Abstract
Alzheimer’s disease (AD) is the most common cause of dementia and is currently one of the biggest public health concerns in the world. Mitochondrial dysfunction in neurons is one of the major hallmarks of AD. Emerging evidence suggests that mitochondrial miRNAs potentially play important roles in the mitochondrial dysfunctions, focusing on synapse in AD progression. In this meta-analysis paper, a comprehensive literature review was conducted to identify and discuss the (1) role of mitochondrial miRNAs that regulate mitochondrial and synaptic functions; (2) the role of various factors such as mitochondrial dynamics, biogenesis, calcium signaling, biological sex, and aging on synapse and mitochondrial function; (3) how synapse damage and mitochondrial dysfunctions contribute to AD; (4) the structure and function of synapse and mitochondria in the disease process; (5) latest research developments in synapse and mitochondria in healthy and disease states; and (6) therapeutic strategies that improve synaptic and mitochondrial functions in AD. Specifically, we discussed how differences in the expression of mitochondrial miRNAs affect ATP production, oxidative stress, mitophagy, bioenergetics, mitochondrial dynamics, synaptic activity, synaptic plasticity, neurotransmission, and synaptotoxicity in neurons observed during AD. However, more research is needed to confirm the locations and roles of individual mitochondrial miRNAs in the development of AD.
Keywords: Alzheimer’s disease, MicroRNAs, Mitochondria, Synapse, Bioenergetics, Synaptic activity
Introduction
Alzheimer’s disease (AD) is a progressive, neurodegenerative disorder and is the most common cause of dementia in older people, making up 60 to 80% of dementia cases. Dementia refers to a decline in brain functioning in multiple areas including memory, reasoning, and language (Bulgart et al., 2020). Cellular changes occur at least two decades before clinical symptoms develop in patients with AD. The main characteristics of AD are memory loss and cognitive impairment that adversely affect patients’ ability to independently carry out their daily activities. Causes of AD are still unknown and no curative treatments exist (DeTure and Dickson, 2019; John and Reddy, 2021; Sheladia and Reddy, 2021).
AD is one of the major public health concerns in the world (DeTure and Dickson, 2019). An estimated 6.2 million Americans over the age of 65 currently live with AD, 72% of them being 75 or older (Alzheimer’sAssociation, 2021). AD is becoming an increasingly bigger problem in the United States as its population with age 65 and older is projected to grow from 58 million in 2021 to 88 million by 2050 with the number of new AD cases predicted to double by 2050 (Alzheimer’s Association, 2021). As the prevalence of AD increases, so will the resulting financial and family burdens.
The predicted aggregate cost of care for people, ages 65 and older with AD or other dementias in 2021 is $355 million in the United States (Alzheimer’sAssociation, 2021). The cost of care for AD patients is predicted to increase to $1.1 trillion due to increases in government spending through Medicare and Medicaid programs and out-of-pocket expenses (Alzheimer’sAssociation, 2021; Wong, 2020). In addition to financial burdens, AD patients have increased emotional and physical stress on family members and friends. Increases in stress is partly due to financial burdens caused by medical costs and reduced earnings by having to work less, increased responsibility of care due to progressively worsening symptoms, presence of other diseases, and watching a family member’s condition deteriorate (Brodaty and Donkin, 2009). A meta-analysis report showed that 30 to 40% of family members who care for someone with dementia have stated having depression (Alzheimer’s Association, 2021).
Research has shown that changes in the brain such as the presence of soluble and insoluble beta-amyloid (Aβ) (Kametani and Hasegawa, 2018; Sadigh-Eteghad et al., 2015), neurofibrillary tangles (NFTs) and accumulation of abnormal form of phosphorylated tau (p-tau) protein inside the neurons (Ashrafian et al., 2021; Hondius et al., 2021), mitochondrial dysfunction (Swerdlow, 2020; Weidling and Swerdlow 2020; Kodavati et al., 2020), atrophy (Planche et al., 2021), and inflammation (Cullen et al., 2021) are found to be associated with AD.
The synapse requires large amounts of energy to effectively transmit signals. Mitochondrial dysfunction in AD prevents the synapse from properly carrying out its function, thus leading to decreased neurotransmission and weaker synaptic connections. This leads to cognitive impairment and worse memory that is present in AD. Therefore, understanding how mitochondrial miRNAs affects mitochondrial function at synapse and the role they play in the progression of AD will allow researchers to use these miRNAs as biomarkers or potential treatments. This article discusses how the change in the expression of mitochondrial miRNAs affects the production of energy by neurons in the brain during AD and how this leads to the progression of AD.
1. Alzheimer’s disease
Alzheimer’s disease occurs in two forms: early-onset (familial) and late-onset (sporadic). Familial AD develops well before the age of 65, typically between the ages of 30 and 50, and is extremely rare (one to two percent of all AD cases). Familial AD is caused by mutations in amyloid-beta precursor protein (APP), presenilin1 (PSEN1), and presenilin 2 (PSEN2) genes, which leads to overproduction of Aβ plaques (Ayodele et al., 2021; Lane et al., 2018; Mao et al., 2011).
Multiple factors are involved in late-onset AD, including lifestyle (unhealthy diet and physical inactivity), traumatic brain injury, diabetes/obesity, cardiovascular, hypertension, kidney disease, epigenetic factors, and environmental and occupational exposures (Sheladia and Reddy, 2021). In addition, APOE4 genotype is a major risk factor. Researchers have found that APOE-e4, a gene that supplies the blueprint for a protein that transports cholesterol in the bloodstream, increases the risk of AD (Safieh et al., 2019). Above all, age is the single most important risk factor causing both familial and sporadic AD and also in combination with aging, single nucleotide polymorphisms largely contribute to late-onset (Chakrabarti et al., 2015; Guerreiro and Bras, 2015; Van Cauwenberghe et al., 2016; Cannon-Albright et al., 2019). The symptoms of AD usually begin after the age of 60 and the percentage of people with AD exponentially increases with age: 5.3% for 65–74 age group, 13.8% for 75–84 age group, and 34.6% of 85 and older age group (Alzheimer’s Association, 2021).
Regarding APOE4 in AD, every person inherits one of the three alleles of the APOE gene (e2, e3, or e4) from each parent leading to six possible APOE pairs (Strittmatter and Roses, 1996). However, occurrences of specific APOE pairs seem to differ in different racial and ethnic groups. For example, the percentage of African Americans inheriting the AD-causing APOE-e4 is about 38.8% compared to 26.2% for European Americans (Alzheimer’sAssociation, 2021). A meta-analysis by Ward and others (Ward et al., 2012) found that 56% of the people diagnosed with AD in the United States had one copy of the APOE-e4 gene followed by 11% with two copies of that gene. However, research findings related to the risk of the presence of APOE-e4 on AD are inconclusive. For example, many AD studies (Green et al., 2002; Liu et al., 2013; Hendrie et al., 2014) that involved Black Americans have had inconsistent results regarding the presence of e4 allele in the risk of developing AD.
1.1. Synapse basis of Alzheimer’s disease
Accumulation of Aβ monomers, oligomers, and deposits/plaques interfere with neuron-to-neuron communications at synapses contributing to neurodegeneration (damage and death of neurons) (He et al., 2019; Kumar and Reddy, 2020). Similarly, tau tangles inside neurons block the transport of essential molecules for the normal functioning and health of neurons (Niewiadomska et al., 2021). Figure 1 depicts how the presence of p-tau and Aβ oligomers contribute to synaptic dysfunction.
Fig. 1. Synapse dysfunction in Alzheimer’s disease.

One of the major hallmarks of AD is synapse dysfunction, which as shown in the figure, leads to the activation of microglia. This figure depicts the presence of p-tau inside the synapse and Aβ oligomers in the extracellular space, both of which contribute to AD and the decrease in synaptic activity. Moreover, this figure illustrates how the presence of dysfunctional mitochondria and the resulting calcium overload in the synapse also contributes to the damage of synapse. The closed channels on the postsynaptic neuron show how neurotransmission is impaired due to synaptic dysfunction (Adapted from “Synaptic Cleft Horizontal”, by BioRender.com (2021). Retrieved from https://app.biorender.com/biorender-templates.).
The research related to the sequence of events that leads to the death of neurons is limited; however, researchers believe that increases in tau are associated with an increase in Aβ (Gerrits et al., 2021; He et al., 2019). Further, the presence of these toxic proteins is believed to activate microglia, the principal immune cell in the brain, to remove toxic proteins and dead cells (Azam et al., 2021). Figure 1 highlights the resulting activation of microglia in the brain. When microglia cannot keep with their cleaning function, chronic inflammation in the brain can occur. Therefore, inflammation is considered a biomarker of AD and can be used to measure the presence or absence of AD (Park et al., 2020). Researchers have become increasingly interested in microRNAs (miRNAs) and their association with mitochondrial and synaptic functions in neurons as the progression of AD involves mitochondrial and synaptic dysfunctions, both of which contribute to the impaired neurotransmission and memory loss that is seen in AD. Many miRNAs have been found to be deregulated in AD, many of which have been associated with mitochondrial and synaptic dysfunctions (Kumar and Reddy, 2020). Therefore, the deregulation of miRNAs is potentially involved in the progression of AD and can serve as treatments or biomarkers.
2. MicroRNAs
MicroRNAs (miRNA) are small, conserved non-coding RNAs (average 22 nucleotides long) that post-transcriptionally regulate gene expression through complementary binding to messenger RNA (mRNA) (O’Brien et al., 2018). Most miRNAs are transcribed in the nucleus by RNA polymerase II or III to primary miRNA (pri-miRNA) (Wahid et al., 2010). Pri-miRNA is cleaved into precursor-miRNA (pre-miRNA) by DROSHA and DGCR8 proteins (O’Brien et al., 2018; Starega-Roslan et al., 2011). The pre-miRNA is transported to the cytoplasm by the Exportin 5/RanGTP complex (Bohnsack et al., 2004). Pre-miRNA is digested by RNAse III Dicer and TBRP, forming a miRNA duplex. Helicase then unwinds the duplex, creating a single-stranded mature-miRNA while the other strand degrades (Ambrus and Frolov, 2009). The mature-miRNA interacts with Ago2 protein to form an RNA-induced silencing complex (RISC) (Treiber et al., 2019). Figure 2 presents the many potential pathways for miRNA biogenesis and localization to the mitochondria. The RISC binds to 3’UTR of the mRNA through complementary base pairing and downregulates gene expression by repressing translation or degrading mRNA (van den Berg et al., 2008; Liu et al., 2014). Some miRNAs have also been found to bind to the 5’UTR region and coding sequence of the mRNA and others have been found to upregulate translation (Orom et al., 2008; Truesdell et al., 2012).
Fig. 2. Mitochondrial miRNA biogenesis.

There are many potential mechanisms of mitochondrial miRNA biogenesis. The first proposed mechanism is that RISC is formed in the cytoplasm and then transported into the mitochondria. Another proposed mechanism is that mature miRNA translocates to the mitochondria and then RISC is formed inside the mitochondria. Moreover, pre-miRNA might translocate into the mitochondria where it is able to be converted into mature miRNA and then RISC. The last proposed mechanism is that mitochondrial miRNA biogenesis occurs entirely in the mitochondria using mtDNA (Created with BioRender.com).
2.1. MicroRNAs and Alzheimer’s disease
Differing levels of miRNA between healthy and diseased states have been discovered in many diseases, such as cancer, atherosclerosis, and many neurodegenerative diseases, including AD (Kemar et al., 2014; Kumar et al., 2017; Kumar and Reddy, 2016; Vijayan et al., 2018; Zhao et al., 2019; Lukiw, 2020). In AD, under expression and overexpression of certain miRNAs have been associated with many of the physiological effects that accompany the disease (Silvestro et al., 2019). For example, the downregulation of miR-193b, which targets Amyloid Beta Precursor Protein (AβPP), is associated with the accumulation of Aβ plaques (Liu et al., 2014). The miRNAs also play important roles in aging (Kenyon, 2010; Smith-Vikos and Slack, 2012), mitochondrial dysfunction, tau accumulation, and in regulating enzymes associated with Aβ production (Reddy et al., 2017). Therefore, further research on the role and mechanism of miRNAs in the brain and how the disease develops from the deregulation of miRNAs will provide great insight into how AD progresses and how miRNAs can be used as potential biomarkers for AD. Kumar and others has currently been focusing on mitochondrial dysfunction in AD, how this dysfunction harm the synaptic function, and how miRNAs may be related to mitochondrial (Gong et al., 2017; Hong et al., 2017; Kumar et al., 2019) and synaptic functions (Di Rita et al., 2020; Wingo et al., 2020).
3. Mitochondria and Alzheimer’s disease
Mitochondria are organelles located within eukaryotic cells that produce about 90% of cell energy as adenosine triphosphate (ATP) among many other functions (Garcia et al., 2019). Mitochondria are believed to have originated from the integration of an alphaproteobacterium into anaerobic host archaea (Roger et al., 2017). There still exists a lot of controversy regarding the specifics of the origin of the mitochondria. However, it is believed that both organisms had a symbiotic relationship where the bacterium aerobically produced energy for the archaea, thus removing oxygen, which would have been toxic to the anaerobe. Evidence for this includes that mitochondrial DNAs are similar to bacterial DNA and they reproduce through binary fission. Over time, the bacterium eventually evolved into the mitochondria seen in eukaryotic cells today.
Reddy’s laboratory has done extensive research on mitochondrial dysfunction in AD and published many articles discussing both what mitochondrial dysfunction looks like in AD and how it develops (Reddy and Oliver, 2019; Oliver and Reddy, 2019; Pradeepkiran and Reddy, 2020; Bhatti et al., 2021). Mitochondrial dysfunction in AD such as impaired dynamics and mitophagy are areas that researchers are focusing on as these functions are heavily involved in synapse impairment (Cai and Tammineni, 2016). Figure 3 depicts the mitochondrial dysfunction that is present at the synapse during AD and highlights the deregulation of mitochondrial miRNAs associated with each dysfunction. An increased emphasis in this article is being placed on synaptic mitochondria and how mitochondrial miRNA affects its function.
Fig. 3. Mitochondrial microRNAs and mitochondrial dysfunction in Alzheimer’s disease.

This figure depicts the various mitochondrial dysfunctions present in the synapse during AD and highlights the deregulation of miRNAs that is related to each impairment. The green and red arrows indicate the upregulation or downregulation, respectively, of a miRNA in AD. Moreover, mitochondrial trafficking, neurotransmission, and calcium regulation are shown to be impeded. As a result, the synapse during AD has many damaged mitochondria and fewer functional mitochondria (Created with BioRender.com).
3.1. Mitochondrial genome
Mitochondrial DNA (mtDNA) is circular and double-stranded with 16569 base pairs and has been found to encode 37 genes, 22 tRNAs, 13 polypeptides, and two rRNAs (Taanman, 1999). The human mitochondria have between two and 10 copies of mtDNA located in the matrix and is replicated using DNA polymerase γ (Copeland, 2010; Turnbull et al., 2010; Oliver and Reddy, 2019; Morton et al., 2021). The mtDNA is predominantly inherited matrilineally (Radzvilavicius et al., 2017) and for this reason, offspring may inherit most maternal mtDNA diseases. Most proteins that localize to the mitochondria are encoded by the nuclear genome. However, complexes I, III, IV, and V are encoded by both the nuclear and mitochondrial genomes (Reddy and Beal, 2005). The mtDNA is more easily mutated than nuclear DNA as it can be damaged by interaction with reactive oxygen species (ROS) produced during oxidative phosphorylation (OXPHOS) in aerobic respiration (Kujoth et al., 2005; Ishikawa et al., 2008).
3.2. Mitochondrial structure
Mitochondria are surrounded by a double membrane consisting of the inner membrane and the outer membrane (Kuhlbrandt, 2015). The space between outer and inner membranes is known as the intermembrane space. One of the main processes associated with this space is the passive diffusion across the inner membrane through ATP synthase to produce ATP (Klingenberg, 2008). The folded protrusions in the inner mitochondrial membrane are known as cristae that penetrate the inner mitochondrial matrix. Cristae have several cylindrical connections, 12–40 nm in diameter, to the inner membrane called cristae junctions (Mannella, 2020). These formations play a key role in inter-mitochondrial communication and increase the surface area for mitochondrial reactions to occur, thus allowing for more ATP to be generated (Mannella, 2020). The matrix is the space within the inner membrane and contains ribosomes, mtDNA, and metabolic enzymes (Klingenberg, 2008; Greber and Ban, 2016). Many biological processes occur in the matrix, one of the main processes being the tricarboxylic acid cycle (TCA cycle), which plays a major role in energy production and biosynthesis in cells (Martinez-Reyes and Chandel, 2020).
The outer membrane controls mitochondrion’s shape and plays a vital role in communicating with other organelles. It consists of voltage-dependent anion channels (VDAC) and many other pores in the membrane, allowing for many molecules, including proteins, ions, and other low molecular weight solutes, to be able to diffuse unrestricted through the outer membrane (Camara et al., 2017). Also, the outer membrane consists of the protein complex, translocase of the outer membrane (TOM), which allows proteins to cross the outer membrane into the intermembrane space (Perry et al., 2008). The inner membrane is significantly less permeable and more selective in what can diffuse across the membrane. Certain molecules are only able to diffuse across the inner membrane through specific transporters. Translocase of the inner membrane (TIM) protein, TIM23, allows for the transport of proteins across the inner membrane into the matrix and TIM22 allows for the integration of proteins into the inner membrane (Jensen and Dunn, 2002). Protons are the main ions that diffuse across the inner membrane to generate the proton gradient for producing ATP during OXPHOS (Kuhlbrandt, 2015). Calcium, cofactors, biosynthetic precursors, and metabolites can also diffuse across the inner membrane (Zorova et al., 2018; Rossi et al., 2019). In addition, embedded in the inner membrane are enzymes involved in the electron transport chain (ETC) (Complexes I-IV) and the production of ATP (ATP Synthase) along with the mitochondrial calcium uniporter (MCU) (Finkel et al., 2015; Kuhlbrandt, 2015). Figure 4 illustrates the general structure of the mitochondria, including various channels present in the outer and inner membranes along with the mtDNA and ribosomes present in the matrix.
Fig. 4. Mitochondrial structure.

Mitochondria consists of a double membrane (outer and inner membrane), intermembrane space, cristae, and the matrix. The outer membrane has TOM complexes and VDAC embedded to allow the transport of proteins and ions respectively from the cytoplasm to the intermembrane space. The inner mitochondrial membrane has TIM complexes that allow for the transport of proteins from the intermembrane space to the matrix (TIM23) and the integration of proteins within the inner membrane (TIM22). MCU and NCLX, which are involved in calcium transport into and out of the matrix, and the respiratory complexes (I-V) that are involved in oxidative phosphorylation are also located in the inner membrane. The matrix consists of ribosomes and mtDNA (Adapted from “Electron Transport Chain”, by BioRender.com (2021). Retrieved from https://app.biorender.com/biorender-templates.).
3.3. Mitochondrial functions
Mitochondria produce ATPs through a variety of catabolic processes including aerobic respiration, ketolysis, and amino acid catabolism (Bhatti et al., 2017; Spinelli and Haigis, 2018). The number of mitochondria in a cell depends on the energy needs of that cell. For example, muscle cells have many mitochondria as they need to produce energy to move the body. The bioenergetics of the mitochondria is discussed in detail later in the article. In addition to producing ATP, mitochondria have many other functions including calcium homeostasis, participating in controlled cell death (apoptosis), cell signaling, redox balance/dealing with oxidative stress, thermogenesis, innate immunity, and being involved in many biosynthetic pathways (Tait and Green, 2012).
Currently, scientists do not completely understand the functions of mtDNA and their implications in diseases. However, recent research has shown that defects in mtDNA are associated with many human diseases such as diabetes, AD, Parkinson’s disease (PD), Huntington’s disease (HD), cancer, and aging (Bender et al., 2006; Wallace, 2010; Di Stefano et al., 2016; Yan et al., 2019) as it causes damage and dysfunctional mitochondria.
3.4. Mitochondria and calcium signaling
Calcium ion (Ca2+) plays a vital role in neuronal function and plasticity (Burgoyne and Haynes, 2012). It serves as a messenger to transmit depolarization status and synaptic activity within a neuron thereby relaying neuronal activity status (Gleichmann and Mattson, 2011). Specifically, an action potential triggers the active transport of Ca2+ into the presynaptic terminal of the neuron by opening up Ca2+ channels which in turn triggers the release of neurotransmitters through exocytosis, thus allowing the action potential to continue to the postsynaptic neuron (Gleichmann and Mattson, 2011; Mochida, 2021). This process increases local Ca2+ concentrations at the presynaptic active zone (Gleichmann and Mattson, 2011). High-intensity Ca2+ signaling requires high ATP consumption for Ca2+ homeostasis in the neuron (Sudhof, 2012). One of the biggest functions of the mitochondria is to maintain Ca2+ homeostasis (Kannurpatti, 2017). Mitochondria works with the ER to maintain Ca2+ balance (Pedriali et al., 2017). It buffers Ca2+ concentrations by transporting calcium into the matrix and out of the mitochondria using VDAC1 in the outer membrane along with Na+-dependent mitochondrial Ca2+ efflux transporter (NCLX) and MCU located in the inner membrane (Reddy, 2013a; Reddy, 2013b; Finkel et al., 2015; Shoshan-Barmatz and De, 2017). When the mitochondria gets overloaded with calcium, mitochondrial permeability transition pores (mPTPs) in the inner mitochondrial membrane open, thereby triggering apoptosis through the release of cytochrome c and activation of caspases in the cytosol, thus making calcium deregulation an important consideration during neurodegenerative diseases (Calvo-Rodriguez et al., 2020).
3.5. Mitochondrial dynamics
Mitochondrial dynamics, which involves both fusion and fission of mitochondria, and degradation of damaged mtDNA, also provide an important function in neurons (Youle and van der Bliek, 2012). The fusion (combining) allows mitochondria to significantly reduce the effects of changes in mtDNA (Vidoni et al., 2013). Changes in mtDNA must reach elevated levels to lead to dysfunction in the ETC, thus through fusion, the mutated mtDNA is split between both mitochondria, thus leading to lower levels within each mitochondrion (Wang et al., 2020b). This allows mitochondria to tolerate high mutation levels. Also, fusion allows mitochondria with damaged components to regain functional versions of those components, thus protecting the cell from dysfunctional mitochondria (Vidoni et al., 2013). After fusion, the mitochondria undergo fission (splitting) back into separate mitochondria (Mishra and Chan, 2014). Due to fusion, the mitochondria that previously had impaired function regain its functionality. As a result, the fusion/fission mechanism leads to a variety of shapes and functions of mitochondria (Pagliuso et al., 2018; Ma et al., 2020). The dendrites consist of long and tubular mitochondria (Rangaraju et al., 2019) while axons consist of short and punctate mitochondria. This allows for the compartmentalization of specialized functions of mitochondria in the neuron and allows for local regulation. For example, the compartmentalization of specific mitochondria in the dendrite and axon are important for local translation, which requires large amounts of energy. Regulation and balancing of fission and fusion processes are managed by a set of fusion (dynamin GTPase regulators Optic Atrophy 1, OPA1; Mitofusin 1, MFN1; Mitofusin 2, MFN2) and fission (dynamin-like GTPase regulator Dynamin related protein 1, DRP1) proteins (Reddy et al., 2011; Kandimalla et al., 2016; Ali and McStay, 2018; Oliver and Reddy, 2019).
3.6. Mitochondrial differences between genders
Researchers have identified sex-based differences in mitochondrial functions in the brain. Human mitochondria and mtDNA are found to be mainly maternally inherited (Giles et al., 1980). However, recent research has found that mtDNA can also be paternally inherited (McWilliams and Suomalainen, 2019). Sex steroid hormones are known to play a role in the function of mitochondria and have been found to have neuroprotective effects. Steroids, known as neuroactive steroids, are produced in the brain, thus can impact neuronal functions (Melcangi et al., 2016). Understanding differences in mitochondrial functions due to sex allows development of better treatment strategies for mitochondrial dysfunction observed during neurodegenerative diseases.
Since more than two-thirds of those with AD are women, sex is an important factor to consider (Alzheimer’sAssociation, 2021). One major sex difference is that men have significantly higher levels of testosterone than women while women have higher levels of estrogen and progesterone. Mitochondrial content is approximately the same between sexes, thus indicating that differences in mitochondrial function are due to impacts by sex-based hormones and proteins associated with the mitochondria (Guevara et al., 2011). Women were found to have greater rates of mitochondrial ATP production from glucose metabolism than men due to increased Pyruvate Dehydrogenase Complex activity and decreased ROS production compared to men, thus resulting in lower oxidative stress (Gaignard et al., 2018). However, OXPHOS activity was found to be the same between males and females (Guevara et al., 2011). A study with rats showed that female rats had greater levels of mitochondrial function than male rats (Guevara et al., 2011). Not much literature was found analyzing the differences in the expression of mitochondrial miRNAs between genders. One study found that mitochondrial metabolism in an overloaded heart was subjected to sex-specific miRNA regulation (Sanchez-Ruderisch et al., 2019). Therefore, researchers should investigate sex-specific miRNA regulation in neurons in the brain as this might give further insight into mitochondrial differences between genders.
3.6.1. Sexual hormones and mitochondria
Estrogen, progesterone, and testosterone are found to affect mitochondrial functions in both men and women as these three hormones are found in both sexes. Differing levels of these hormones between sexes might indicate which hormones have significant impact in each sex and how hormonal changes due to aging affect mitochondrial function. Studies showed that mitochondrial respiration was decreased substantially after the removal of ovaries but not from the removal of testicles, thereby showing that female sex hormones (estrogen and progesterone) have a greater impact on mitochondrial function than testosterone (Guevara et al., 2011). Estradiol, the prominent estrogen in humans, was found to increase OXPHOS activity, inhibit the production of ROS, and stabilize the potential of the mitochondrial membrane (Lejri et al., 2018). In addition, estrogen was found to suppress apoptosis and promote mitochondrial biogenesis (Klinge, 2008). Progesterone was also found to increase energy production by the mitochondria and reduce oxidative stress (Gaignard et al., 2016). Testosterone deficiency was found to damage mitochondria in the substantia nigra in the brain through increased oxidative stress and reduced the activity of Complex I, thus signifying the potential effect that testosterone has on mitochondrial dysfunction in the brain (Yan et al., 2017).
Researchers have found differences in brain mitochondrial dysfunction between sexes. As a person ages, the amount of sex hormones in the body decreases. Since these steroid hormones play a role in mitochondrial function, a decrease in these hormones contributes to mitochondrial dysfunction. Moreover, research has found that the mitochondria play a role in the synthesis of steroids, thus dysfunction in the mitochondria could in turn also be responsible for reduction in sex hormones’ levels (Gaignard et al., 2017). Women have a higher life expectancy than men, thus indicating that age is one of the main contributors to why more women have AD as age is the most significant risk factor for AD (Ginter and Simko, 2013). However, differences in sex hormones and the decreased production of sex hormones as people get older may also play a major role in this discrepancy. Learning more about how differences in mitochondrial function between genders change with aging will allow researchers to get a better understanding of why more women have AD.
3.7. Mitochondrial function and aging
Mitochondrial changes occur with age in humans and all mammals, leading to progressively worse function. As a result, mitochondrial dysfunctions related to redox homeostasis, bioenergetics, dynamics, and mitophagy have been associated with many neurodegenerative diseases, including AD. Mitochondrial function changes with aging involve mtDNA changes and decreased ATP production, which then leads to damage/dysfunction of mitochondrial proteins.
ROS production contributes a significant amount to mitochondrial dysfunction during aging. Complexes I and III, which are part of the ETC, are known to produce ROS (Hirst et al., 2008). Complex I produce ROS in the FMN site and CoQ binding site due to electron leak that occurs when electrons transfer from NADH to CoQ, resulting in the reduction of O2 to O2− (Zhao et al., 2019). In complex III, the electron from CoQH− leaks out and reduces O2 to O2−. The ROS produced by CIII is released into the matrix and the intermembrane space where O2− can be converted to H2O2 utilizing SOD1 and exit the mitochondria into the cytosol (Zhao et al., 2019). Complex II was also found to be involved in pathways associated with ROS production (Lenaz, 2001). Complex II generates ROS due to electron leak that occurs from electrons supplied from succinate and also in the reverse direction from electrons supplied by CoQH2 (Quinlan et al. 2012). In addition, alpha-ketoglutarate, an enzyme that is involved in the TCA cycle, was found to produce ROS (Starkov et al., 2004). At appropriate levels, ROS participate in many signaling pathways that contribute to important brain functions, such as synaptic plasticity (Cobley et al., 2018). However, as a person ages, oxidative stress increases due to the dysregulation of redox homeostasis. The build-up of ROS associated with aging contributes to increased oxidative stress and leads to changes in mtDNA (Stefanatos and Sanz, 2018). ROS that is generated in the mitochondria also damages mtDNA through base modifications caused by oxidation, resulting in the formation of 8-hydroxy-2’-deoxyguanosine (Mao and Reddy, 2011; Torres-Gonzalez et al., 2014). Furthermore, antioxidant enzymes in the mitochondria become damaged with age, further making the mitochondria of older people more susceptible to oxidative stress (Wei and Lee, 2002). Due to increased oxidative stress, the mitochondria of older people exhibit impaired functions.
In healthy adults, the mitochondria play a vital role in producing most of the energy used in neuronal function. Therefore, a vast number of functional mitochondria are needed for neurons to meet their high energy demands. As previously mentioned, mitochondria become increasingly damaged with age due to higher levels of oxidative stress. The energy provided by the mitochondria is used in a variety of mitochondrial and synaptic functions in neurons that are discussed throughout this paper. As a result, aging is associated with decreased ATP production by the mitochondria (Chistiakov et al., 2014). Decreased energy production in neurons contributes to further mitochondrial dysfunction, synaptic dysfunction and leads to neuronal death.
Mitochondrial trafficking, dynamics, and mitophagy also experience alterations as people get older. Researchers found that mitochondrial trafficking decreases early on in adulthood and continues to decline as people age (Morsci et al., 2016; Morton et al., 2021). Moreover, aging was found to impair mitophagy (Tyrrell et al., 2020). In healthy adults, mitophagy removes damaged mitochondria and trafficking allows for the transport of functional mitochondria to these areas. However, due to aging and age-dependent increased accumulation of mutant proteins, and abnormal interactions between Abeta/p-tau with Drp1 and VDAC1, mitophagy is decreased, thus contributing to the continued presence of dysfunctional mitochondria (Morton et al 2021, Manczak et al 2011, Manczak and Reddy 2012). As a result, these synaptic areas are unable to produce the energy needed for proper functioning. For example, neurotransmission has been found to be decreased in AD due in part to impaired mitophagy in neurons (Han et al. 2020; Kerr at al. 2017).
4.1. Mitochondrial dysfunction in AD and neurodegenerative diseases
Alzheimer’s disease is closely associated with various forms of mitochondrial dysfunctions including excessive ROS production, low ATP production, mitochondrial Ca2+ dyshomeostasis, and defects in mitochondrial dynamics and transport, and mitophagy. Figure 1 displays how the disruption of calcium homeostasis and increased presence of damaged mitochondria contributes to synaptic dysfunction. These physiological changes contribute to the impaired function of mitochondria in neurons. Aβ plaques and NFTs work independently as well as together in causing mitochondrial dysfunction. Considering the critical role of mitochondria in energy production, calcium buffering and cell health, any impediment in the mitochondrial movement and function can lead to neurological (Deheshi et al., 2013; Misgeld and Schwarz, 2017; Norat et al., 2020) and neuropsychiatric (Millar et al., 2000; Belmaker and Agam, 2008; Pei and Wallace, 2018; Zilocchi et al., 2020) disorders.
4.2. Impaired mitochondrial ATP production in Alzheimer’s disease
Production of ATP by mitochondria through OXPHOS is essential for cellular functions (Verschueren et al., 2019). Mitochondria produce ATP using five enzyme complexes in their inner membrane. Numerous studies have shown declined levels of all five-enzyme complexes in multiple areas of the AD brain (Liang et al., 2008). A decrease in the production of ATP in neuronal cells is associated with ROS overproduction and may fail to meet the energy requirements of neurons and glia. Aβ buildup is also associated with decreased energy production. Research has shown that Aβ oligomers inhibit the activities of cytochrome c oxidase (Complex IV) and ATP synthase, thus impairing the ATP production during OXPHOS (Morton et al., 2021; Wang et al., 2020b). In addition, the α-ketoglutarate dehydrogenase complex, pyruvate dehydrogenase complex showed decreased activity. Decreased energy production prevents the neuron from carrying out its function properly, thus leading to neuronal death. Low ATP production impairs neurons’ ability to release neurotransmitters, leading to decreased neurotransmission and cognitive impairment.
4.3. Oxidative stress in Alzheimer’s disease
The overproduction of ROS in turn causes mitochondrial dysfunction through oxidative stress. Oxidative stress occurs when the production of ROS significantly increases, thus exceeding the antioxidant enzyme capabilities of a cell (Reddy and Beal, 2005; Reddy, 2006; Reddy, 2007; Reddy, 2009a; Reddy 2009b). The ROS are produced as a by-product of metabolism in mitochondria (Sies and Jones, 2020). Aβ has been found to contribute to the increased production of ROS by mitochondria by forming coordination compounds with metal ions (Cu and Fe) in the brain (Cheignon et al., 2018). In AD brains, increased ROS production is due to deficiency of cytochrome c oxidase in the mitochondrial ETC (Rak et al., 2016). The ROS consists of both radical and non-radical oxygen species such as superoxide radical anion (O2−), hydrogen peroxide (H2O2), hydroxyl radical (HO), nitric oxide (NO), and peroxynitrite (ONOO−). An imbalance between production and degradation of ROS can cause oxidative stress as they are capable of oxidizing nucleic acids (DNA & RNA), proteins, and lipids and consequently damage brain cells (Arimon et al., 2015; Dumanovic et al., 2020; Misrani et al., 2021; Tauffenberger and Magistretti, 2021). Many studies have shown that ROS accounts for cellular injury in aging and many neurodegenerative diseases (Misrani et al., 2021). As a person ages, increasing ROS levels have been found to be associated with the accumulation of damaged mitochondria.
Oxidative stress leads to changes in mtDNA. The mtDNA changes that occur with age and are not inherited are known as somatic mutations. These mtDNA mutations have been associated with decreased functioning of OXPHOS as mtDNAs have shown to help code for enzymes used in the ETC, thus damaging mitochondria and consequently leading to reduced ATP production. ROS also damage proteins and lipids associated with bioenergetics, such as cardiolipin found in the inner membrane (Reddy and Beal, 2005; Reddy, 2006; Reddy, 2007; Reddy, 2009a; Reddy, 2009b). As a result, neurons die over time from apoptosis/necrosis due to not meeting their energy needs or increased damage by ROS, thus leading to the development and progression of neuroinflammation and neurodegenerative diseases. Moreover, increased ROS levels are associated with promoting senescence in other cells by leading to the damage and shortening of telomeres in nuclear DNA and through ROS-mediated cell signaling pathways. Oxidative stress and consequent substantial increase in their by-products have been widely reported in AD (Bhattacharyya et al., 2014). These oxidized biomolecule by-products are more stable and therefore used as ROS markers in monitoring the progression of AD (Marrocco et al., 2017). Table 1 presents some of the by-products that are studied in AD as oxidative stress markers. Neuronal mitochondria themselves are the victims of oxidative stress as it leads to their progressive destruction. Increased oxidative stress is important as it damages mitochondria, thus causing neurons to become dysfunctional. Oxidative stress also damages mtDNA, thus further damaging the respiratory complexes. The overproduction of ROS in turn causes mitochondrial dysfunction through oxidative stress. This vicious cycle of decreased ATP production due to compromised OXPHOS, elevated oxidative stress, mitochondrial dysfunction, and cell death is the hallmark of AD progression (Reddy and Beal, 2008).
Table 1.
Oxidized biomolecule by-products produced in the brain cell due to overproduction of ROS in AD.
| Sl. No. | Oxidation of | By-products or Oxidative stress markers | References |
|---|---|---|---|
| 1 | Nucleic acids (DNAs and RNAs) | 8-OH-2’-deoxyguanosine 8-OH-guanosine |
(Pena-Bautista et al., 2019; Zhang et al., 2013) |
| 2 | Proteins | Protein carbonyls 3-nitrotyrosine (3-NT) |
(Butterfield and Boyd-Kimball, 2020; Butterfield et al., 2007) |
| 3 | Lipids | 4-hydroxy-2-trans-nonenal (HNE) Malondialdehyde (MDA) 2-propenal (acrolein) F2-isoprostanes F4-neuroprostanes Thiobarbituric acid-reactive substances (TBARS) Free fatty acid release |
(Dei et al., 2002; Di Domenico et al., 2017; Gamba et al., 2019; Lovell et al., 1997; Moldogazieva et al., 2019) |
| 4 | Carbohydrates | Advanced glycation end products | (Dei et al., 2002; Srikanth et al., 2011) |
4.4. Impaired mitochondrial Ca2+ production in Alzheimer’s disease
Ca2+ is a crucial second messenger for the normal functioning of neurons as it modulates different neuronal processes such as secretion, motility, metabolic regulation, synaptic plasticity, proliferation, gene expression, and apoptosis (Horigane et al., 2019). It also plays a key role in learning and memory (Berridge, 2014). Studies have found that dysregulation involves elevated levels of cytosolic calcium due to increased influx of Ca2+ through voltage-gated calcium channels, reduced calcium buffering, reduced expression of Ca2+-binding proteins, and impaired handling of calcium by the mitochondria (Catterall, 2011; Nanou and Catterall, 2018; Strickland et al., 2019; Ureshino et al., 2019). Sustained overloading of cytosolic Ca2+ promotes cell death while their deficiency might affect signal transmissions (Esteras and Abramov, 2020). Research has found that Aβ plaques build up significantly in the synaptic mitochondria contributes to Ca2+ dysregulation (Calvo-Rodriguez et al., 2020). In cortical neurons, Aβ is believed to increase the release of calcium from ER, leading to a significant increase in cytosolic calcium levels (Green, 2009). This results in increased uptake of Ca2+ by the mitochondria (Tillement et al., 2011). High cyclophilin D levels also have been found to disrupt Ca2+ balance by synaptic mitochondria (Naga et al., 2007; Modesti et al., 2021). The excessive amount of Ca2+ in the mitochondria contributes to the collapse of mitochondrial membrane potential due to the opening of mPTPs, activation of pro-apoptotic proteins, and increased production of ROS (Calvo-Rodriguez et al., 2020). Also, the Aβ-mediated impediments in Ca2+ regulation at multiple cellular levels include increased inositol 1,4,5-trisphosphate receptor activity at the ER (Supnet and Bezprozvanny, 2010) and the dysregulation of voltage-operated channels. This impairment in mitochondria’s activity has been widely observed in AD brains (Berridge, 2014; Britti et al., 2018). Figure 5 summarizes Ca2+ regulation in healthy synapse compared to its deregulation in AD brains.
Fig. 5. Mitochondria and calcium regulation in healthy synapse (left) and in Alzheimer’s disease (right).

During neurotransmitter release, the synapse experiences an influx of calcium through voltage-gated calcium channels. Mitochondria play a significant role in regulating calcium levels in the synapse. Calcium crosses the outer mitochondrial membrane through VDAC1 and then passes through the inner mitochondrial membrane and into the matrix through MCU. While the mitochondria control calcium levels, synaptic vesicles form in the membrane and fuse with the synaptic membrane to release neurotransmitters by exocytosis. However, in AD, cytosolic calcium levels are significantly elevated due to a greater influx of calcium ions through the voltage-gated calcium channels and increased release of calcium into the synapse by the endoplasmic reticulum. Calcium deregulation was found to occur due in part to Aβ. MPTPs open in the inner mitochondrial membrane and mitochondria experience a substantially greater influx of calcium. As a result, the mitochondria in the synapse become dysfunctional, thus leading to neuronal apoptosis and consequently impaired neurotransmission and decreased synaptic plasticity (Created with BioRender.com).
4.5. Impaired mitochondrial dynamics and mitophagy in Alzheimer’s disease
The transport of mitochondria in neurons is essential for supplying ATP to synapses, Ca2+ buffering, and repairing of mitochondria (Calkins et al., 2011; Course and Wang, 2016). This is facilitated through microtubule tracks. The integrity of these tracks is known to be affected by tau proteins in AD cells negatively impacting mitochondrial transport (Perez et al., 2018). Further, fusion protein MFN2 has been shown to participate in mitochondrial transport by interacting with mitochondrial Rho small GTPase (MIRO), a protein associated with motor proteins to transport mitochondria in the anterograde direction (Majstrowicz et al., 2021). Mutations or deletions in MFN2 lead to the slowing of axonal transport in both directions (Misko et al., 2010). In AD, impaired mitochondrial axonal transport is known to precede the accumulation of toxic protein compounds and is linked to distressed synaptic function (Calkins et al., 2011; Calkins et al., 2012; Cai and Tammineni, 2017; Morton et al., 2021). However, precise underlying molecular mechanisms of how it affects mitochondrial transport are not known. The development of neurodegenerative diseases has also been associated with the reduction in mitophagy (Bakula and Scheibye-Knudsen, 2020; Swerdlow and Wilkins, 2020). Mitophagy is the selective degradation of mitochondria through autophagy. PD, HD and AD all have been found to have decreased mitophagy (Cai and Jeong, 2020; Norat et al., 2020; Choi and Han, 2021). Moreover, cristae disorders have been observed in humans with diseases such as ALS, AD, and PD (Stanga et al., 2020).
Disruption in mitochondrial dynamics through increased fission and the accumulation of changes in mtDNA also contribute to the increased presence of dysfunctional mitochondria. Aβ interacts with many proteins located in the mitochondria, thus altering mitochondrial dynamics. For example, Aβ interacts with VDAC1 and stops the transport of mitochondrial proteins, resulting in the formation of free radicals (Rajmohan and Reddy, 2017). Researchers have found that Aβ interacts with Drp1 which causes increased mitochondrial fragmentation and reduced fusion in the synapses (Manczak et al., 2011). In addition, mitochondrial fission is more pronounced with an increase in Aβ and Tau levels as they interact with fission regulator proteins (Kandimalla et al., 2016). Tau has been found to disrupt the transport of mitochondria along microtubules to the synapse, thus hindering the release of neurotransmitters and leading to synaptic dysfunction (Forner et al., 2017). Consequently, increased fission leads to reduced mitophagy, thus leading to the presence of more dysfunctional mitochondria fission. As a result, if dysfunctional mitochondria are not removed, it can lead to build-up of ROS and decreased energy production, ultimately resulting in neuronal death (Misrani et al., 2021).
5. Mitochondrial Bioenergetics
The mitochondria are responsible for up to 95% of the energy produced in the Central Nervous System (CNS) (Benaroya, 2020). Moreover, the brain has the highest energy demand in the body due to the constant synaptic activity, generation of action potentials in neurons, and complex processing. As previously mentioned, the brain consumes about 20% of the total energy of the body while accounting for only 2% of the body weight (Silver and Erecinska, 1998). There are an estimated two million mitochondria in a neuron (Misgeld and Schwarz, 2017).
Most of the energy produced in the brain is used for synaptic activity (Harris et al., 2012). Mitochondria in neurons produce energy in a variety of different pathways. The main pathway utilized to make ATP is aerobic respiration as glucose is the most used fuel source by neurons. The first step of aerobic respiration, glycolysis, takes place in the cytosol. During glycolysis, glucose is oxidized into two molecules of pyruvate (Yetkin-Arik et al., 2019). Pyruvate is then able to enter the pyruvate dehydrogenase (PDH) complex in the mitochondrial matrix (Gray et al., 2014). Another contributor to the formation of pyruvate is the formation of lactate in astrocytes (Descalzi et al., 2019). The lactate produced by astrocytes can be transported to neurons and converted into pyruvate for use in energy production. PDH oxidizes pyruvate into acetyl-CoA, producing carbon dioxide and NADH. Acetyl-CoA then enters the TCA cycle where Acetyl-CoA is used to produce ATP through substrate-level phosphorylation and NADH/FADH2 which are electron carriers (Gray et al., 2014). Both NADH and FADH2 are then used in the respiratory chain (Complexes I-IV) to pump protons across the inner membrane from the matrix to the intermembrane space (Cooper, 2000). Then the protons flow across its gradient through ATP synthase, generating the production of ATP through oxidative phosphorylation.
Under conditions of prolonged starvation, the brain can derive up to ⅔ of its energy from ketone bodies (Naithani and Karn, 2020). Ketone bodies can cross the blood-brain barrier and be used as a fuel source for cells in the CNS. Through ketolysis, ketone bodies are converted into acetyl-CoA, which can then enter the TCA cycle. Amino acids and fatty acids can also feed into the TCA cycle (Swerdlow, 2012). The localization of miRNAs to the mitochondria allows them to regulate energy production along with all other mitochondrial functions that have been discussed. Learning about the mechanism of miRNA localization to various cellular organelles will allow for a better understanding of the specific mechanisms of how miRNAs localize and impact mitochondrial functions.
6. Mitochondria and neuronal function
The human brain consists of about 86 billion neuronal cells wired in complex networks from which our emotions, movements, and experiences arise (Azevedo et al., 2009). Not surprisingly, the CNS is known for extraordinarily high metabolic rate as neurons are highly differentiated cells that require large amounts of ATP for maintaining ion gradients across cell membranes and for neurotransmission (Chamberlain and Sheng, 2019). For example, ATP is required to power the Na+/K+ pump to create the ion gradients necessary to generate action potentials along the axon. Not surprisingly, the brain consumes about 20% of the total energy produced by the body while accounting for only 2% of the body weight (Silver and Erecinska, 1998). Any deficit in the generation of energy can have catastrophic effects on neuronal functioning (Lax et al., 2011). Mitochondria are central to various processes including ATP production in neurons, intracellular Ca2+ signaling, and activating apoptosis while also producing ROS (Ames, 2000; Kann and Kovács, 2007). Therefore, mitochondrial dysfunction has harmful effects on the integrity of cells including cell death, and may be critically involved in aging, metabolic and degenerative diseases (Wallace, 2005; Reddy et al., 2012; Mandal and Drerup, 2019; Sharma et al., 2021).
7. Cellular organelles and miRNAs locations
Organelles are subcellular structures that have one or more specific functions to perform within a cell. miRNAs have been identified in many different organelles including endoplasmic reticulum (ER), trans-Golgi network, nucleus, and mitochondria (Maurel and Chevet, 2013). The mechanism of subcellular localization and trafficking of miRNA is not fully understood (Simion et al., 2020; Trabucchi and Mategot, 2020). However, it is known that miRNA expression is related to the regulation of proteins that help with the function of many organelles in the cell (Leung, 2015; O’Brien et al., 2018). For example, there is a functional connection between the expression of miRNAs and ERUPR signaling that has shown that miRNA regulates ER as well as regulated by ER signaling (Maurel and Chevet, 2013; Almanza et al., 2019; Ortega et al., 2020). By targeting the expression of proteins that are found in different organelles, miRNA plays a significant role in the function of organelles. Moreover, research has found miRNAs that are localized to the mitochondria and alter mitochondrial gene expression by targeting mRNA located in the mitochondria (John et al., 2020). Therefore, studying miRNA specific to the mitochondria will provide great insight in determining the mechanism that miRNA has towards the progression of AD and as biomarkers for the early detection of this disease. The next sections focus specifically on the function of mitochondria and how alterations in levels of mitochondrial miRNAs are associated with mitochondrial dysfunction that is typical of AD.
8. Mitochondrial miRNAs
Mitochondrial miRNAs are target genes that code for proteins that are essential for proper mitochondrial function. A vast number of mitochondrial miRNAs have the potential to be directly involved in the pathogenesis of AD. Table 2 gives a list of mitochondrial miRNAs that researchers have found to be differentially expressed in AD and present in neurons in the brain. It highlights the target gene, mRNA binding region, mitochondrial functions, and the importance of these miRNAs to AD. In addition, Table 3 shows how the miRNAs are related to other neurodegenerative diseases such as PD, HD, and Amyotrophic Lateral Sclerosis (ALS) as these diseases are similar to AD. These miRNAs have the potential to be therapeutic targets for the treatment of AD or as biomarkers.
Table 2.
Mitochondrial miRNAs associated with Alzheimer’s disease.
| Sl. No. | microRNA | Target Gene(s) | Regulation | Region | Function | Importance | References |
|---|---|---|---|---|---|---|---|
| 1 | miR-7 | VDAC1 | Downregulation of target gene | 3’ UTR | Stabilizes mitochondrial membrane potential | VDAC1 significantly increased in AD so miR-7 might also have impact on AD | (Chaudhuri et al., 2016) |
| 2 | miR-15b | SIRT4, BACE1 | Downregulation of target gene | 3’ UTR | Plays role in redox homeostasis and counteracts senescence-related mitochondrial dysfunction | Found downregulated in AD and plays a role in repressing BACE1. Downregulation of mir-15b could also upregulate SIRT4 in AD, leading to mitochondrial dysfunction. | (Lang et al., 2016; Gong et al., 2017) |
| 3 | miR-16-5p | BCL-2 | Downregulation of target gene | 3’ UTR | Disrupts mitochondrial membrane integrity and promote apoptosis | Upregulated in AD due to Aβ, leading to neuronal cell death through apoptosis | (Leggio et al., 2017; Liguori et al., 2018; Kim et al., 2020) |
| 4 | miR-23a/b | SIRT1, GLS | Downregulation of target gene | 3’ UTR | Mediates protective neuronal stress response and regulates mitochondrial metabolism by targeting glutaminase | Found upregulated in frontal cortex in AD, thus indicating that mir-23a/b might play potential role in disease progression by decreasing energy production and stress response in neurons, leading to neuronal death | (Gao et al., 2009; Weinberg et al., 2015; Tang, 2016) |
| 5 | miR-29a | VDAC1, VDAC2, VDAC3 | Downregulation of target gene | 3’ UTR | Triggers apoptosis by targeting mitochondrial voltage dependent anion channels in outer membrane of mitochondria | Downregulated in AD, thus potentially disrupting the control of neuron apoptosis and proliferation in brain | (Shioya et al., 2010; Bargaje et al., 2012) |
| 6 | miR-29b | Bcl2L2, Mcl-1 | Downregulation of target gene | 3’ UTR | Promotes apoptosis and loss of mitochondrial membrane potential | Downregulated in AD, thus potentially serving a neuroprotective role | (Mott et al., 2007; Hebert et al., 2008; Shi et al., 2012; Lungu et al., 2013) |
| 7 | miR-30 | DRP1, p53 | Downregulation of target gene | 3’ UTR | Inhibits mitochondrial fission and apoptosis | Downregulated in AD, which then can lead to increased neuronal death through apoptosis | (Li et al., 2010) |
| 8 | mir-34a | NDUFC2, SDHC, UQCRB, UQCRQ, COX10, Bcl-2, SIRT1, Txnrd2 | Downregulation of target gene | 3’ UTR | Downregulates antioxidant mitochondrial enzymes, impair energy production, and induce apoptosis | Upregulated in AD, leading to widespread mitochondrial dysfunction | (Yamakuchi et al., 2008; Bai et al., 2011; Lin et al., 2015) |
| 9 | miR-98 | HEY2 | Downregulation of target gene | 3’ UTR | Inhibits neuronal apoptosis and ameliorates oxidative stress and mitochondrial dysfunction | Downregulated in mice with AD and found to reduce the production of Aβ and improve mitochondrial dysfunction | (Chen et al., 2019a) |
| 10 | miR-125b | FOXQ1 | Downregulation of target gene | 3’ UTR | Induces apoptosis by upregulating Bax and downregulating Bcl-2 | Found upregulated in AD, thus suggesting that the increase in miR-125b during AD enhances apoptosis in neurons | (Hong et al., 2017; Ma et al., 2017) |
| 11 | miR-132/212 | PTEN, FOXO3a, P300, NOS1 | Downregulation of target gene | 3’ UTR | Inhibits neuronal apoptosis | Downregulated in AD neurons, thus contributing to the promotion of apoptosis of neurons in the brain | (Wong et al., 2013; Wang et al., 2017) |
| 12 | miR-137 | FUNDC1, NIX | Downregulation of target gene | 3’ UTR | Inhibit mitophagy | Downregulated in AD patients, thus potentially having role in promoting mitophagy | (Geekiyanage and Chan, 2011; Li et al., 2014c; Li et al., 2017) |
| 13 | miR-140 | PINK1, mfn1 | Downregulation of target gene | 3’ UTR | Increases mitochondrial dysfunction and ROS production while also promoting autophagy, mitochondrial fission, and apoptosis | Reduced PINK1 and upregulated miR-140 is associated with AD. Induced PINK1 overexpression leads to decreased mitochondrial ROS and injury of the mitochondrial membrane, thereby preventing neuronal cell apoptosis. Therefore, silencing of miR-140 poses a novel therapeutic target for AD. | (Li et al., 2014c; Liang et al., 2021) |
| 14 | miR-195 | mfn2 | Downregulation of target gene | 3’ UTR | Decreases mitochondrial membrane potential and activity | Mfn2 is found to be downregulated in hippocampal neurons during AD, therefore upregulation of mfn2 by inhibiting miR-195 is a potential therapeutic strategy for AD. | (Zhang et al., 2016) |
| 15 | miR-204 | TRPML1 | Downregulation of target gene | 3’ UTR | Increases mitochondrial damage, ROS production, and mitophagy | Downregulation of miR-204 inhibits mitochondrial damage, ROS production, and mitophagy caused by Aβ | (Zhang et al., 2021) |
| 16 | miR-210 | ISCU1/2, COX10 | Downregulation of target gene | 3’ UTR | Downregulates mitochondrial respiration and activates glycolysis and ROS generation | Downregulated in the hippocampus of AD. Soluble Aβ leads to upregulation of miR-210, thus leading to greater inhibition of ETC and increased production of ROS. Downregulation of miR-210 during AD serves as protection against toxic effects of Aβ and reduces oxidative stress. | (Cogswell et al., 2008; Chen et al., 2010; Huang et al., 2010; Li et al., 2014b) |
| 17 | miR-218 | PRKN | Downregulation of target gene | 3’ UTR | Inhibits mitophagy and mitochondrial clearance and protects against neurotoxicity that is caused by metallic ions and other neurotoxins | PRKN found at higher levels in mice AD brain, thus showing that miR-218 is potentially downregulated in AD, thus leading to the dysreuglation of mitophagy and neuron homeostasis. miR-218 was found to be a potential therapeutic target for AD. | (Di Rita et al., 2020; Hou et al., 2020) |
| 18 | miR-335 | SOD2, Txnrd2 | Downregulation of target gene | 3’ UTR | Downregulates antioxidant enzymes in mitochondria, leading to increase in ROS | Upregulated in AD and associated with senescence, thus potentially contributing to mitochondrial dysfunction associated with AD | (Bai et al., 2011; Satoh et al., 2015) |
| 19 | miR-338 | COXIV | Downregulation of target gene | 3’ UTR | Decreases metabolic activity in axonal mitochondria | Overexpression of this brain-specific miRNA contributes to decreased energy production in neurons and could be involved in pathogenesis of AD | (Aschrafi et al., 2008) |
| 20 | miR-351-5p | Miro2 | Downregulation of target gene | 3’ UTR | Regulates mitochondrial GTPase and induces mitochondrial fission, leading to mitochondrial dysfunction and promoting mitophagy | Found upregulated in AD and contributes to hippocampal neural progenitor cell death, which is associated with AD, | (Woo et al., 2021) |
| 21 | miR-375 | SIX4 | Downregulation of target gene | 3’ UTR | Promotes oxidative stress injury and apoptosis | Found upregulated in AD, which was found to promote oxidative stress injury and apoptosis in neurons | (Wang et al., 2020a) |
| 22 | miR-455-3p | APP | Downregulation of target gene | 3’ UTR | Regulates mitochondrial dynamics (decrease mitochondrial fission/increase mitochondrial fusion) and supports mitochondrial biogenesis and synaptic plasticity | Found upregulated in AD neurons compared to control neurons in response to increase in Aβ | (Kumar et al., 2017; Kumar et al., 2019; Kumar and Reddy, 2019) |
| 23 | miR-484 | Fis1, BCL2L13 | Downregulation of target gene | Coding region (Fis1) and 3’ UTR (BCL2L13) | Inhibits mitochondrial fission and apoptosis | Lower level of miR-484 associated with mild cognitive impairment, cognitive decline and higher risk of having AD | (Wang et al., 2012; Wingo et al., 2020; Liu et al., 2021) |
| 24 | miR-743a | mdh2 | Downregulation of target gene | 3’ UTR | Suppresses malate dehydrogenase activity | Malate dehydrogenase activity is elevated in humans with AD, thus indicating the potential inhibition of miR-743 a due to increased oxidative stress | (Shi and Gibson, 2011) |
Table 3.
Mitochondrial miRNAs associated with Huntington Disease (HD), Parkinson’s Disease (HD) and Amyotrophic lateral sclerosis (ALS).
8.1. Mitochondria localized miRNAs
Mitochondria-localized miRNAs (mitomiRs) are miRNAs that are found inside the mitochondria. These mitomiRs have been hypothesized to be encoded by the nuclear genome or by the mitochondrial genome. Figure 2 illustrates the many potential mechanisms of mitomiR biogenesis. The presence of miRNA in the mitochondria has only been discovered this past decade in various species. In 2009, Kren and others discovered the localization of mitochondria in rat liver and Bian et al. found miRNA located in the mitochondria of mouse liver (Kren et al., 2009; Bian et al., 2010). Research on mitomiRs in humans is limited. In 2011, Bandiera and others found 13 miRNAs at high levels in mitochondria that were purified from HeLa cells (Bandiera et al., 2011). The same study found that the outsourcing of miRNA by the nucleus to the mitochondria is highly conserved in mammals. Barrey and others have found both pre-miRNA (pre-mir-302a and pre-let-7b) and miRNA (miR-365) that are localized to the mitochondria in human myocytes, thus showing how both pre-miRNA and miRNA are found in the mitochondria (Barrey et al., 2011).
8.2. MitomiRs biogenesis
Where the mitomiRs originate from, how they are transported, how they know where to localize, if RISC forms in the cytosol and/or the mitochondria, and whether they are synthesized in the mitochondria or only processed in the mitochondria are not known. In addition, if and how interactions occur between mitomiR and mtDNA transcripts are not fully understood. Most mitomiRs encoded in the nucleus were found to originate from loci that are either transcribed from mitochondrial gene clusters or located close to mitochondrial genes, thus the linking to mitochondrial-related genes could play a role in mitomiR being able to localize to the mitochondria (Vendramin et al., 2017). Also, mitomiRs atypical size (17–25 nucleotide length) and distinctive thermodynamic properties increase the possibility that their structures are involved in mitochondrial localization and being able to transport through the mitochondrial membranes (Bandiera et al., 2011; Vendramin et al., 2017). Since, Ago2 was found to localize to the mitochondria, this suggests that miRNA may be processed in the mitochondria; however, Ago2 has functions unrelated to miRNA processing, thus the localization might not be related to mitomiRs (Zhang et al., 2014). P-bodies have been stated to allow the transport of the miRNA-Ago2 complexes directly into the mitochondria (Macgregor-Das and Das, 2018). Another proposed mechanism is that miRNA-Ago2 complexes can travel through TOM20 and SAM50 into the outer membrane and then polynucleotide phosphorylase (PNPAse) in the intermembrane space can assist in the transportation of the complex across the inner membrane and into the matrix through TIM complexes (Macgregor-Das and Das, 2018).
MitomiRs are hypothesized to play a regulatory role in various mitochondrial functions. No specific research has been done regarding mitomiRs in neurons. Because mitochondrial dysfunction is one of the main indicators of AD, discovering the specific functions and mechanism of actions of mitomiRs in human neurons and the differential expression of mitomiRs in neurons during AD will provide greater insight in the impact of mitomiRs in the progression of AD and potential targets for treatments.
8.3. How to study mitochondrial miRNA in neurons
A few studies have been performed in analyzing the localization and specific function of mitochondrial miRNA in neurons, especially in the synapse where mitochondrial miRNA have important regulatory functions (John et al., 2020). Separation of the cytosol and synaptosome components of the neuron through centrifugation and extraction reagents allows for the isolation of miRNA that is specifically located in the synapse. Moreover, mitochondria can be extracted from the synapse using commercial kits or differential centrifugation and then characterized using transmission electron microscopy and through the immunoblotting of mitochondrial marker genes (Liao et al., 2020). Afterwards, mitochondrial miRNA can be extracted and used to produce cDNA, and then qRT-PCR can be used to quantify how much of a specific miRNA is present in the mitochondria from the synaptosome. This procedure can be used with transgenic APP or Tau mice tissue samples, postmortem human tissue samples, or transfected cells to analyze the altered regulation of miRNA in AD vs WT samples. Next, miRNAs can be quantified in both healthy and diseased states to identify mitochondrial miRNAs of interest. The removal or addition of specific mitochondrial miRNA into neurons can be used to better understand what functions they regulate by measuring mitochondrial activity.
8.4. Techniques for mitochondrial miRNAs analysis
Mitochondrial miRNAs can be analyzed utilizing many different methods. Once mitomiRs of interest are isolated, qRT-PCR, Northern blot, NC2 hybridization, miRNA microarray, and cellular fractionation combined with deep sequencing can be used to learn more about them (Larriba et al., 2016). Moreover, mitochondrial miRNAs can be knocked-in or knocked-out of mice/isolated cells and then changes in mitochondrial function can be analyzed through various techniques. Mitochondria biogenesis/mitophagy can be measured using electron microscopy, fluorescence microscopy, and western blotting along with recently developed techniques using MitoTimer, Mt-Keima, and Mito-QC (Williams et al., 2017; Pradeepkiran and Reddy, 2020). Mitochondrial dynamics can be measured using fluorescence wide-field microscopy and 3D image processing (Song et al., 2008). More specifically, mitochondrial fusion is measured using polyethylene glycol (PEG) fusion assay alongside imaging flow cyclometry (Nascimento et al., 2017). Gokerkucuk and others have identified a variety of methods to measure mitophagy, dynamics, and calcium signaling using fluorescent dyes and proteins (mitophagy = GFP-LC3; dynamics = tetramethylrhodamine ethyl ester (TMRE), Rhodamine 123 (R123), tetramethylrhodamine methyl ester (TMRM), and 5,5,6,6’-tetrachloro-1,1’,3,3’ tetraethylbenzimidazoylcarbocyanine iodide (JC-1), naphthalimide (NPA-TPP), boron-dipyrromethene (BODIPY)) and genetically-encoded sensors (mitophagy = RFP-GFP; dynamics = mitochondria-targeted photoactivable GFP (PAGFPmt), mitochondrially-targeted DsRed fluorescent protein (mtDsRed); calcium signaling = genetically-encoded Ca2+ indicators (GECIs) (Gokerkucuk et al., 2020).
Mitochondrial calcium regulation can also be measured using fluorescence microscopy with Fluo-4, AM, TMRM, Fura-2, Indo-1, Rhod-2 as indicators (McKenzie et al., 2017; Gokerkucuk et al., 2020). ATP production can be measured using bioluminescence assays and high resolution respirometry (Drew and Leeuwenburgh, 2003; Lanza and Nair, 2010). Amplex Red Assays, which detects the presence of H2O2, can be used to quantify ROS production (Karakuzu et al., 2019). Mitochondrial trafficking can be analyzed by using a photo-switchable fluorescent protein (dendra2) that can track movement and be used to determine a velocity distribution or utilizing fluorescent microscopy on neurons that are transfected with fluorescent proteins, such as enhanced yellow fluorescent protein (eYFP), that target the mitochondria and can be used to measure mitochondria localization frequency, mitochondrial residence time, number of passing mitochondria per minute, and fraction of passing mitochondria that stop (Chang et al., 2006; Niescier et al., 2016). Apoptosis can be analyzed using flow cytometry (Chen et al., 2019b; Wang et al., 2020b).
Furthermore, researchers can work to develop methods to isolate mitochondria from tissue samples and isolate miRNA. Mitochondrial miRNAs can be analyzed to see if they bind to mtDNA and to which sequences in the mitochondrial genome and then observe mitochondrial activities for any changes. Moreover, finding ways to label a specific miRNA sequence that is known to localize to the mitochondria and observing its path to the mitochondria while also determining the presence of certain proteins associated with miRNA biogenesis in the mitochondria can give better understanding of how mitomiRs localize to the mitochondria. The next steps in mitochondrial miRNA analysis are to determine: 1) how mitomiRs are localized to the mitochondria, 2) the specific mechanism of actions of these mitomiRs, 3) to identity the function of all neuronal mitomiRs and which mitomiRs are specific to synaptic function, 4) the deregulation of specific mitomiRs in AD, and 5) how these mitomiRs can be used as a treatments or biomarkers of AD. Figure 6 presents the current techniques in sequential order, which researchers can follow when studying mitochondrial miRNAs.
Fig. 6. Mitochondrial miRNA analysis flow chart.

This figure depicts a flow chart of how to study mitochondrial miRNAs. The first half of the flow chart involves how to identify and quantify mitochondrial miRNAs and mitomiRs in the synapse and mitochondria. Next, the chart portrays how these identified miRNAs should be analyzed for their target gene and whether or not they upregulate or downregulate that gene. The chart goes on to show how researchers should explore if these identified miRNAs are upregulated or downregulated in AD. At the bottom, the chart shows how the mitochondrial functions of these miRNAs can be determined by knocking-in or knocking-out a specific miRNA and measuring mitochondrial activity (Created with BioRender.com).
8.5. Mitochondrial miRNAs and ATP production
miRNAs downregulate genes that code for proteins involved in energy production. For example, miR-743a was found to suppress malate dehydrogenase activity (Shi and Gibson, 2011). Malate dehydrogenase is involved in the oxidation of malate to oxaloacetate during the TCA cycle, producing NADH. Malate dehydrogenase activity is upregulated in AD, thus miR-743a might be downregulated in AD (Shi and Gibson, 2011). miR-23a/b are also known to disrupt the TCA cycle by downregulating glutaminase (Gao et al., 2009). Furthermore, miR-210 (Chen et al., 2010), miR-338 (Aschrafi et al., 2008), and miR-34a (Sarkar et al., 2016) inhibit the activity of enzymes involved in OXPHOS. Most of the ATP produced in neurons comes from OXPHOS, thus the suppression of this process prevents neurons from meeting their high energy demand, leading to neuronal death.
Membrane potential is important for maintaining the ETC. miRNAs such as miR-16-5p (Kim et al., 2020), miR-195 (Zhang et al., 2016), and miR-29b (Lungu et al., 2013) are known to disrupt mitochondrial membrane potential and integrity while miR-7 (Chaudhuri et al., 2016) is known to stabilize the membrane potential. Decreased production of energy also inhibits neurons’ abilities to transmit signals. Ultimately, disrupted transmission and the loss of neurons contribute to the cognitive impairments seen in AD. Therefore, understanding how mitochondrial miRNA disrupts bioenergetics in neurons during AD will provide greater insight into the progression of AD and lead to the development of potential diagnostics and treatments for this disease.
8.6. Mitochondrial miRNAs and oxidative stress
Oxidative stress is a major contributor to mitochondrial dysfunction during AD. The dysregulation of many miRNAs in neurons have been found to be related to increased ROS production by mitochondria. Mitochondrial miRNAs were also found to affect the function of antioxidant enzymes that play a role in redox homeostasis. miR-98, miR-204, miR-34a, miR-15b, miR-375, miR-140, and miR-335 are all involved in the mitochondria’s handling of oxidative stress. MiR-98 (Chen et al., 2019a), and miR-15b (Lang et al., 2016) were found to promote redox homeostasis and reduce oxidative stress. In AD, these miRNAs are downregulated, leading to increased ROS production and oxidative damage to neurons, which in turn, leads to cell death (Gong et al., 2017; Chen et al., 2019a). miR-204 (Zhang et al., 2021), miR-34a (Sarkar et al., 2016), miR-375 (Wang et al., 2020a), miR-140 (Liang et al., 2021), and miR-335 (Bai et al., 2011) were found to promote ROS production and inhibit the function of antioxidant enzymes in the mitochondria. Therefore, the upregulation of these miRNAs leads to apoptosis in neurons during AD due to increased oxidative stress.
8.7. Mitochondrial miRNAs and mitophagy
Mitophagy is the removal of dead mitochondria through autophagy. Dysfunctional mitophagy leads to the increased presence of damaged mitochondria in neurons. Damaged mitochondria are unable to produce enough energy for neuron survival. Therefore, mitochondrial miRNAs play a vital role in regulating mitophagy and clearing damaged mitochondria. miR-137 (Li et al., 2014c), miR-34a (Xiong et al., 2019), and miR-218 (Di Rita et al., 2020) were found to inhibit mitophagy, thus the downregulation of these miRNAs in AD can increase the number of functional mitochondria in neurons. miR-204 (Zhang et al., 2021) and miR-351-5p (Woo et al., 2021) promote mitophagy and are found to be downregulated in AD.
8.8. Mitochondrial miRNAs and mitochondrial biogenesis
Mitochondrial biogenesis is the formation of new mitochondria and leads to an increase in the number of mitochondria in neurons. This process is important as it allows neurons to meet their high energy needs. Researchers have found that miR-455-3p (Kumar and Reddy, 2019) and miR-34a (Xiong et al., 2019) are involved in mitochondrial biogenesis while miR-23a/b regulates SIRT1, which is associated with mitochondrial biogenesis, thereby indicating a potentially link between miR-23a/b and biogenesis (Weinberg et al., 2015; Tang, 2016). Altered levels of these miRNAs in AD play a role in disrupting biogenesis, thus decreasing the number of functional mitochondria in neurons, leading to neuronal death.
8.9. Mitochondrial miRNAs and mitochondrial dynamics
Mitochondrial dynamics involve the fission and fusion balance of mitochondria and are important in maintaining functional mitochondria in neurons. Mitochondrial miRNAs were found to affect mitochondrial dynamics by either inhibiting or promoting fission or fusion. miRNAs including miR-484, miR-455-3p, miR-30, miR-351-5p, and miR-140 are associated with mitochondrial dynamics. In AD, miR-484 (Wang et al., 2012) and miR-30 (Li et al., 2010) were downregulated while miR-351-5p (Woo et al., 2021) and miR-140 (Li et al., 2014a) were upregulated, thereby leading to increased mitochondrial fission and neuronal cell death through apoptosis. miR-455-3p is produced in response to Aβ production and acts as a negative feedback inhibitor of APP, enhancing fusion (Kumar et al., 2017., Kumar et al 2019).
8.10. Mitochondrial miRNAs and apoptosis
Increased apoptosis in the brain leads to neuronal death during AD. Mitochondria are involved in the mechanisms of this pathway. Widespread neuronal death disrupts pathways involved in learning and memory; thus, miRNA works to limit apoptosis to only when needed. Therefore, the dysregulation of mitochondrial miRNA(s) related to apoptosis plays a key role in neuron survival and death. miR-7 (Chaudhuri et al., 2016), miR-98 (Chen et al., 2019a), miR-30 (Li et al., 2010), miR-132/212 (Wong et al., 2013), and miR-484 (Liu et al., 2021) were found to inhibit apoptosis. The downregulation of these miRNAs in AD leads to neuronal death via apoptosis. Conversely, miR-16-5p (Kim et al., 2020), miR-34a (Lin et al., 2015), miR-375 (Wang et al., 2020a), miR-125b (Hong et al., 2017), miR-29a (Bargaje et al., 2012), miR-29b (Shi et al., 2012), and miR-140 (Liang et al., 2021) induce apoptosis and are consequently found upregulated in AD.
9. Synapse function in the brain
The synapse is a major component of the neuron and is responsible for neurotransmission (John and Reddy, 2021). Researchers have found that the human neocortex has approximately 150 trillion synapses (Finnema et al., 2016). The entire human brain was found to have around a total of quadrillion synapses (Ho et al., 2011). Due to the vast number of synapses in the human brain, the synapses are significantly involved in major brain functions. In neurons, action potentials travel from the axon hillock to the synapse. For the action potential to travel to the next neuron, the synapse must release neurotransmitters into the synaptic cleft via exocytosis at chemical synapses or by ions traveling through gap junctions directly into the next neuron at electrical synapses (John and Reddy, 2021). Both types of synapses allow for the transmission of signals from one neuron to the next. To release neurotransmitters, an influx of Ca2+ travels through voltage-gated calcium channels into the synapse. This influx of calcium ions leads to the fusion of vesicles filled with neurotransmitters to the synaptic terminals, resulting in the release of neurotransmitters. The neurotransmitters can travel through the synaptic cleft and bind to the dendrite of the postsynaptic neuron (John and Reddy, 2021).
9.1. Chemical and electrical synapse
Synapses are the locations where signals are transmitted from one neuron (presynaptic neuron) to the subsequent neuron (postsynaptic neuron). Synapses can be classified in many ways. For instance, synapses can be either chemical or electrical. Chemical synapses involve a gap between the presynaptic and postsynaptic neuron known as the synaptic cleft (John and Reddy, 2021). The presynaptic neuron releases neurotransmitters into the synaptic cleft where the neurotransmitters can bind to ionotropic or metabotropic receptors on the postsynaptic neuron. Ionotropic receptors initiate a rapid response but last for a short period while metabotropic receptors lead to slower, but longer-lasting responses. Chemical synapses also allow the postsynaptic neuron to receive and integrate signals from multiple presynaptic neurons. On the other hand, electrical synapses are formed by the connection of the presynaptic neuron to the postsynaptic neuron via gap junctions. This allows direct transfer of ions and signals from the presynaptic neuron to the postsynaptic neuron. Because the cytoplasm of the neurons is directly connected by gap junctions, electrical synapses transmit signals significantly faster than chemical synapses.
9.2. Excitatory and inhibitory synapse
Synapses can be classified as excitatory or inhibitory. Excitatory synapses contribute to the depolarization of the postsynaptic neuron by allowing the influx of Na+ ions. On the other hand, inhibitory synapses contribute to the hyperpolarization of the postsynaptic neuron by allowing the influx of Cl− ions. In chemical synapses, the postsynaptic neuron sums the signals it receives from both excitatory synapses (excitatory postsynaptic potentials) and inhibitory synapses (inhibitory postsynaptic potential), thus leading to either the transmission of the signal to the postsynaptic neuron or the termination of the signal. Glutamate is the main excitatory neurotransmitter in the CNS while γ-aminobutyric acid (GABA) is the main inhibitory neurotransmitter in the CNS.
9.3. Astrocytes, microglia and synapse
Many different cells in the brain are involved in synaptic processes. Astrocytes are glial cells that have been found to form the blood-brain barrier. Researchers have found that astrocytes are involved in the clearance of neurotransmitters along with the formation, maturation, alteration, and elimination of synapses (Chung et al., 2015). Moreover, cholesterol secreted by astrocytes has been found to enhance synaptic transmission by increasing the chance of the presynaptic neuron releasing glutamate (Mauch et al., 2001). Microglia are brain-specific macrophages and are known to play a role in the elimination of synapses, such as during synaptic pruning (Paolicelli et al., 2011). As a result, microglia have been found to play an important role in the removal of synapses during AD (Rajendran and Paolicelli, 2018). Finally, the synapses of neurons in the brain are involved in the transmission of signals, thus playing a vital role in neural processing and memory formation through long-term potentiation (LTP). The LTP is the long-lasting strengthening of synapses of neurons due to increased stimulation. Therefore, these different types of cells in the brain are all important to proper brain function, thus making them targets for analysis during AD.
9.4. Synapse and synaptic plasticity
Synaptic plasticity is the strengthening or weakening of synapses and connections between neurons, which occurs due to either increased or decreased activity, respectively. The activation of neurons leads to the phosphorylation of synapsin, thus increasing the number of synaptic vesicles that are ready for release, thereby allowing for neurotransmitters to be released more quickly and efficiently in response to signals. (Ho et al., 2011). Also, RIM proteins regulate the interactions between the influx of calcium and the corresponding release of neurotransmitters and the localization of vesicles in the active zone and preparation for release (Ho et al., 2011). Synaptic plasticity can be both short-term, involving transient changes in the synapse over a few minutes, and long-term, leading to long-lasting changes in the strength of synaptic connections and the formation of new synaptic connections that can last from a few hours to a person’s lifetime and contributes to the depression or strengthening of synaptic connections (Deperrois and Graupner, 2020). In the brain, synaptic plasticity has its greatest importance in forming and maintaining long-term memory. The hippocampus is known to play a significant role in memory and learning, thus LTP is of vital importance in the hippocampus (Nicoll, 2017).
9.5. Mitochondria and synaptic energy
Mitochondria play an important role in the synapse as energy is needed for local translation in the synapse and synaptic maturation (Devine and Kittler, 2018). Mitochondria located in the dendrite have been found to provide the energy needed for local translation during synaptic plasticity (Rossi and Pekkurnaz, 2019). Studies show that blocking mitochondrial function was related to decreased synaptic potentiation and neurotransmission (Guo et al., 2017; Todorova and Blokland, 2017). More specifically, blocking oxidative phosphorylation in mitochondria in neurons inhibits long-term potentiation (Flippo and Strack, 2017; Todorova and Blokland, 2017). Long-term potentiation is important for the formation and retrieval of long-term memory and learning, thus mitochondrial dysfunction can lead to weaker synaptic connections and less excitable neurons, leading to worse memory (Lynch, 2004). Synaptic activity requires large amounts of energy, which is provided by a large number of mitochondria present in the synapse, which also helps with calcium buffering (Devine and Kittler, 2018). ATP is used in the synapse for many functions involved in neurotransmission. For example, ATP is needed for synaptic recycling (Pathak et al., 2015). Also, ATP is needed for the removal of calcium from inside the neuron using calcium-ATPase, resetting potassium and sodium concentration gradients during the refractory period, synthesis of molecules, and mitochondrial trafficking to the synapse (Harris et al., 2012). Trafficking is also essential for the clearance of damaged mitochondria (Sheng, 2014). Therefore, mitochondrial dysfunction present in AD has a considerable impact on impairing neurotransmission and other synaptic functions.
9.6. Mitochondrial trafficking in the synapse
Proper positioning and a healthy pool of mitochondria in neurons are essential to develop and maintain functional neuronal connections (Mandal and Drerup, 2019). The neuron has mechanisms that transport mitochondria to the synapse in order for the synapse to have enough mitochondria to execute its function (Sheng, 2017). Mitochondria are synthesized in the cell soma and are transported along the axons to the synapse using kinesins associated with microtubules and KIF5 factors (Course and Wang, 2016). Mitochondria can travel bi-directionally between the soma and presynapse using motor protein complexes, kinesins for anterograde, and dyneins for retrograde directions (Pilling et al., 2006, Calkins et al., 2012). Mitochondria travel toward the synapse and arrest there due to the higher intracellular calcium levels in the synapse (Saotome et al., 2008). Calcium disrupts interactions between mitochondria and transport complexes consisting of kinesins and Miro1, thus allowing mitochondria to localize and remain in the synapse (Wang and Schwarz, 2009). However, the transport of mitochondria becomes less frequent as neuron ages (Morsci et al., 2016). The movement of mitochondria must be coupled with their proper positioning throughout the neurons to meet the varying energy demands. However, the positioning of these mitochondria is controlled by both docking proteins and local environmental conditions (Cai and Sheng, 2009). For example, docking of mitochondria in neurons is affected by intracellular glucose (Course and Wang, 2016) and calcium (Saotome et al., 2008) concentrations with mitochondria positioning itself in areas where these concentrations are higher. This allows more mitochondria to be available in the part of the neuron that is needing the most energy, such as the synapse and nodes of Ranvier (Misgeld and Schwarz, 2017).
In AD and other neurodegenerative diseases, mitochondrial trafficking is impaired, resulting in fewer mitochondria located in the synapse. As a result, the synapse has less energy available to perform its functions, thus leading to a variety of pathophysiologies during AD. For instance, impaired trafficking results in decreased clearance of dysfunctional mitochondria (Correia et al., 2015). Dysfunctional trafficking occurs for many reasons. Damaged bioenergetics and mitochondrial dynamics have been found to limit mitochondrial transport (Correia et al., 2012; Correia et al., 2016). Energy is required to transport mitochondria, so if the neuron is not producing enough energy, mitochondria are not able to localize to the synapse. Aβ and Tau were also found to disrupt mitochondrial trafficking (Zhao et al., 2010; Calkins and Reddy, 2011; Calkins et al., 2012; Shahpasand et al., 2012; Morton et al., 2021). Both Aβ and Tau destabilize microtubules involved in transport and lead to potential loss, thus decreasing the polarity of the microtubules (Correia et al., 2016). In addition, they contribute to mitochondrial dysfunction thus limiting the energy available for mitochondrial trafficking. As a result, defective mitochondrial trafficking results in the breakdown of synapses, leading to neuronal death (Correia et al., 2016). With less functional synapses, neurotransmission, neural processing, learning, and formation/accessing memories are all impaired.
9.7. Synapse differences between genders
The organization and function of synapses are different between the biological sexes. In 2008, a study found that the synaptic density was significantly higher in men than in women in all layers of the temporal cortex (Alonso-Nanclares et al., 2008). Although men were found to have more synaptic connections in the cortex, this does not indicate greater and more complex processing capabilities. However, it is not known why these areas have greater synaptic densities in men than in women and if yes, how that leads to different functions in those areas of the brain between men and women is not known. Moreover, in men, nitrous oxide is involved in LTP while women are much less dependent on nitrous oxide (Hyer et al., 2018). Researchers also found differences in the mechanism of synaptic plasticity between sexes (Hyer et al., 2018).
Sex steroid hormones are involved in a variety of pathways that influence synapses. Estrogen is a part of signaling pathways that are involved in the formation of synapses in the prefrontal cortex and hippocampus, thus playing a key role in neurological development (Hara et al., 2015). As a result, estrogen is important for synaptic plasticity. Estradiol is of significant interest as it is involved in synaptic plasticity in both men and women as men can convert testosterone into estradiol using aromatase enzymes (Gillies and McArthur, 2010). Therefore, estrogen plays a protective role in the brain and the significant decrease in estrogen after menopause might contribute to why more women have neurodegenerative diseases than men as women are more reliant on estrogen. Currently, estrogen is being explored as a potential treatment for neurodegenerative diseases, but no treatments have been developed yet. Progesterone also has been found to assist in synaptic function and LTP alongside estrogen, although its role is less known (Baudry et al., 2013). Testosterone, the predominant sex hormone in men, also plays a role in synaptic function. One study found that the administration of testosterone to mice increased synaptic density in hippocampal neurons (Fattoretti et al., 2019). Moreover, testosterone was found to maintain the function and levels of presynaptic proteins and the lengths of neurites, thus indicating the neuroprotective effects of testosterone against Aβ. As previously mentioned, testosterone is also able to be converted to estradiol, thus further impacting synapse formation and function.
Therefore, hormonal differences between genders play a role in the formation and maintenance of the synapse, resulting in different mechanisms of neuroprotection. As a result, the development and progression of neurodegenerative diseases are different between the biological sexes. Learning more about how the differences in biological sex contribute to synaptic function and how decreases in hormone levels and other body responses contribute to the progression of the disease will allow researchers to potentially develop more effective treatments.
9.8. Synapse function and aging
The decreased synaptic function is associated with aging, thus leading to decreased neurotransmission over time. This section analyzes the effects of specific changes that happen in the synapse. One major mechanism of synaptic dysfunction attributed to aging as mitochondrial dysfunction starts occurring when person gets older. Mitochondria’s functional changes concerning aging are discussed in Section 3.7. Reduced mitochondrial trafficking, decreased mitophagy, and impaired energy production prevents the synapse from making the appropriate amount of energy needed for its function. Therefore, synaptic impairment increases due to synapses not being able to recycle synaptic vesicles, regulate intracellular synaptic concentrations and reset concentration gradients across the axon and synapse. This leads to synaptic dysfunction and the loss of synaptic connections due to neuronal death which in turn decreases neurotransmission. Furthermore, synaptic strength decreases, as a person gets older. Researchers have found that aging is associated with the loss of synaptic connections (Bishop et al., 2010). Also, alterations in neurotransmitter levels are involved in aging (Anyanwu, 2007). In conjunction, aging results in decreased postsynaptic responsiveness. In addition, aging inhibits LTP and promotes LTD, thus making the formation and access of long-term memories more difficult (Foster, 1999). Together, these alterations weaken neurotransmission and decrease synaptic plasticity. These changes in synapses due to aging make older people more susceptible to AD and other neurodegenerative diseases. The deterioration of neurons leads to fewer synaptic connections, thereby leading to cognitive decline. For example, the loss and weakening of synaptic connections in the hippocampus contribute to the reduction in hippocampal volume and worsening memory (Kashyap et al., 2019). Furthermore, the weakening of existing connections due to aging leads to decreased processing capabilities of older people. Synaptic function changes associated with aging are also seen in synapse AD, thus aging-related changes could play a role in the early development of AD. Analyzing how synapses change due to age will allow researchers to better understand how synaptic changes due to age relate to synaptic changes due to AD. This allows researchers to find how synapses change due to AD and what treatments can be developed to preserve and enhance synapse function with age in elderly and patients with AD. Figure 7 illustrates how the variation and amounts of sex hormones in both genders are related to changing synapse density and mitochondrial respiratory activity as people age. Based on this figure, researchers can potentially explore the effects of sex hormones on mitochondrial and synaptic activity and density in different genders at various periods of life.
Fig. 7. Mitochondrial respiratory activity and synapse density related to age and gender.

In men, testosterone levels are significantly higher than estrogen and progesterone levels. Testosterone is initially high at birth before dropping down after the first week of life (Tomlinson et al., 2004). As boys gets older and goes through puberty, testosterone levels increase, peaking around 21 years of age (Chodick et al., 2020). Afterward, testosterone levels in men gradually decrease throughout adulthood (Chodick et al., 2020). About 80% of estradiol in men is derived from testosterone, thus estrogen in the figure is shown to follow roughly the same trend since less testosterone means less estradiol that can be produced (Finkelstein et al., 2013). Progesterone is involved in the synthesis of testosterone in men and is shown to remain constant as men get older (Hall and Klein, 2017; Oettel and Mukhopadhyay, 2004). For women, estrogen and progesterone levels are significantly higher than testosterone levels. Estrogen in girls stays low until puberty when estrogen levels significantly increase and then levels off around one year after menarche (Hoyt and Falconi, 2015). Perimenopause (transition to menopause) occurs in women in their 40s with menopause beginning, on average, between ages 45 and 51 (McCarthy and Raval, 2020). At early perimenopause, estrogen levels become elevated but drop off substantially in later stages, thus estrogen levels are low by menopause (Hoyt and Falconi, 2015). Progesterone levels are low until puberty where it significantly increases (Hall and Klein, 2017). Progesterone remains constant in adulthood prior to menopause (Hall and Klein, 2017). As women approach menopause, progesterone begins to drastically decrease until progesterone levels are low during menopause. Testosterone in women is found to increase from birth till adulthood and then gradually decrease as women continue to age (Ankarberg and Norjavaara, 1999; Davison et al., 2005). Mitochondrial respiratory activity is proposed to follow hormone changes during aging with decreasing hormone levels corresponding to decreasing activity. Mitochondrial respiratory activity is known to be significantly higher in women than men as reflected in the figure (Gaignard et al., 2018). Moreover, men were found to have higher synaptic density than women in certain areas of the brain (Alonso-Nanclares et al., 2008). In both men and women, synapse density increases and peaks when people are 1–2 years old and then decrease through synaptic pruning as people become adults (Sakai, 2020). Because men have greater synapse density than women, the peak for men is predicted to be higher than that for women. Also, researchers found that the synapse density in layer 3 of the middle frontal gyrus increased till about age one, decreased thereafter until age 16 due to synaptic pruning, which removes unused synapses, and then remains constant till age 72 before decreasing significantly (Huttenlocher, 1979). Men are shown to follow the previously stated trend while this figure predicts women to experience a notable decrease in density closer to menopause. This figure is not to scale and only general trends and relative levels are shown (Created with BioRender.com).
9.9. Synapse dysfunction in Alzheimer’s disease
In AD, synaptic dysfunction leads to impaired neurotransmission, thereby contributing to a variety of pathophysiologies that are observed in AD (John and Reddy, 2021). The deterioration of synapses occurs early on in AD (Shankar and Walsh, 2009). Synaptic damage can occur due to disruption of mitochondrial trafficking (covered in a different section), the presence of toxic substances in the synapse, and due to mitochondrial dysfunction.
9.9.1. Aβ and synapse dysfunction
The accumulation of Aβ has been shown to contribute to neurotoxicity and corresponding dysfunction and damage in the synapse. Aβ disrupts the release of neurotransmitters, regeneration of synaptic vesicles, and anterograde transport of mitochondria to the synapse (Calkins and Reddy, 2011; Rajmohan and Reddy, 2017; Chen et al., 2019b; John and Reddy, 2021; Morton et al., 2021; Pradeepkiran and Reddy, 2020). Furthermore, Aβ were found to promote N-methyl-D-aspartate (NMDA)-dependent Long-Term Depression (LTD) and inhibit NMDA-dependent LTP, thus leading to weaker synaptic connections (Marsh and Alifragis, 2018; Tu et al., 2014). LTD is also enhanced by decreased reuptake of glutamate by the synapse (Li et al., 2009).
9.9.2. P-tau and synapse dysfunction
P-tau has also been implicated in synaptic dysfunction during AD. Synaptogyrin-3, a protein that interacts with Tau and is involved in vesicle movement, was found to restrict the movement of synaptic vesicles and decrease the release of neurotransmitters from vesicles in the presence of Tau (Zhou et al., 2017; McInnes et al., 2018; John an Reddy, 2021). Tau was also found to interact with microtubules, thus interfering with the binding of dyneins and kinesins with microtubules, thereby decreasing the movement of mitochondria via axonal transport (Dixit et al., 2008; Cheng and Bai, 2018), thus diminishing neurotransmission. Synaptic activity has been found to increase the spread of Tau to synapses, further leading to synaptic dysfunction (Calafate et al., 2015). Researchers found that the interactions between hyperphosphorylated tau and Drp1 lead to enhanced mitochondrial fission, thus leading to decreased functional mitochondria present in the synapse (Manczak and Reddy, 2012). Due to these pathologies, the number of synapses decreases in the brain due to degradation (Kashyap et al., 2019). Synaptic dysfunction contributes to slower and disrupted neurotransmission, leading to the memory loss and cognitive impairment that is characteristic of AD. As synapse continues to deteriorate and neurons die as a result, so does the neural processing capabilities of those with AD, thereby leading to dementia and the progression of AD. Recently our lab identified the critical synaptic miRNAs and their function in AD (Kumar and Reddy, 2020). Mitochondrial miRNAs are involved in various synaptic functions. Table 4 shows mitochondrial miRNAs that are specifically associated with various synaptic functions. This table highlights target genes, the dysregulation of these miRNAs in AD, and how they are related to synaptic function. The downregulation of these genes by binding to the 3’UTR of target mRNAs are found to contribute to various synaptic functions that are listed in this table. Figure 8 depicts which mitochondria miRNAs are associated with each synaptic function, whether these miRNAs are upregulated or downregulated in AD, and whether these miRNAs promote or inhibit a certain synaptic function. Understanding how synaptic dysfunction occurs in AD will allow researchers a better understanding of how AD progresses and determine potential targets for treatment.
Table 4.
Mitochondrial miRNAs associated with Synaptic Function.
| Sl. No. | microRNA | Function | Target Genes | Regulation in AD | References |
|---|---|---|---|---|---|
| 1 | mir-34a | Impacts presynaptic and postsynaptic activity and promotes the dysfunction of synaptic plasticity | VAMP2, SYT1, HCN, NR2A, GLUR1 | Upregulated in AD | (Sarkar et al., 2016) |
| 2 | miR-132/212 | Enhance neurotransmission and involved in synaptic plasticity | PTEN, FOXO3a, P300, NOS1, MMP-9 (miR-132), p250GAP (miR-132) | Downregulated in AD | (Wayman et al., 2008; Lambert et al., 2010; Remenyi et al., 2013; Wong et al., 2013; Jasinska et al., 2016; Wang et al., 2017) |
| 3 | miR-218 | Downregulates protein involved in protection against neurotoxins, thus potentially resulting in synaptotoxicity | PRKN | Downregulated in AD | (Di Rita et al., 2020) |
| 4 | miR-455-3p | Enhances synaptic activity and is associated with the upregulation of the synaptic genes, synaptophysin and PSD95 | APP | Upregulated in AD | (Kumar and Reddy, 2019) |
| 5 | miR-484 | Potentially plays role in neurotransmission and long-term plasticity | Fis1, BCL2L13, 46 genes in common between predicted targets of miR-484 and module M1 turquoise | Downregulated in AD | (Wingo et al., 2020) |
Fig 8. Mitochondrial miRNAs and synapse function in Alzheimer’s disease.

Mitochondrial miRNAs that are shown in the figure are associated with synaptic functions and deregulated in AD. The green and red arrows indicate the upregulation or downregulation, respectively, of the miRNA in AD. miR-455-3p and miR-218 seem to have a neuroprotective impact on neurons while miR-34a, miR-132, and miR-212 are associated with synaptic dysfunction (Created with BioRender.com).
9.10. Mitochondrial miRNAs and synaptic activity
Synaptic activity is essential for neurotransmission and brain function. The impairment of this activity can lead to neuronal death, weakened transmission, and decreased synaptic plasticity. Decreased synaptic activity leads to slower neural processing and decreases a person’s ability to form or access long-term memories. Two mitochondrial miRNAs have been found to enhance synaptic activity: miR-455-3p and miR-34a. miR-455-3p was found to enhance synaptic activity, thus the upregulation of miR-455-3p in AD indicates that this miRNA serves a protective role in neurons and can be used as a biomarker for AD (Kumar and Reddy, 2019). In addition, miR-34a leads to dysfunction in resting presynaptic and postsynaptic activities, thus the upregulation of this miRNA in AD shows that this miRNA could be involved in the pathogenesis of AD (Sarkar et al., 2016).
9.11. Mitochondrial miRNAs and synaptotoxicity
Protection against toxic molecules is a major function of synapses. Synaptotoxicity results in dysfunctional synapses, thus impairing neurotransmission and leading to neuronal death. miR-218 is a mitochondrial miRNA that was found to downregulate PRKN (Di Rita et al., 2020). The PRKN plays a role in safeguarding neurons from toxins and metallic ions, thus allowing for synapses to remain functional (Di Rita et al., 2020). The downregulation of miR-218 in AD, suggests that this miRNA may potentially have a protective effect against synaptotoxicity, thus showing how maintaining miR-218 levels in neurons could be of importance in neuronal survival.
9.12. Mitochondrial miRNAs and neurotransmission
The primary role of synapse in neurons is neurotransmission, which is accomplished through the release of neurotransmitters. A few mitochondrial miRNAs (miR-484, miR-132, and miR-212) are related to this vital function. miR-132 and miR-212 were found to enhance neurotransmission, thus the impairment of these miRNAs in AD leads to decreased neurotransmission, thus limiting neural processing capabilities by those with AD (Remenyi et al., 2013). miR-484 was also found to be involved in neurotransmission, thus the deregulation of this miRNA has the potential to also alter signal transmission in AD patients (Wingo et al., 2020). Therefore, mitochondrial miRNAs are important to neurotransmission as altered levels of these miRNAs in neurons can contribute to disease.
9.13. Mitochondrial miRNAs and synaptic plasticity
Synaptic plasticity is essential for memory and learning. Mitochondria provide the energy necessary for plasticity and aid in calcium buffering that occurs at the synapse. Mitochondrial miRNAs have been found to be involved in synaptic plasticity. miR-132 (Lambert et al., 2010) has a role in regulating short-term plasticity while miR-484 (Wingo et al., 2020) impacts long-term plasticity. Both miRNAs are deregulated in AD. miR-34a is associated with the dysfunction of synaptic plasticity and is upregulated in AD, thus this miRNA could be implicated in the pathogenesis of AD (Sarkar et al., 2016). These miRNAs are most likely regulated to where synaptic plasticity is defective and existing connections are damaged.
Conclusions and future directions
The science of mitochondrial microRNAs is emerging and relatively new in mitochondrial and synapse areas of research in Alzheimer’s disease. It is known that miRNAs play important roles in performing mitochondrial functions. Naturally, mitochondrial miRNAs are of higher interest to researchers pursuing to develop biomarkers, treatments, and/or targets for the treatment of AD. However, knowledge on mitochondrial miRNAs affecting specific synaptic functions is limited. For example, no miRNA has been associated with mitochondrial trafficking, Ca2+ signaling, and synaptic vesicle formation, all of which are important in neurotransmission. Moreover, only a few mitochondrial miRNAs were associated with synaptic activity, neurotransmission, synaptic plasticity, and neurotoxicity. Identifying more mitochondrial miRNAs in the synapse associated with these functions and whether they are deregulated in AD or not will give a greater understanding of which miRNA should be of focus when analyzing the synaptic dysfunction present in AD.
In this meta-analysis study, we have identified 24 mitochondrial miRNAs that are associated with AD (Table 2) and mostly binding to the 3’ UTR of the mRNA. For a better understanding, these miRNAs are classified into major mitochondrial functions, including bioenergetics, redox homeostasis, mitophagy, biogenesis, mitochondrial dynamics, and apoptosis. In addition, five mitochondrial miRNAs were found to be related to synaptic functions, including synaptic activity, neurotransmission, neurotoxicity, and synaptic plasticity. Many of the miRNAs are found to be involved in more than one function and are associated with other neurodegenerative diseases such as PD, HD, and ALS. Many of the studies that we reviewed also indicated that many miRNAs contribute to mitochondrial dysfunction in AD brains. However, which miRNAs initiate and contribute the most to the development and progression of AD is still unknown. Therefore, more research is needed to determine interrelationships among the different mitochondrial functions and their miRNAs.
Further exploration of mechanisms and proteins involved in mitochondrial and synaptic dysfunction is needed. Recently, defective mitophagy has been hypothesized as an early cellular event in neurodegenerative diseases. More research is needed to understand how mitochondrial miRNAs regulate mitophagy and how the deregulation of these miRNAs occurs. Further, how miRNAs localize to the mitochondria to exert their effects needs to be discovered. Enhancing synaptic activity can serve as one possible method of treatment for AD. In addition, researchers need to develop ways to decrease the harmful effects that Aβ and p-tau have on mitochondrial dysfunction.
Research on treatment strategies is of utmost importance. Mitochondrial miRNAs have a major potential to be used as biomarkers for the early detection and treating AD. Researchers can explore what happens when certain mitochondrial miRNAs are added at greater levels or completely knocked out and how this affects mitochondrial function in both WT and AD mice to determine the potential use of certain mitochondrial miRNA as treatments. Also, sex hormones are being analyzed as potential treatments for AD. Although many researchers are analyzed the protective effects of sex hormones, no successful treatment has been developed. Therefore, there are research opportunities to further explore sex hormones to identify potential biomarkers of AD, allowing for earlier diagnosis and to the development of treatments, and/or targets for treatment to prevent the progression of this disease.
Highlights.
Mitochondrial dysfunction is one of the major hallmarks of AD.
Mitochondrial miRNAs are crucial for mitochondrial functions in neurotransmission at synapse.
Mitochondria works differently with age and gender in AD.
Synapse numbers varies with age and gender in AD.
Mitochondrial miRNAs are potential targets for mitochondria and synapses in AD.
Acknowledgements
The authors would like to thank NIH for funding various projects - R01AG042178, R01AG47812, R01NS105473, AG060767, AG069333, and AG066347 to (P.H.R) and K99AG065645 to (S.K.).
Footnotes
Declaration of competing interest
We would like to inform you that we have a pending patent ‘MicroRNA-455-3p as a Potential Peripheral Biomarker for Alzheimer’s Disease (US 20200255900)’ related to the contents of our manuscript.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- Ali S, McStay GP, 2018. Regulation of Mitochondrial Dynamics by Proteolytic Processing and Protein Turnover. Antioxidants (Basel) 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Almanza A, Carlesso A, Chintha C, Creedican S, Doultsinos D, Leuzzi B, Luis A, McCarthy N, Montibeller L, More S, Papaioannou A, Puschel F, Sassano ML, Skoko J, Agostinis P, de Belleroche J, Eriksson LA, Fulda S, Gorman AM, Healy S, Kozlov A, Munoz-Pinedo C, Rehm M, Chevet E, Samali A, 2019. Endoplasmic reticulum stress signalling - from basic mechanisms to clinical applications. FEBS J 286, 241–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alonso-Nanclares L, Gonzalez-Soriano J, Rodriguez JR, DeFelipe J, 2008. Gender differences in human cortical synaptic density. Proc Natl Acad Sci U S A 105, 14615–14619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alzheimer’sAssociation, 2021. 2021 Alzheimer’s disease facts and figures. Alzheimers Dement 17, 327–406. [DOI] [PubMed] [Google Scholar]
- Ambrus AM, Frolov MV, 2009. The diverse roles of RNA helicases in RNAi. Cell Cycle 8, 3500–3505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ames A 3rd, 2000. CNS energy metabolism as related to function. Brain Res Brain Res Rev 34, 42–68. [DOI] [PubMed] [Google Scholar]
- Ankarberg C, Norjavaara E, 1999. Diurnal rhythm of testosterone secretion before and throughout puberty in healthy girls: correlation with 17beta-estradiol and dehydroepiandrosterone sulfate. J Clin Endocrinol Metab 84, 975–984. [DOI] [PubMed] [Google Scholar]
- Anyanwu EC, 2007. Neurochemical changes in the aging process: implications in medication in the elderly. ScientificWorldJournal 7, 1603–1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arimon M, Takeda S, Post KL, Svirsky S, Hyman BT, Berezovska O, 2015. Oxidative stress and lipid peroxidation are upstream of amyloid pathology. Neurobiol Dis 84, 109–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aschrafi A, Schwechter AD, Mameza MG, Natera-Naranjo O, Gioio AE, Kaplan BB, 2008. MicroRNA-338 regulates local cytochrome c oxidase IV mRNA levels and oxidative phosphorylation in the axons of sympathetic neurons. J Neurosci 28, 12581–12590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ashrafian H, Zadeh EH, Khan RH, 2021. Review on Alzheimer’s disease: Inhibition of amyloid beta and tau tangle formation. Int J Biol Macromol 167, 382–394. [DOI] [PubMed] [Google Scholar]
- Ayodele T, Rogaeva E, Kurup JT, Beecham G, Reitz C, 2021. Early-Onset Alzheimer’s Disease: What Is Missing in Research? Curr Neurol Neurosci Rep 21, 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Azam S, Haque ME, Kim IS, Choi DK, 2021. Microglial Turnover in Ageing-Related Neurodegeneration: Therapeutic Avenue to Intervene in Disease Progression. Cells 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Azevedo FA, Carvalho LR, Grinberg LT, Farfel JM, Ferretti RE, Leite RE, Jacob Filho W, Lent R, Herculano-Houzel S, 2009. Equal numbers of neuronal and nonneuronal cells make the human brain an isometrically scaled-up primate brain. J Comp Neurol 513, 532–541. [DOI] [PubMed] [Google Scholar]
- Ba Q, Cui C, Wen L, Feng S, Zhou J, Yang K, 2015. Schisandrin B shows neuroprotective effect in 6-OHDA-induced Parkinson’s disease via inhibiting the negative modulation of miR-34a on Nrf2 pathway. Biomed Pharmacother 75, 165–172. [DOI] [PubMed] [Google Scholar]
- Bai XY, Ma Y, Ding R, Fu B, Shi S, Chen XM, 2011. miR-335 and miR-34a Promote renal senescence by suppressing mitochondrial antioxidative enzymes. J Am Soc Nephrol 22, 1252–1261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bakula D, Scheibye-Knudsen M, 2020. MitophAging: Mitophagy in Aging and Disease. Front Cell Dev Biol 8, 239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bandiera S, Ruberg S, Girard M, Cagnard N, Hanein S, Chretien D, Munnich A, Lyonnet S, Henrion-Caude A, 2011. Nuclear outsourcing of RNA interference components to human mitochondria. PLoS One 6, e20746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bargaje R, Gupta S, Sarkeshik A, Park R, Xu T, Sarkar M, Halimani M, Roy SS, Yates J, Pillai B, 2012. Identification of novel targets for miR-29a using miRNA proteomics. PLoS One 7, e43243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barrey E, Saint-Auret G, Bonnamy B, Damas D, Boyer O, Gidrol X, 2011. Pre-microRNA and mature microRNA in human mitochondria. PLoS One 6, e20220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baudry M, Bi X, Aguirre C, 2013. Progesterone-estrogen interactions in synaptic plasticity and neuroprotection. Neuroscience 239, 280–294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Belmaker RH, Agam G, 2008. Major depressive disorder. N Engl J Med 358, 55–68. [DOI] [PubMed] [Google Scholar]
- Benaroya H, 2020. Brain energetics, mitochondria, and traumatic brain injury. Rev Neurosci 31, 363–390. [DOI] [PubMed] [Google Scholar]
- Bender A, Krishnan KJ, Morris CM, Taylor GA, Reeve AK, Perry RH, Jaros E, Hersheson JS, Betts J, Klopstock T, Taylor RW, Turnbull DM, 2006. High levels of mitochondrial DNA deletions in substantia nigra neurons in aging and Parkinson disease. Nat Genet 38, 515–517. [DOI] [PubMed] [Google Scholar]
- Berridge MJ, 2014. Calcium regulation of neural rhythms, memory and Alzheimer’s disease. J Physiol 592, 281–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhattacharyya A, Chattopadhyay R, Mitra S, Crowe SE, 2014. Oxidative stress: an essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol Rev 94, 329–354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhatti JS, Bhatti GK, Reddy PH, 2017. Mitochondrial dysfunction and oxidative stress in metabolic disorders - A step towards mitochondria based therapeutic strategies. Biochim Biophys Acta Mol Basis Dis 1863, 1066–1077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhatti JS, Tamarai K, Kandimalla R, Manczak M, Yin X, Ramasubramanian B, Sawant N, Pradeepkiran JA, Vijayan M, Kumar S, Reddy PH, 2021. Protective effects of a mitochondria-targeted small peptide SS31 against hyperglycemia-induced mitochondrial abnormalities in the liver tissues of diabetic mice, Tallyho/JngJ mice. Mitochondrion 58, 49–58. [DOI] [PubMed] [Google Scholar]
- Bhinge A, Namboori SC, Bithell A, Soldati C, Buckley NJ, Stanton LW, 2016. MiR-375 is Essential for Human Spinal Motor Neuron Development and May Be Involved in Motor Neuron Degeneration. Stem Cells 34, 124–134. [DOI] [PubMed] [Google Scholar]
- Bian Z, Li LM, Tang R, Hou DX, Chen X, Zhang CY, Zen K, 2010. Identification of mouse liver mitochondria-associated miRNAs and their potential biological functions. Cell Res 20, 1076–1078. [DOI] [PubMed] [Google Scholar]
- Bishop NA, Lu T, Yankner BA, 2010. Neural mechanisms of ageing and cognitive decline. Nature 464, 529–535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bohnsack MT, Czaplinski K, Gorlich D, 2004. Exportin 5 is a RanGTP-dependent dsRNA-binding protein that mediates nuclear export of pre-miRNAs. RNA 10, 185–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Britti E, Delaspre F, Tamarit J, Ros J, 2018. Mitochondrial calcium signalling and neurodegenerative diseases. Neuronal Signal 2, NS20180061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brodaty H, Donkin M, 2009. Family caregivers of people with dementia. Dialogues Clin Neurosci 11, 217–228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bulgart HR, Neczypor EW, Wold LE, Mackos AR, 2020. Microbial involvement in Alzheimer disease development and progression. Mol Neurodegener 15, 42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Butterfield DA, Boyd-Kimball D, 2020. Mitochondrial Oxidative and Nitrosative Stress and Alzheimer Disease. Antioxidants (Basel) 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Butterfield DA, Reed TT, Perluigi M, De Marco C, Coccia R, Keller JN, Markesbery WR, Sultana R, 2007. Elevated levels of 3-nitrotyrosine in brain from subjects with amnestic mild cognitive impairment: implications for the role of nitration in the progression of Alzheimer’s disease. Brain Res 1148, 243–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai Q, Jeong YY, 2020. Mitophagy in Alzheimer’s Disease and Other Age-Related Neurodegenerative Diseases. Cells 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai Q, Sheng ZH, 2009. Mitochondrial transport and docking in axons. Exp Neurol 218, 257–267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai Q, Tammineni P, 2016. Alterations in Mitochondrial Quality Control in Alzheimer’s Disease. Front Cell Neurosci 10, 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai Q, Tammineni P, 2017. Mitochondrial Aspects of Synaptic Dysfunction in Alzheimer’s Disease. J Alzheimers Dis 57, 1087–1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calafate S, Buist A, Miskiewicz K, Vijayan V, Daneels G, de Strooper B, de Wit J, Verstreken P, Moechars D, 2015. Synaptic Contacts Enhance Cell-to-Cell Tau Pathology Propagation. Cell Rep 11, 1176–1183. [DOI] [PubMed] [Google Scholar]
- Calkins MJ, Manczak M, Mao P, Shirendeb U, Reddy PH, 2011. Impaired mitochondrial biogenesis, defective axonal transport of mitochondria, abnormal mitochondrial dynamics and synaptic degeneration in a mouse model of Alzheimer’s disease. Hum Mol Genet 20, 4515–4529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calkins MJ, Manczak M, Reddy PH, 2012. Mitochondria-Targeted Antioxidant SS31 Prevents Amyloid Beta-Induced Mitochondrial Abnormalities and Synaptic Degeneration in Alzheimer’s Disease. Pharmaceuticals (Basel) 5, 1103–1119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calkins MJ, Reddy PH, 2011. Amyloid beta impairs mitochondrial anterograde transport and degenerates synapses in Alzheimer’s disease neurons. Biochim Biophys Acta 1812, 507–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calvo-Rodriguez M, Hou SS, Snyder AC, Kharitonova EK, Russ AN, Das S, Fan Z, Muzikansky A, Garcia-Alloza M, Serrano-Pozo A, Hudry E, Bacskai BJ, 2020. Increased mitochondrial calcium levels associated with neuronal death in a mouse model of Alzheimer’s disease. Nat Commun 11, 2146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Camara AKS, Zhou Y, Wen PC, Tajkhorshid E, Kwok WM, 2017. Mitochondrial VDAC1: A Key Gatekeeper as Potential Therapeutic Target. Front Physiol 8, 460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Campos-Melo D, Droppelmann CA, He Z, Volkening K, Strong MJ, 2013. Altered microRNA expression profile in Amyotrophic Lateral Sclerosis: a role in the regulation of NFL mRNA levels. Mol Brain 6, 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cannon-Albright LA, Foster NL, Schliep K, Farnham JM, Teerlink CC, Kaddas H, Tschanz J, Corcoran C, Kauwe JSK, 2019. Relative risk for Alzheimer disease based on complete family history. Neurology 92, e1745–e1753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Catterall WA, 2011. Voltage-gated calcium channels. Cold Spring Harb Perspect Biol 3, a003947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chakrabarti S, Khemka VK, Banerjee A, Chatterjee G, Ganguly A, Biswas A, 2015. Metabolic Risk Factors of Sporadic Alzheimer’s Disease: Implications in the Pathology, Pathogenesis and Treatment. Aging Dis 6, 282–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chamberlain KA, Sheng ZH, 2019. Mechanisms for the maintenance and regulation of axonal energy supply. J Neurosci Res 97, 897–913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang DT, Honick AS, Reynolds IJ, 2006. Mitochondrial trafficking to synapses in cultured primary cortical neurons. J Neurosci 26, 7035–7045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chaudhuri AD, Choi DC, Kabaria S, Tran A, Junn E, 2016. MicroRNA-7 Regulates the Function of Mitochondrial Permeability Transition Pore by Targeting VDAC1 Expression. J Biol Chem 291, 6483–6493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheignon C, Tomas M, Bonnefont-Rousselot D, Faller P, Hureau C, Collin F, 2018. Oxidative stress and the amyloid beta peptide in Alzheimer’s disease. Redox Biol 14, 450–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen FZ, Zhao Y, Chen HZ, 2019a. MicroRNA-98 reduces amyloid beta-protein production and improves oxidative stress and mitochondrial dysfunction through the Notch signaling pathway via HEY2 in Alzheimer’s disease mice. Int J Mol Med 43, 91–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y, Fu AKY, Ip NY, 2019b. Synaptic dysfunction in Alzheimer’s disease: Mechanisms and therapeutic strategies. Pharmacol Ther 195, 186–198. [DOI] [PubMed] [Google Scholar]
- Chen Z, Li Y, Zhang H, Huang P, Luthra R, 2010. Hypoxia-regulated microRNA-210 modulates mitochondrial function and decreases ISCU and COX10 expression. Oncogene 29, 4362–4368. [DOI] [PubMed] [Google Scholar]
- Cheng Y, Bai F, 2018. The Association of Tau With Mitochondrial Dysfunction in Alzheimer’s Disease. Front Neurosci 12, 163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chistiakov DA, Sobenin IA, Revin VV, Orekhov AN, Bobryshev YV, 2014. Mitochondrial aging and age-related dysfunction of mitochondria. Biomed Res Int 2014, 238463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chodick G, Epstein S, Shalev V, 2020. Secular trends in testosterone- findings from a large state-mandate care provider. Reprod Biol Endocrinol 18, 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choi GE, Han HJ, 2021. Glucocorticoid impairs mitochondrial quality control in neurons. Neurobiol Dis 152, 105301. [DOI] [PubMed] [Google Scholar]
- Chung WS, Allen NJ, Eroglu C, 2015. Astrocytes Control Synapse Formation, Function, and Elimination. Cold Spring Harb Perspect Biol 7, a020370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cobley JN, Fiorello ML, Bailey DM, 2018. 13 reasons why the brain is susceptible to oxidative stress. Redox Biol 15, 490–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cogswell JP, Ward J, Taylor IA, Waters M, Shi Y, Cannon B, Kelnar K, Kemppainen J, Brown D, Chen C, Prinjha RK, Richardson JC, Saunders AM, Roses AD, Richards CA, 2008. Identification of miRNA changes in Alzheimer’s disease brain and CSF yields putative biomarkers and insights into disease pathways. J Alzheimers Dis 14, 27–41. [DOI] [PubMed] [Google Scholar]
- Cooper GM, 2000. The cell : a molecular approach, 2nd ed. ASM Press; Sinauer Associates, Washington, D.C. Sunderland, Mass. [Google Scholar]
- Copeland WC, 2010. The mitochondrial DNA polymerase in health and disease. Subcell Biochem 50, 211–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Correia SC, Perry G, Moreira PI, 2016. Mitochondrial traffic jams in Alzheimer’s disease - pinpointing the roadblocks. Biochim Biophys Acta 1862, 1909–1917. [DOI] [PubMed] [Google Scholar]
- Correia SC, Resende R, Moreira PI, Pereira CM, 2015. Alzheimer’s disease-related misfolded proteins and dysfunctional organelles on autophagy menu. DNA Cell Biol 34, 261–273. [DOI] [PubMed] [Google Scholar]
- Correia SC, Santos RX, Cardoso S, Carvalho C, Candeias E, Duarte AI, Placido AI, Santos MS, Moreira PI, 2012. Alzheimer disease as a vascular disorder: where do mitochondria fit? Exp Gerontol 47, 878–886. [DOI] [PubMed] [Google Scholar]
- Course MM, Wang X, 2016. Transporting mitochondria in neurons. F1000Res 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cullen NC, Malarstig AN, Stomrud E, Hansson O, Mattsson-Carlgren N, 2021. Accelerated inflammatory aging in Alzheimer’s disease and its relation to amyloid, tau, and cognition. Sci Rep 11, 1965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- da Silva FC, Iop RD, Vietta GG, Kair DA, Gutierres Filho PJ, de Alvarenga JG, da Silva R, 2016. microRNAs involved in Parkinson’s disease: A systematic review. Mol Med Rep 14, 4015–4022. [DOI] [PubMed] [Google Scholar]
- Davison SL, Bell R, Donath S, Montalto JG, Davis SR, 2005. Androgen levels in adult females: changes with age, menopause, and oophorectomy. J Clin Endocrinol Metab 90, 3847–3853. [DOI] [PubMed] [Google Scholar]
- De Felice B, Manfellotto F, Fiorentino G, Annunziata A, Biffali E, Pannone R, Federico A, 2018. Wide-Ranging Analysis of MicroRNA Profiles in Sporadic Amyotrophic Lateral Sclerosis Using Next-Generation Sequencing. Front Genet 9, 310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Luna N, Turon-Sans J, Cortes-Vicente E, Carrasco-Rozas A, Illan-Gala I, Dols-Icardo O, Clarimon J, Lleo A, Gallardo E, Illa I, Rojas-Garcia R, 2020. Downregulation of miR-335-5P in Amyotrophic Lateral Sclerosis Can Contribute to Neuronal Mitochondrial Dysfunction and Apoptosis. Sci Rep 10, 4308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deheshi S, Pasqualotto BA, Rintoul GL, 2013. Mitochondrial trafficking in neuropsychiatric diseases. Neurobiol Dis 51, 66–71. [DOI] [PubMed] [Google Scholar]
- Dei R, Takeda A, Niwa H, Li M, Nakagomi Y, Watanabe M, Inagaki T, Washimi Y, Yasuda Y, Horie K, Miyata T, Sobue G, 2002. Lipid peroxidation and advanced glycation end products in the brain in normal aging and in Alzheimer’s disease. Acta Neuropathol 104, 113–122. [DOI] [PubMed] [Google Scholar]
- Deperrois N, Graupner M, 2020. Short-term depression and long-term plasticity together tune sensitive range of synaptic plasticity. PLoS Comput Biol 16, e1008265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Descalzi G, Gao V, Steinman MQ, Suzuki A, Alberini CM, 2019. Lactate from astrocytes fuels learning-induced mRNA translation in excitatory and inhibitory neurons. Commun Biol 2, 247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeTure MA, Dickson DW, 2019. The neuropathological diagnosis of Alzheimer’s disease. Mol Neurodegener 14, 32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Devine MJ, Kittler JT, 2018. Mitochondria at the neuronal presynapse in health and disease. Nat Rev Neurosci 19, 63–80. [DOI] [PubMed] [Google Scholar]
- Di Domenico F, Tramutola A, Butterfield DA, 2017. Role of 4-hydroxy-2-nonenal (HNE) in the pathogenesis of alzheimer disease and other selected age-related neurodegenerative disorders. Free Radic Biol Med 111, 253–261. [DOI] [PubMed] [Google Scholar]
- Di Rita A, Maiorino T, Bruqi K, Volpicelli F, Bellenchi GC, Strappazzon F, 2020. miR-218 Inhibits Mitochondrial Clearance by Targeting PRKN E3 Ubiquitin Ligase. Int J Mol Sci 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Stefano F, Kas A, Habert MO, Decazes P, Lamari F, Lista S, Hampel H, Teichmann M, 2016. The phenotypical core of Alzheimer’s disease-related and nonrelated variants of the corticobasal syndrome: A systematic clinical, neuropsychological, imaging, and biomarker study. Alzheimers Dement 12, 786–795. [DOI] [PubMed] [Google Scholar]
- Diez-Planelles C, Sanchez-Lozano P, Crespo MC, Gil-Zamorano J, Ribacoba R, Gonzalez N, Suarez E, Martinez-Descals A, Martinez-Camblor P, Alvarez V, Martin-Hernandez R, Huerta-Ruiz I, Gonzalez-Garcia I, Cosgaya JM, Visioli F, Davalos A, Iglesias-Gutierrez E, Tomas-Zapico C, 2016. Circulating microRNAs in Huntington’s disease: Emerging mediators in metabolic impairment. Pharmacol Res 108, 102–110. [DOI] [PubMed] [Google Scholar]
- Ding H, Huang Z, Chen M, Wang C, Chen X, Chen J, Zhang J, 2016. Identification of a panel of five serum miRNAs as a biomarker for Parkinson’s disease. Parkinsonism Relat Disord 22, 68–73. [DOI] [PubMed] [Google Scholar]
- Dixit R, Ross JL, Goldman YE, Holzbaur EL, 2008. Differential regulation of dynein and kinesin motor proteins by tau. Science 319, 1086–1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Drew B, Leeuwenburgh C, 2003. Method for measuring ATP production in isolated mitochondria: ATP production in brain and liver mitochondria of Fischer-344 rats with age and caloric restriction. Am J Physiol Regul Integr Comp Physiol 285, R1259–1267. [DOI] [PubMed] [Google Scholar]
- Dumanovic J, Nepovimova E, Natic M, Kuca K, Jacevic V, 2020. The Significance of Reactive Oxygen Species and Antioxidant Defense System in Plants: A Concise Overview. Front Plant Sci 11, 552969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Esteras N, Abramov AY, 2020. Mitochondrial Calcium Deregulation in the Mechanism of Beta-Amyloid and Tau Pathology. Cells 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fattoretti P, Malatesta M, Mariotti R, Zancanaro C, 2019. Testosterone administration increases synaptic density in the gyrus dentatus of old mice independently of physical exercise. Exp Gerontol 125, 110664. [DOI] [PubMed] [Google Scholar]
- Finkel T, Menazza S, Holmstrom KM, Parks RJ, Liu J, Sun J, Liu J, Pan X, Murphy E, 2015. The ins and outs of mitochondrial calcium. Circ Res 116, 1810–1819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Finkelstein JS, Yu EW, Burnett-Bowie SA, 2013. Gonadal steroids and body composition, strength, and sexual function in men. N Engl J Med 369, 2457. [DOI] [PubMed] [Google Scholar]
- Finnema SJ, Nabulsi NB, Eid T, Detyniecki K, Lin SF, Chen MK, Dhaher R, Matuskey D, Baum E, Holden D, Spencer DD, Mercier J, Hannestad J, Huang Y, Carson RE, 2016. Imaging synaptic density in the living human brain. Sci Transl Med 8, 348ra396. [DOI] [PubMed] [Google Scholar]
- Flippo KH, Strack S, 2017. Mitochondrial dynamics in neuronal injury, development and plasticity. J Cell Sci 130, 671–681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Forner S, Baglietto-Vargas D, Martini AC, Trujillo-Estrada L, LaFerla FM, 2017. Synaptic Impairment in Alzheimer’s Disease: A Dysregulated Symphony. Trends Neurosci 40, 347–357. [DOI] [PubMed] [Google Scholar]
- Foster TC, 1999. Involvement of hippocampal synaptic plasticity in age-related memory decline. Brain Res Brain Res Rev 30, 236–249. [DOI] [PubMed] [Google Scholar]
- Gaignard P, Frechou M, Liere P, Therond P, Schumacher M, Slama A, Guennoun R, 2018. Sex differences in brain mitochondrial metabolism: influence of endogenous steroids and stroke. J Neuroendocrinol 30. [DOI] [PubMed] [Google Scholar]
- Gaignard P, Frechou M, Schumacher M, Therond P, Mattern C, Slama A, Guennoun R, 2016. Progesterone reduces brain mitochondrial dysfunction after transient focal ischemia in male and female mice. J Cereb Blood Flow Metab 36, 562–568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaignard P, Liere P, Therond P, Schumacher M, Slama A, Guennoun R, 2017. Role of Sex Hormones on Brain Mitochondrial Function, with Special Reference to Aging and Neurodegenerative Diseases. Front Aging Neurosci 9, 406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gamba P, Staurenghi E, Testa G, Giannelli S, Sottero B, Leonarduzzi G, 2019. A Crosstalk Between Brain Cholesterol Oxidation and Glucose Metabolism in Alzheimer’s Disease. Front Neurosci 13, 556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao P, Tchernyshyov I, Chang TC, Lee YS, Kita K, Ochi T, Zeller KI, De Marzo AM, Van Eyk JE, Mendell JT, Dang CV, 2009. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature 458, 762–765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garcia GC, Bartol TM, Phan S, Bushong EA, Perkins G, Sejnowski TJ, Ellisman MH, Skupin A, 2019. Mitochondrial morphology provides a mechanism for energy buffering at synapses. Sci Rep 9, 18306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geekiyanage H, Chan C, 2011. MicroRNA-137/181c regulates serine palmitoyltransferase and in turn amyloid beta, novel targets in sporadic Alzheimer’s disease. J Neurosci 31, 14820–14830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gerrits E, Brouwer N, Kooistra SM, Woodbury ME, Vermeiren Y, Lambourne M, Mulder J, Kummer M, Moller T, Biber K, Dunnen W, De Deyn PP, Eggen BJL, Boddeke E, 2021. Distinct amyloid-beta and tau-associated microglia profiles in Alzheimer’s disease. Acta Neuropathol 141, 681–696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Giles RE, Blanc H, Cann HM, Wallace DC, 1980. Maternal inheritance of human mitochondrial DNA. Proc Natl Acad Sci U S A 77, 6715–6719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gillies GE, McArthur S, 2010. Estrogen actions in the brain and the basis for differential action in men and women: a case for sex-specific medicines. Pharmacol Rev 62, 155–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ginter E, Simko V, 2013. Women live longer than men. Bratisl Lek Listy 114, 45–49. [DOI] [PubMed] [Google Scholar]
- Gleichmann M, Mattson MP, 2011. Neuronal calcium homeostasis and dysregulation. Antioxid Redox Signal 14, 1261–1273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goh SY, Chao YX, Dheen ST, Tan EK, Tay SS, 2019. Role of MicroRNAs in Parkinson’s Disease. Int J Mol Sci 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gokerkucuk EB, Tramier M, Bertolin G, 2020. Imaging Mitochondrial Functions: from Fluorescent Dyes to Genetically-Encoded Sensors. Genes (Basel) 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gong G, An F, Wang Y, Bian M, Yu LJ, Wei C, 2017. miR-15b represses BACE1 expression in sporadic Alzheimer’s disease. Oncotarget 8, 91551–91557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gray LR, Tompkins SC, Taylor EB, 2014. Regulation of pyruvate metabolism and human disease. Cell Mol Life Sci 71, 2577–2604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greber BJ, Ban N, 2016. Structure and Function of the Mitochondrial Ribosome. Annu Rev Biochem 85, 103–132. [DOI] [PubMed] [Google Scholar]
- Green KN, 2009. Calcium in the initiation, progression and as an effector of Alzheimer’s disease pathology. J Cell Mol Med 13, 2787–2799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Green RC, Cupples LA, Go R, Benke KS, Edeki T, Griffith PA, Williams M, Hipps Y, Graff-Radford N, Bachman D, Farrer LA, Group MS, 2002. Risk of dementia among white and African American relatives of patients with Alzheimer disease. JAMA 287, 329–336. [DOI] [PubMed] [Google Scholar]
- Guerreiro R, Bras J, 2015. The age factor in Alzheimer’s disease. Genome Med 7, 106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guevara R, Gianotti M, Roca P, Oliver J, 2011. Age and sex-related changes in rat brain mitochondrial function. Cell Physiol Biochem 27, 201–206. [DOI] [PubMed] [Google Scholar]
- Guo L, Tian J, Du H, 2017. Mitochondrial Dysfunction and Synaptic Transmission Failure in Alzheimer’s Disease. J Alzheimers Dis 57, 1071–1086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hall OJ, Klein SL, 2017. Progesterone-based compounds affect immune responses and susceptibility to infections at diverse mucosal sites. Mucosal Immunol 10, 1097–1107. [DOI] [PubMed] [Google Scholar]
- Han S, Jeong YY, Sheshadri P, Cai Q, 2020. Mitophagy coordination with retrograde transport ensures the integrity of synaptic mitochondria. Autophagy 16, 1925–1927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hara Y, Waters EM, McEwen BS, Morrison JH, 2015. Estrogen Effects on Cognitive and Synaptic Health Over the Lifecourse. Physiol Rev 95, 785–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harris JJ, Jolivet R, Attwell D, 2012. Synaptic energy use and supply. Neuron 75, 762–777. [DOI] [PubMed] [Google Scholar]
- He Y, Wei M, Wu Y, Qin H, Li W, Ma X, Cheng J, Ren J, Shen Y, Chen Z, Sun B, Huang FD, Shen Y, Zhou YD, 2019. Amyloid beta oligomers suppress excitatory transmitter release via presynaptic depletion of phosphatidylinositol-4,5-bisphosphate. Nat Commun 10, 1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hebert SS, Horre K, Nicolai L, Papadopoulou AS, Mandemakers W, Silahtaroglu AN, Kauppinen S, Delacourte A, De Strooper B, 2008. Loss of microRNA cluster miR-29a/b-1 in sporadic Alzheimer’s disease correlates with increased BACE1/beta-secretase expression. Proc Natl Acad Sci U S A 105, 6415–6420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hendrie HC, Murrell J, Baiyewu O, Lane KA, Purnell C, Ogunniyi A, Unverzagt FW, Hall K, Callahan CM, Saykin AJ, Gureje O, Hake A, Foroud T, Gao S, 2014. APOE epsilon4 and the risk for Alzheimer disease and cognitive decline in African Americans and Yoruba. Int Psychogeriatr 26, 977–985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hirst J, King MS, Pryde KR, 2008. The production of reactive oxygen species by complex I. Biochem Soc Trans 36, 976–980. [DOI] [PubMed] [Google Scholar]
- Ho VM, Lee JA, Martin KC, 2011. The cell biology of synaptic plasticity. Science 334, 623–628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hondius DC, Koopmans F, Leistner C, Pita-Illobre D, Peferoen-Baert RM, Marbus F, Paliukhovich I, Li KW, Rozemuller AJM, Hoozemans JJM, Smit AB, 2021. The proteome of granulovacuolar degeneration and neurofibrillary tangles in Alzheimer’s disease. Acta Neuropathol 141, 341–358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hong H, Li Y, Su B, 2017. Identification of Circulating miR-125b as a Potential Biomarker of Alzheimer’s Disease in APP/PS1 Transgenic Mouse. J Alzheimers Dis 59, 1449–1458. [DOI] [PubMed] [Google Scholar]
- Horigane SI, Ozawa Y, Yamada H, Takemoto-Kimura S, 2019. Calcium signalling: a key regulator of neuronal migration. J Biochem 165, 401–409. [DOI] [PubMed] [Google Scholar]
- Hou X, Watzlawik JO, Cook C, Liu CC, Kang SS, Lin WL, DeTure M, Heckman MG, Diehl NN, Al-Shaikh FSH, Walton RL, Ross OA, Melrose HL, Ertekin-Taner N, Bu G, Petrucelli L, Fryer JD, Murray ME, Dickson DW, Fiesel FC, Springer W, 2020. Mitophagy alterations in Alzheimer’s disease are associated with granulovacuolar degeneration and early tau pathology. Alzheimers Dement. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoye ML, Regan MR, Jensen LA, Lake AM, Reddy LV, Vidensky S, Richard JP, Maragakis NJ, Rothstein JD, Dougherty JD, Miller TM, 2018. Motor neuron-derived microRNAs cause astrocyte dysfunction in amyotrophic lateral sclerosis. Brain 141, 2561–2575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoyt LT, Falconi AM, 2015. Puberty and perimenopause: reproductive transitions and their implications for women’s health. Soc Sci Med 132, 103–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang X, Le QT, Giaccia AJ, 2010. MiR-210--micromanager of the hypoxia pathway. Trends Mol Med 16, 230–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huttenlocher PR, 1979. Synaptic density in human frontal cortex - developmental changes and effects of aging. Brain Res 163, 195–205. [DOI] [PubMed] [Google Scholar]
- Hyer MM, Phillips LL, Neigh GN, 2018. Sex Differences in Synaptic Plasticity: Hormones and Beyond. Front Mol Neurosci 11, 266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishikawa K, Takenaga K, Akimoto M, Koshikawa N, Yamaguchi A, Imanishi H, Nakada K, Honma Y, Hayashi J, 2008. ROS-generating mitochondrial DNA mutations can regulate tumor cell metastasis. Science 320, 661–664. [DOI] [PubMed] [Google Scholar]
- Jasinska M, Milek J, Cymerman IA, Leski S, Kaczmarek L, Dziembowska M, 2016. miR-132 Regulates Dendritic Spine Structure by Direct Targeting of Matrix Metalloproteinase 9 mRNA. Mol Neurobiol 53, 4701–4712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jensen RE, Dunn CD, 2002. Protein import into and across the mitochondrial inner membrane: role of the TIM23 and TIM22 translocons. Biochim Biophys Acta 1592, 25–34. [DOI] [PubMed] [Google Scholar]
- John A, Kubosumi A, Reddy PH, 2020. Mitochondrial MicroRNAs in Aging and Neurodegenerative Diseases. Cells 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- John A, Reddy PH, 2021. Synaptic basis of Alzheimer’s disease: Focus on synaptic amyloid beta, P-tau and mitochondria. Ageing Res Rev 65, 101208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kametani F, Hasegawa M, 2018. Reconsideration of Amyloid Hypothesis and Tau Hypothesis in Alzheimer’s Disease. Front Neurosci 12, 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kandimalla R, Manczak M, Fry D, Suneetha Y, Sesaki H, Reddy PH, 2016. Reduced dynamin-related protein 1 protects against phosphorylated Tau-induced mitochondrial dysfunction and synaptic damage in Alzheimer’s disease. Hum Mol Genet 25, 4881–4897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kann O, Kovács R, 2007. Mitochondria and neuronal activity. Am J Physiol Cell Physiol 292, C641–657. [DOI] [PubMed] [Google Scholar]
- Kannurpatti SS, 2017. Mitochondrial calcium homeostasis: Implications for neurovascular and neurometabolic coupling. J Cereb Blood Flow Metab 37, 381–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karakuzu O, Cruz MR, Liu Y, Garsin DA, 2019. Amplex Red Assay for Measuring Hydrogen Peroxide Production from Caenorhabditis elegans. Bio Protoc 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kashyap G, Bapat D, Das D, Gowaikar R, Amritkar RE, Rangarajan G, Ravindranath V, Ambika G, 2019. Synapse loss and progress of Alzheimer’s disease -A network model. Sci Rep 9, 6555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kenyon CJ, 2010. The genetics of ageing. Nature 464, 504–512. [DOI] [PubMed] [Google Scholar]
- Kerr JS, Adriaanse BA, Greig NH, Mattson MP, Cader MZ, Bohr VA, Fang EF, 2017. Mitophagy and Alzheimer’s Disease: Cellular and Molecular Mechanisms. Trends Neurosci 40, 151–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim YJ, Kim SH, Park Y, Park J, Lee JH, Kim BC, Song WK, 2020. miR-16-5p is upregulated by amyloid beta deposition in Alzheimer’s disease models and induces neuronal cell apoptosis through direct targeting and suppression of BCL-2. Exp Gerontol 136, 110954. [DOI] [PubMed] [Google Scholar]
- Klinge CM, 2008. Estrogenic control of mitochondrial function and biogenesis. J Cell Biochem 105, 1342–1351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klingenberg M, 2008. The ADP and ATP transport in mitochondria and its carrier. Biochim Biophys Acta 1778, 1978–2021. [DOI] [PubMed] [Google Scholar]
- Kodavati M, Wang H, Hegde ML. Altered Mitochondrial Dynamics in Motor Neuron Disease: An Emerging Perspective. Cells. 2020;9:1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kovanda A, Leonardis L, Zidar J, Koritnik B, Dolenc-Groselj L, Ristic Kovacic S, Curk T, Rogelj B, 2018. Differential expression of microRNAs and other small RNAs in muscle tissue of patients with ALS and healthy age-matched controls. Sci Rep 8, 5609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kozlowska E, Krzyzosiak WJ, Koscianska E, 2013. Regulation of huntingtin gene expression by miRNA-137, −214, −148a, and their respective isomiRs. Int J Mol Sci 14, 16999–17016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kren BT, Wong PY, Sarver A, Zhang X, Zeng Y, Steer CJ, 2009. MicroRNAs identified in highly purified liver-derived mitochondria may play a role in apoptosis. RNA Biol 6, 65–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuhlbrandt W, 2015. Structure and function of mitochondrial membrane protein complexes. BMC Biol 13, 89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kujoth GC, Hiona A, Pugh TD, Someya S, Panzer K, Wohlgemuth SE, Hofer T, Seo AY, Sullivan R, Jobling WA, Morrow JD, Van Remmen H, Sedivy JM, Yamasoba T, Tanokura M, Weindruch R, Leeuwenburgh C, Prolla TA, 2005. Mitochondrial DNA mutations, oxidative stress, and apoptosis in mammalian aging. Science 309, 481–484. [DOI] [PubMed] [Google Scholar]
- Kumar S, Chawla YK, Ghosh S, Chakraborti A, 2014. Severity of hepatitis C virus (genotype-3) infection positively correlates with circulating microRNA-122 in patients sera. Dis Markers 2014, 435476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S, Batra A, Kanthaje S, Ghosh S, Chakraborti A, 2017. Crosstalk between microRNA-122 and FOX family genes in HepG2 cells. Exp Biol Med 242, 436–440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S, Reddy AP, Yin X, Reddy PH, 2019. Novel MicroRNA-455-3p and its protective effects against abnormal APP processing and amyloid beta toxicity in Alzheimer’s disease. Biochim Biophys Acta Mol Basis Dis 1865, 2428–2440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S, Reddy PH, 2016. Are circulating microRNAs peripheral biomarkers for Alzheimer’s disease? Biochim Biophys Acta 1862, 1617–1627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S, Reddy PH, 2019. A New Discovery of MicroRNA-455-3p in Alzheimer’s Disease. J Alzheimers Dis 72, S117–S130. [DOI] [PubMed] [Google Scholar]
- Kumar S, Vijayan M, Reddy PH, 2017. MicroRNA-455-3p as a potential peripheral biomarker for Alzheimer’s disease. Hum Mol Genet 26, 3808–3822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S, Reddy PH, (2020). The role of synaptic microRNAs in Alzheimer’s disease. Biochim Biophys Acta Mol Basis Dis. 866, 165937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lambert TJ, Storm DR, Sullivan JM, 2010. MicroRNA132 modulates short-term synaptic plasticity but not basal release probability in hippocampal neurons. PLoS One 5, e15182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lane CA, Hardy J, Schott JM, 2018. Alzheimer’s disease. Eur J Neurol 25, 59–70. [DOI] [PubMed] [Google Scholar]
- Lang A, Grether-Beck S, Singh M, Kuck F, Jakob S, Kefalas A, Altinoluk-Hambuchen S, Graffmann N, Schneider M, Lindecke A, Brenden H, Felsner I, Ezzahoini H, Marini A, Weinhold S, Vierkotter A, Tigges J, Schmidt S, Stuhler K, Kohrer K, Uhrberg M, Haendeler J, Krutmann J, Piekorz RP, 2016. MicroRNA-15b regulates mitochondrial ROS production and the senescence-associated secretory phenotype through sirtuin 4/SIRT4. Aging (Albany NY) 8, 484–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lanza IR, Nair KS, 2010. Mitochondrial metabolic function assessed in vivo and in vitro. Curr Opin Clin Nutr Metab Care 13, 511–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Larriba E, Rial E, Del Mazo J, 2018. The landscape of mitochondrial small non-coding RNAs in the PGCs of male mice, spermatogonia, gametes and in zygotes. BMC Genomics 19, 634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lax NZ, Turnbull DM, Reeve AK, 2011. Mitochondrial mutations: newly discovered players in neuronal degeneration. Neuroscientist 17, 645–658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee ST, Chu K, Im WS, Yoon HJ, Im JY, Park JE, Park KH, Jung KH, Lee SK, Kim M, Roh JK, 2011. Altered microRNA regulation in Huntington’s disease models. Exp Neurol 227, 172–179. [DOI] [PubMed] [Google Scholar]
- Leggio L, Vivarelli S, L’Episcopo F, Tirolo C, Caniglia S, Testa N, Marchetti B, Iraci N, 2017. microRNAs in Parkinson’s Disease: From Pathogenesis to Novel Diagnostic and Therapeutic Approaches. Int J Mol Sci 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lejri I, Grimm A, Eckert A, 2018. Mitochondria, Estrogen and Female Brain Aging. Front Aging Neurosci 10, 124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lenaz G, 2001. The mitochondrial production of reactive oxygen species: mechanisms and implications in human pathology. IUBMB Life 52, 159–164. [DOI] [PubMed] [Google Scholar]
- Leung AKL, 2015. The Whereabouts of microRNA Actions: Cytoplasm and Beyond. Trends Cell Biol 25, 601–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J, Donath S, Li Y, Qin D, Prabhakar BS, Li P, 2010. miR-30 regulates mitochondrial fission through targeting p53 and the dynamin-related protein-1 pathway. PLoS Genet 6, e1000795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J, Li Y, Jiao J, Wang J, Li Y, Qin D, Li P, 2014a. Mitofusin 1 is negatively regulated by microRNA 140 in cardiomyocyte apoptosis. Mol Cell Biol 34, 1788–1799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li JJ, Dolios G, Wang R, Liao FF, 2014b. Soluble beta-amyloid peptides, but not insoluble fibrils, have specific effect on neuronal microRNA expression. PLoS One 9, e90770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li N, Pan X, Zhang J, Ma A, Yang S, Ma J, Xie A, 2017. Plasma levels of miR-137 and miR-124 are associated with Parkinson’s disease but not with Parkinson’s disease with depression. Neurol Sci 38, 761–767. [DOI] [PubMed] [Google Scholar]
- Li S, Hong S, Shepardson NE, Walsh DM, Shankar GM, Selkoe D, 2009. Soluble oligomers of amyloid Beta protein facilitate hippocampal long-term depression by disrupting neuronal glutamate uptake. Neuron 62, 788–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li W, Zhang X, Zhuang H, Chen HG, Chen Y, Tian W, Wu W, Li Y, Wang S, Zhang L, Chen Y, Li L, Zhao B, Sui S, Hu Z, Feng D, 2014c. MicroRNA-137 is a novel hypoxia-responsive microRNA that inhibits mitophagy via regulation of two mitophagy receptors FUNDC1 and NIX. J Biol Chem 289, 10691–10701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liang C, Mu Y, Tian H, Wang D, Zhang S, Wang H, Liu Y, Di C, 2021. MicroRNA-140 silencing represses the incidence of Alzheimer’s disease. Neurosci Lett 758, 135674. [DOI] [PubMed] [Google Scholar]
- Liang WS, Reiman EM, Valla J, Dunckley T, Beach TG, Grover A, Niedzielko TL, Schneider LE, Mastroeni D, Caselli R, Kukull W, Morris JC, Hulette CM, Schmechel D, Rogers J, Stephan DA, 2008. Alzheimer’s disease is associated with reduced expression of energy metabolism genes in posterior cingulate neurons. Proc Natl Acad Sci U S A 105, 4441–4446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liao PC, Bergamini C, Fato R, Pon LA, Pallotti F, 2020. Isolation of mitochondria from cells and tissues. Methods Cell Biol 155, 3–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liguori M, Nuzziello N, Introna A, Consiglio A, Licciulli F, D’Errico E, Scarafino A, Distaso E, Simone IL, 2018. Dysregulation of MicroRNAs and Target Genes Networks in Peripheral Blood of Patients With Sporadic Amyotrophic Lateral Sclerosis. Front Mol Neurosci 11, 288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin Q, Mao Y, Song Y, Huang D, 2015. MicroRNA34a induces apoptosis in PC12 cells by reducing Bcell lymphoma 2 and sirtuin1 expression. Mol Med Rep 12, 5709–5714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu CC, Liu CC, Kanekiyo T, Xu H, Bu G, 2013. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat Rev Neurol 9, 106–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu CG, Song J, Zhang YQ, Wang PC, 2014. MicroRNA-193b is a regulator of amyloid precursor protein in the blood and cerebrospinal fluid derived exosomal microRNA-193b is a biomarker of Alzheimer’s disease. Mol Med Rep 10, 2395–2400. [DOI] [PubMed] [Google Scholar]
- Liu X, Wang X, Zhang L, Zhou Y, Yang L, Yang M, 2021. By targeting apoptosis facilitator BCL2L13, microRNA miR-484 alleviates cerebral ischemia/reperfusion injury-induced neuronal apoptosis in mice. Bioengineered 12, 948–959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lovell MA, Ehmann WD, Mattson MP, Markesbery WR, 1997. Elevated 4-hydroxynonenal in ventricular fluid in Alzheimer’s disease. Neurobiol Aging 18, 457–461. [DOI] [PubMed] [Google Scholar]
- Lukiw WJ, 2020. microRNA-146a signaling in Alzheimer’s disease (AD) and prion disease (PrD). Front Neurol 11, 462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lungu G, Stoica G, Ambrus A, 2013. MicroRNA profiling and the role of microRNA-132 in neurodegeneration using a rat model. Neurosci Lett 553, 153–158. [DOI] [PubMed] [Google Scholar]
- Lynch MA, 2004. Long-term potentiation and memory. Physiol Rev 84, 87–136. [DOI] [PubMed] [Google Scholar]
- Ma X, Liu L, Meng J, 2017. MicroRNA-125b promotes neurons cell apoptosis and Tau phosphorylation in Alzheimer’s disease. Neurosci Lett 661, 57–62. [DOI] [PubMed] [Google Scholar]
- Ma Y, Wang L, Jia R, 2020. The role of mitochondrial dynamics in human cancers. Am J Cancer Res 10, 1278–1293. [PMC free article] [PubMed] [Google Scholar]
- Macgregor-Das AM, Das S, 2018. A microRNA’s journey to the center of the mitochondria. Am J Physiol Heart Circ Physiol 315, H206–H215. [DOI] [PubMed] [Google Scholar]
- Majstrowicz K, Honnert U, Nikolaus P, Schwarz V, Oeding SJ, Hemkemeyer SA, Bahler M, 2021. Coordination of mitochondrial and cellular dynamics by the actin-based motor Myo19. J Cell Sci 134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Manczak M, Calkins MJ, Reddy PH, 2011. Impaired mitochondrial dynamics and abnormal interaction of amyloid beta with mitochondrial protein Drp1 in neurons from patients with Alzheimer’s disease: implications for neuronal damage. Hum Mol Genet 20, 2495–2509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Manczak M, Reddy PH, 2012. Abnormal interaction between the mitochondrial fission protein Drp1 and hyperphosphorylated tau in Alzheimer’s disease neurons: implications for mitochondrial dysfunction and neuronal damage. Hum Mol Genet 21, 2538–2547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mandal A, Drerup CM, 2019. Axonal Transport and Mitochondrial Function in Neurons. Front Cell Neurosci 13, 373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mannella CA, 2020. Consequences of Folding the Mitochondrial Inner Membrane. Front Physiol 11, 536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mao P, Manczak M, Calkins MJ, Truong Q, Reddy TP, Reddy AP, Shirendeb U, Lo HH, Rabinovitch PS, Reddy PH, 2012. Mitochondria-targeted catalase reduces abnormal APP processing, amyloid beta production and BACE1 in a mouse model of Alzheimer’s disease: implications for neuroprotection and lifespan extension. Hum Mol Genet 21, 2973–2990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mao P, Reddy PH, 2011. Aging and amyloid beta-induced oxidative DNA damage and mitochondrial dysfunction in Alzheimer’s disease: implications for early intervention and therapeutics. Biochim Biophys Acta 1812, 1359–1370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marrocco I, Altieri F, Peluso I, 2017. Measurement and Clinical Significance of Biomarkers of Oxidative Stress in Humans. Oxid Med Cell Longev 2017, 6501046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marsh J, Alifragis P, 2018. Synaptic dysfunction in Alzheimer’s disease: the effects of amyloid beta on synaptic vesicle dynamics as a novel target for therapeutic intervention. Neural Regen Res 13, 616–623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marti E, Pantano L, Banez-Coronel M, Llorens F, Minones-Moyano E, Porta S, Sumoy L, Ferrer I, Estivill X, 2010. A myriad of miRNA variants in control and Huntington’s disease brain regions detected by massively parallel sequencing. Nucleic Acids Res 38, 7219–7235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martinez-Reyes I, Chandel NS, 2020. Mitochondrial TCA cycle metabolites control physiology and disease. Nat Commun 11, 102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mauch DH, Nagler K, Schumacher S, Goritz C, Muller EC, Otto A, Pfrieger FW, 2001. CNS synaptogenesis promoted by glia-derived cholesterol. Science 294, 1354–1357. [DOI] [PubMed] [Google Scholar]
- Maurel M, Chevet E, 2013. Endoplasmic reticulum stress signaling: the microRNA connection. Am J Physiol Cell Physiol 304, C1117–1126. [DOI] [PubMed] [Google Scholar]
- McCarthy M, Raval AP, 2020. The peri-menopause in a woman’s life: a systemic inflammatory phase that enables later neurodegenerative disease. J Neuroinflammation 17, 317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McInnes J, Wierda K, Snellinx A, Bounti L, Wang YC, Stancu IC, Apostolo N, Gevaert K, Dewachter I, Spires-Jones TL, De Strooper B, De Wit J, Zhou L, Verstreken P, 2018. Synaptogyrin-3 Mediates Presynaptic Dysfunction Induced by Tau. Neuron 97, 823–835 e828. [DOI] [PubMed] [Google Scholar]
- McKenzie M, Lim SC, Duchen MR, 2017. Simultaneous Measurement of Mitochondrial Calcium and Mitochondrial Membrane Potential in Live Cells by Fluorescent Microscopy. J Vis Exp. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McWilliams TG, Suomalainen A, 2019. Mitochondrial DNA can be inherited from fathers, not just mothers. Nature 565, 296–297. [DOI] [PubMed] [Google Scholar]
- Melcangi RC, Giatti S, Garcia-Segura LM, 2016. Levels and actions of neuroactive steroids in the nervous system under physiological and pathological conditions: Sex-specific features. Neurosci Biobehav Rev 67, 25–40. [DOI] [PubMed] [Google Scholar]
- Millar JK, Wilson-Annan JC, Anderson S, Christie S, Taylor MS, Semple CA, Devon RS, St Clair DM, Muir WJ, Blackwood DH, Porteous DJ, 2000. Disruption of two novel genes by a translocation co-segregating with schizophrenia. Hum Mol Genet 9, 1415–1423. [DOI] [PubMed] [Google Scholar]
- Misgeld T, Schwarz TL, 2017. Mitostasis in Neurons: Maintaining Mitochondria in an Extended Cellular Architecture. Neuron 96, 651–666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mishra P, Chan DC, 2014. Mitochondrial dynamics and inheritance during cell division, development and disease. Nat Rev Mol Cell Biol 15, 634–646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Misko A, Jiang S, Wegorzewska I, Milbrandt J, Baloh RH, 2010. Mitofusin 2 is necessary for transport of axonal mitochondria and interacts with the Miro/Milton complex. J Neurosci 30, 4232–4240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Misrani A, Tabassum S, Yang L, 2021. Mitochondrial Dysfunction and Oxidative Stress in Alzheimer’s Disease. Front Aging Neurosci 13, 617588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mochida S, 2021. Neurotransmitter Release Site Replenishment and Presynaptic Plasticity. Int J Mol Sci 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Modesti L, Danese A, Angela Maria Vitto V, Ramaccini D, Aguiari G, Gafa R, Lanza G, Giorgi C, Pinton P, 2021. Mitochondrial Ca(2+) Signaling in Health, Disease and Therapy. Cells 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moldogazieva NT, Mokhosoev IM, Mel’nikova TI, Porozov YB, Terentiev AA, 2019. Oxidative Stress and Advanced Lipoxidation and Glycation End Products (ALEs and AGEs) in Aging and Age-Related Diseases. Oxid Med Cell Longev 2019, 3085756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morsci NS, Hall DH, Driscoll M, Sheng ZH, 2016. Age-Related Phasic Patterns of Mitochondrial Maintenance in Adult Caenorhabditis elegans Neurons. J Neurosci 36, 1373–1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morton H, Kshirsagar S, Orlov E, Bunquin LE, Sawant N, Boleng L, George M, Basu T, Ramasubramanian B, Pradeepkiran JA, Kumar S, Vijayan M, Reddy AP, Reddy PH, 2021. Defective mitophagy and synaptic degeneration in Alzheimer’s disease: Focus on aging, mitochondria and synapse. Free Radic Biol Med 172, 652–667. [DOI] [PubMed] [Google Scholar]
- Mott JL, Kobayashi S, Bronk SF, Gores GJ, 2007. mir-29 regulates Mcl-1 protein expression and apoptosis. Oncogene 26, 6133–6140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Naga KK, Sullivan PG, Geddes JW, 2007. High cyclophilin D content of synaptic mitochondria results in increased vulnerability to permeability transition. J Neurosci 27, 7469–7475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Naithani D, Karn SK, 2020. The Role of Ketone Bodies in Improving Neurological Function and Efficiency. Health Scope 9, e86578. [Google Scholar]
- Nanou E, Catterall WA, 2018. Calcium Channels, Synaptic Plasticity, and Neuropsychiatric Disease. Neuron 98, 466–481. [DOI] [PubMed] [Google Scholar]
- Nascimento A, Lannigan J, Kashatus D, 2017. Detection and Quantification of Mitochondrial Fusion Using Imaging Flow Cytometry. Curr Protoc Cytom 81, 9 53 51–59 53 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nicoll RA, 2017. A Brief History of Long-Term Potentiation. Neuron 93, 281–290. [DOI] [PubMed] [Google Scholar]
- Niescier RF, Kwak SK, Joo SH, Chang KT, Min KT, 2016. Dynamics of Mitochondrial Transport in Axons. Front Cell Neurosci 10, 123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niewiadomska G, Niewiadomski W, Steczkowska M, Gasiorowska A, 2021. Tau Oligomers Neurotoxicity. Life (Basel) 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Norat P, Soldozy S, Sokolowski JD, Gorick CM, Kumar JS, Chae Y, Yagmurlu K, Prada F, Walker M, Levitt MR, Price RJ, Tvrdik P, Kalani MYS, 2020. Mitochondrial dysfunction in neurological disorders: Exploring mitochondrial transplantation. NPJ Regen Med 5, 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Brien J, Hayder H, Zayed Y, Peng C, 2018. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front Endocrinol (Lausanne) 9, 402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oettel M, Mukhopadhyay AK, 2004. Progesterone: the forgotten hormone in men? Aging Male 7, 236–257. [DOI] [PubMed] [Google Scholar]
- Oliveira SR, Dionisio PA, Gaspar MM, Correia Guedes L, Coelho M, Rosa MM, Ferreira JJ, Amaral JD, Rodrigues CMP, 2021. miR-335 Targets LRRK2 and Mitigates Inflammation in Parkinson’s Disease. Front Cell Dev Biol 9, 661461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oliver D, Reddy PH, 2019. Dynamics of Dynamin-Related Protein 1 in Alzheimer’s Disease and Other Neurodegenerative Diseases. Cells 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Orom UA, Nielsen FC, Lund AH, 2008. MicroRNA-10a binds the 5’UTR of ribosomal protein mRNAs and enhances their translation. Mol Cell 30, 460–471. [DOI] [PubMed] [Google Scholar]
- Ortega MA, Fraile-Martinez O, Guijarro LG, Casanova C, Coca S, Alvarez-Mon M, Bujan J, Garcia-Honduvilla N, Asunsolo A, 2020. The Regulatory Role of Mitochondrial MicroRNAs (MitomiRs) in Breast Cancer: Translational Implications Present and Future. Cancers (Basel) 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pagliuso A, Cossart P, Stavru F, 2018. The ever-growing complexity of the mitochondrial fission machinery. Cell Mol Life Sci 75, 355–374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, Giustetto M, Ferreira TA, Guiducci E, Dumas L, Ragozzino D, Gross CT, 2011. Synaptic pruning by microglia is necessary for normal brain development. Science 333, 1456–1458. [DOI] [PubMed] [Google Scholar]
- Parisi C, Napoli G, Amadio S, Spalloni A, Apolloni S, Longone P, Volonte C, 2016. MicroRNA-125b regulates microglia activation and motor neuron death in ALS. Cell Death Differ 23, 531–541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park JC, Han SH, Mook-Jung I, 2020. Peripheral inflammatory biomarkers in Alzheimer’s disease: a brief review. BMB Rep 53, 10–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pathak D, Shields LY, Mendelsohn BA, Haddad D, Lin W, Gerencser AA, Kim H, Brand MD, Edwards RH, Nakamura K, 2015. The role of mitochondrially derived ATP in synaptic vesicle recycling. J Biol Chem 290, 22325–22336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pedriali G, Rimessi A, Sbano L, Giorgi C, Wieckowski MR, Previati M, Pinton P, 2017. Regulation of Endoplasmic Reticulum-Mitochondria Ca(2+) Transfer and Its Importance for Anti-Cancer Therapies. Front Oncol 7, 180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pei L, Wallace DC, 2018. Mitochondrial Etiology of Neuropsychiatric Disorders. Biol Psychiatry 83, 722–730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pena-Bautista C, Tirle T, Lopez-Nogueroles M, Vento M, Baquero M, Chafer-Pericas C, 2019. Oxidative Damage of DNA as Early Marker of Alzheimer’s Disease. Int J Mol Sci 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perez MJ, Jara C, Quintanilla RA, 2018. Contribution of Tau Pathology to Mitochondrial Impairment in Neurodegeneration. Front Neurosci 12, 441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perry AJ, Rimmer KA, Mertens HD, Waller RF, Mulhern TD, Lithgow T, Gooley PR, 2008. Structure, topology and function of the translocase of the outer membrane of mitochondria. Plant Physiol Biochem 46, 265–274. [DOI] [PubMed] [Google Scholar]
- Pilling AD, Horiuchi D, Lively CM, Saxton WM, 2006. Kinesin-1 and Dynein are the primary motors for fast transport of mitochondria in Drosophila motor axons. Mol Biol Cell 17, 2057–2068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Planche V, Bouteloup V, Mangin JF, Dubois B, Delrieu J, Pasquier F, Blanc F, Paquet C, Hanon O, Gabelle A, Ceccaldi M, Annweiler C, Krolak-Salmon P, Habert MO, Fischer C, Chupin M, Bejot Y, Godefroy O, Wallon D, Sauvee M, Bourdel-Marchasson I, Jalenques I, Tison F, Chene G, Dufouil C, the M.S.g., 2021. Clinical relevance of brain atrophy subtypes categorization in memory clinics. Alzheimers Dement 17, 641–652. [DOI] [PubMed] [Google Scholar]
- Pradeepkiran JA, Reddy PH, 2020. Defective mitophagy in Alzheimer’s disease. Ageing Res Rev 64, 101191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quinlan CL, Orr AL, Perevoshchikova IV, Treberg JR, Ackrell BA, Brand MD, 2012. Mitochondrial complex II can generate reactive oxygen species at high rates in both the forward and reverse reactions. J Biol Chem 287, 27255–27264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Radzvilavicius AL, Lane N, Pomiankowski A, 2017. Sexual conflict explains the extraordinary diversity of mechanisms regulating mitochondrial inheritance. BMC Biol 15, 94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rajendran L, Paolicelli RC, 2018. Microglia-Mediated Synapse Loss in Alzheimer’s Disease. J Neurosci 38, 2911–2919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rajmohan R, Reddy PH, 2017. Amyloid-Beta and Phosphorylated Tau Accumulations Cause Abnormalities at Synapses of Alzheimer’s disease Neurons. J Alzheimers Dis 57, 975–999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rak M, Benit P, Chretien D, Bouchereau J, Schiff M, El-Khoury R, Tzagoloff A, Rustin P, 2016. Mitochondrial cytochrome c oxidase deficiency. Clin Sci (Lond) 130, 393–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rangaraju V, Lewis TL Jr., Hirabayashi Y, Bergami M, Motori E, Cartoni R, Kwon SK, Courchet J, 2019. Pleiotropic Mitochondria: The Influence of Mitochondria on Neuronal Development and Disease. J Neurosci 39, 8200–8208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reddy PH, 2006. Mitochondrial oxidative damage in aging and Alzheimer’s disease: implications for mitochondrially targeted antioxidant therapeutics. J Biomed Biotechnol 2006, 31372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reddy PH, 2007. Mitochondrial dysfunction in aging and Alzheimer’s disease: strategies to protect neurons. Antioxid Redox Signal 9, 1647–1658. [DOI] [PubMed] [Google Scholar]
- Reddy PH, 2009a. Amyloid beta, mitochondrial structural and functional dynamics in Alzheimer’s disease. Exp Neurol 218, 286–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reddy PH, 2009b. Role of mitochondria in neurodegenerative diseases: mitochondria as a therapeutic target in Alzheimer’s disease. CNS Spectr 14, 8–13; discussion 16–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reddy PH, 2013a. Amyloid beta-induced glycogen synthase kinase 3beta phosphorylated VDAC1 in Alzheimer’s disease: implications for synaptic dysfunction and neuronal damage. Biochim Biophys Acta 1832, 1913–1921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reddy PH, 2013b. Is the mitochondrial outermembrane protein VDAC1 therapeutic target for Alzheimer’s disease? Biochim Biophys Acta 1832, 67–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reddy PH, Beal MF, 2005. Are mitochondria critical in the pathogenesis of Alzheimer’s disease? Brain Res Brain Res Rev 49, 618–632. [DOI] [PubMed] [Google Scholar]
- Reddy PH, Beal MF, 2008. Amyloid beta, mitochondrial dysfunction and synaptic damage: implications for cognitive decline in aging and Alzheimer’s disease. Trends Mol Med 14, 45–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reddy PH, Oliver DM, 2019. Amyloid Beta and Phosphorylated Tau-Induced Defective Autophagy and Mitophagy in Alzheimer’s Disease. Cells 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reddy PH, Reddy TP, Manczak M, Calkins MJ, Shirendeb U, Mao P, 2011. Dynamin-related protein 1 and mitochondrial fragmentation in neurodegenerative diseases. Brain Res Rev 67, 103–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reddy PH, Tonk S, Kumar S, Vijayan M, Kandimalla R, Kuruva CS, Reddy AP, 2017. A critical evaluation of neuroprotective and neurodegenerative MicroRNAs in Alzheimer’s disease. Biochem Biophys Res Commun 483, 1156–1165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reddy PH, Tripathi R, Troung Q, Tirumala K, Reddy TP, Anekonda V, Shirendeb UP, Calkins MJ, Reddy AP, Mao P, Manczak M, 2012. Abnormal mitochondrial dynamics and synaptic degeneration as early events in Alzheimer’s disease: implications to mitochondria-targeted antioxidant therapeutics. Biochim Biophys Acta 1822, 639–649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Remenyi J, van den Bosch MW, Palygin O, Mistry RB, McKenzie C, Macdonald A, Hutvagner G, Arthur JS, Frenguelli BG, Pankratov Y, 2013. miR-132/212 knockout mice reveal roles for these miRNAs in regulating cortical synaptic transmission and plasticity. PLoS One 8, e62509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rizzuti M, Filosa G, Melzi V, Calandriello L, Dioni L, Bollati V, Bresolin N, Comi GP, Barabino S, Nizzardo M, Corti S, 2018. MicroRNA expression analysis identifies a subset of downregulated miRNAs in ALS motor neuron progenitors. Sci Rep 8, 10105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roger AJ, Munoz-Gomez SA, Kamikawa R, 2017. The Origin and Diversification of Mitochondria. Curr Biol 27, R1177–R1192. [DOI] [PubMed] [Google Scholar]
- Rossi A, Pizzo P, Filadi R, 2019. Calcium, mitochondria and cell metabolism: A functional triangle in bioenergetics. Biochim Biophys Acta Mol Cell Res 1866, 1068–1078. [DOI] [PubMed] [Google Scholar]
- Rossi MJ, Pekkurnaz G, 2019. Powerhouse of the mind: mitochondrial plasticity at the synapse. Curr Opin Neurobiol 57, 149–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sadigh-Eteghad S, Sabermarouf B, Majdi A, Talebi M, Farhoudi M, Mahmoudi J, 2015. Amyloid-beta: a crucial factor in Alzheimer’s disease. Med Princ Pract 24, 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Safieh M, Korczyn AD, Michaelson DM, 2019. ApoE4: an emerging therapeutic target for Alzheimer’s disease. BMC Med 17, 64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sakai J, 2020. Core Concept: How synaptic pruning shapes neural wiring during development and, possibly, in disease. Proc Natl Acad Sci U S A 117, 16096–16099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanchez-Ruderisch H, Queiros AM, Fliegner D, Eschen C, Kararigas G, Regitz-Zagrosek V, 2019. Sex-specific regulation of cardiac microRNAs targeting mitochondrial proteins in pressure overload. Biol Sex Differ 10, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saotome M, Safiulina D, Szabadkai G, Das S, Fransson A, Aspenstrom P, Rizzuto R, Hajnoczky G, 2008. Bidirectional Ca2+-dependent control of mitochondrial dynamics by the Miro GTPase. Proc Natl Acad Sci U S A 105, 20728–20733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sarkar S, Jun S, Rellick S, Quintana DD, Cavendish JZ, Simpkins JW, 2016. Expression of microRNA-34a in Alzheimer’s disease brain targets genes linked to synaptic plasticity, energy metabolism, and resting state network activity. Brain Res 1646, 139–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Satoh J, Kino Y, Niida S, 2015. MicroRNA-Seq Data Analysis Pipeline to Identify Blood Biomarkers for Alzheimer’s Disease from Public Data. Biomark Insights 10, 21–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shahpasand K, Uemura I, Saito T, Asano T, Hata K, Shibata K, Toyoshima Y, Hasegawa M, Hisanaga S, 2012. Regulation of mitochondrial transport and inter-microtubule spacing by tau phosphorylation at the sites hyperphosphorylated in Alzheimer’s disease. J Neurosci 32, 2430–2441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shankar GM, Walsh DM, 2009. Alzheimer’s disease: synaptic dysfunction and Abeta. Mol Neurodegener 4, 48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sharma C, Kim S, Nam Y, Jung UJ, Kim SR, 2021. Mitochondrial Dysfunction as a Driver of Cognitive Impairment in Alzheimer’s Disease. Int J Mol Sci 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sheladia S, Reddy PH, 2021. Age-Related Chronic Diseases and Alzheimer’s Disease in Texas: A Hispanic Focused Study. J Alzheimers Dis Rep 5, 121–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sheng ZH, 2014. Mitochondrial trafficking and anchoring in neurons: New insight and implications. J Cell Biol 204, 1087–1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sheng ZH, 2017. The Interplay of Axonal Energy Homeostasis and Mitochondrial Trafficking and Anchoring. Trends Cell Biol 27, 403–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi G, Liu Y, Liu T, Yan W, Liu X, Wang Y, Shi J, Jia L, 2012. Upregulated miR-29b promotes neuronal cell death by inhibiting Bcl2L2 after ischemic brain injury. Exp Brain Res 216, 225–230. [DOI] [PubMed] [Google Scholar]
- Shi Q, Gibson GE, 2011. Up-regulation of the mitochondrial malate dehydrogenase by oxidative stress is mediated by miR-743a. J Neurochem 118, 440–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shioya M, Obayashi S, Tabunoki H, Arima K, Saito Y, Ishida T, Satoh J, 2010. Aberrant microRNA expression in the brains of neurodegenerative diseases: miR-29a decreased in Alzheimer disease brains targets neurone navigator 3. Neuropathol Appl Neurobiol 36, 320–330. [DOI] [PubMed] [Google Scholar]
- Shoshan-Barmatz V, De S, 2017. Mitochondrial VDAC, the Na(+)/Ca(2+) Exchanger, and the Ca(2+) Uniporter in Ca(2+) Dynamics and Signaling. Adv Exp Med Biol 981, 323–347. [DOI] [PubMed] [Google Scholar]
- Sies H, Jones DP, 2020. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol 21, 363–383. [DOI] [PubMed] [Google Scholar]
- Silver I, Erecinska M, 1998. Oxygen and ion concentrations in normoxic and hypoxic brain cells. Adv Exp Med Biol 454, 7–16. [DOI] [PubMed] [Google Scholar]
- Silvestro S, Bramanti P, Mazzon E, 2019. Role of miRNAs in Alzheimer’s Disease and Possible Fields of Application. Int J Mol Sci 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simion V, Henriet E, Juric V, Aquino R, Loussouarn C, Laurent Y, Martin F, Midoux P, Garcion E, Pichon C, Baril P, 2020. Intracellular trafficking and functional monitoring of miRNA delivery in glioblastoma using lipopolyplexes and the miRNA-ON RILES reporter system. J Control Release 327, 429–443. [DOI] [PubMed] [Google Scholar]
- Smith-Vikos T, Slack FJ, 2012. MicroRNAs and their roles in aging. J Cell Sci 125, 7–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song W, Bossy B, Martin OJ, Hicks A, Lubitz S, Knott AB, Bossy-Wetzel E, 2008. Assessing mitochondrial morphology and dynamics using fluorescence wide-field microscopy and 3D image processing. Methods 46, 295–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spinelli JB, Haigis MC, 2018. The multifaceted contributions of mitochondria to cellular metabolism. Nat Cell Biol 20, 745–754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Srikanth V, Maczurek A, Phan T, Steele M, Westcott B, Juskiw D, Munch G, 2011. Advanced glycation endproducts and their receptor RAGE in Alzheimer’s disease. Neurobiol Aging 32, 763–777. [DOI] [PubMed] [Google Scholar]
- Stanga S, Caretto A, Boido M, Vercelli A, 2020. Mitochondrial Dysfunctions: A Red Thread across Neurodegenerative Diseases. Int J Mol Sci 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Starega-Roslan J, Koscianska E, Kozlowski P, Krzyzosiak WJ, 2011. The role of the precursor structure in the biogenesis of microRNA. Cell Mol Life Sci 68, 2859–2871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Starkov AA, Fiskum G, Chinopoulos C, Lorenzo BJ, Browne SE, Patel MS, Beal MF, 2004. Mitochondrial alpha-ketoglutarate dehydrogenase complex generates reactive oxygen species. J Neurosci 24, 7779–7788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stefanatos R, Sanz A, 2018. The role of mitochondrial ROS in the aging brain. FEBS Lett 592, 743–758. [DOI] [PubMed] [Google Scholar]
- Strickland M, Yacoubi-Loueslati B, Bouhaouala-Zahar B, Pender SLF, Larbi A, 2019. Relationships Between Ion Channels, Mitochondrial Functions and Inflammation in Human Aging. Front Physiol 10, 158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strittmatter WJ, Roses AD, 1996. Apolipoprotein E and Alzheimer’s disease. Annu Rev Neurosci 19, 53–77. [DOI] [PubMed] [Google Scholar]
- Sudhof TC, 2012. Calcium control of neurotransmitter release. Cold Spring Harb Perspect Biol 4, a011353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Supnet C, Bezprozvanny I, 2010. Neuronal calcium signaling, mitochondrial dysfunction, and Alzheimer’s disease. J Alzheimers Dis 20 Suppl 2, S487–498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swerdlow NS, Wilkins HM, 2020. Mitophagy and the Brain. Int J Mol Sci 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swerdlow RH, 2012. Mitochondria and cell bioenergetics: increasingly recognized components and a possible etiologic cause of Alzheimer’s disease. Antioxid Redox Signal 16, 1434–1455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swerdlow RH, 2020. The mitochondrial hypothesis: Dysfunction, bioenergetic defects, and the metabolic link to Alzheimer’s disease. Int Rev Neurobiol 154, 207–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taanman JW, 1999. The mitochondrial genome: structure, transcription, translation and replication. Biochim Biophys Acta 1410, 103–123. [DOI] [PubMed] [Google Scholar]
- Taguchi YH, Wang H, 2018. Exploring MicroRNA Biomarkers for Parkinson’s Disease from mRNA Expression Profiles. Cells 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tait SW, Green DR, 2012. Mitochondria and cell signalling. J Cell Sci 125, 807–815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Talepoor Ardakani M, Rostamian Delavar M, Baghi M, Nasr-Esfahani MH, Kiani-Esfahani A, Ghaedi K, 2019. Upregulation of miR-200a and miR-204 in MPP(+) -treated differentiated PC12 cells as a model of Parkinson’s disease. Mol Genet Genomic Med 7, e548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang BL, 2016. Sirt1 and the Mitochondria. Mol Cells 39, 87–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tauffenberger A, Magistretti PJ, 2021. Reactive Oxygen Species: Beyond Their Reactive Behavior. Neurochem Res 46, 77–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tillement L, Lecanu L, Papadopoulos V, 2011. Alzheimer’s disease: effects of beta-amyloid on mitochondria. Mitochondrion 11, 13–21. [DOI] [PubMed] [Google Scholar]
- Titze-de-Almeida SS, Soto-Sanchez C, Fernandez E, Koprich JB, Brotchie JM, Titze-de-Almeida R, 2020. The Promise and Challenges of Developing miRNA-Based Therapeutics for Parkinson’s Disease. Cells 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Todorova V, Blokland A, 2017. Mitochondria and Synaptic Plasticity in the Mature and Aging Nervous System. Curr Neuropharmacol 15, 166–173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tomlinson C, Macintyre H, Dorrian CA, Ahmed SF, Wallace AM, 2004. Testosterone measurements in early infancy. Arch Dis Child Fetal Neonatal Ed 89, F558–559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Torres-Gonzalez M, Gawlowski T, Kocalis H, Scott BT, Dillmann WH, 2014. Mitochondrial 8-oxoguanine glycosylase decreases mitochondrial fragmentation and improves mitochondrial function in H9C2 cells under oxidative stress conditions. Am J Physiol Cell Physiol 306, C221–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trabucchi M, Mategot R, 2020. Subcellular Heterogeneity of the microRNA Machinery. Trends Genet 36, 70. [DOI] [PubMed] [Google Scholar]
- Treiber T, Treiber N, Meister G, 2019. Regulation of microRNA biogenesis and its crosstalk with other cellular pathways. Nat Rev Mol Cell Biol 20, 5–20. [DOI] [PubMed] [Google Scholar]
- Truesdell SS, Mortensen RD, Seo M, Schroeder JC, Lee JH, LeTonqueze O, Vasudevan S, 2012. MicroRNA-mediated mRNA translation activation in quiescent cells and oocytes involves recruitment of a nuclear microRNP. Sci Rep 2, 842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tu S, Okamoto S, Lipton SA, Xu H, 2014. Oligomeric Abeta-induced synaptic dysfunction in Alzheimer’s disease. Mol Neurodegener 9, 48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turnbull HE, Lax NZ, Diodato D, Ansorge O, Turnbull DM, 2010. The mitochondrial brain: From mitochondrial genome to neurodegeneration. Biochim Biophys Acta 1802, 111–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tyrrell DJ, Blin MG, Song J, Wood SC, Goldstein DR, 2020. Aging Impairs Mitochondrial Function and Mitophagy and Elevates Interleukin 6 Within the Cerebral Vasculature. J Am Heart Assoc 9, e017820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ureshino RP, Erustes AG, Bassani TB, Wachilewski P, Guarache GC, Nascimento AC, Costa AJ, Smaili SS, Pereira G, 2019. The Interplay between Ca(2+) Signaling Pathways and Neurodegeneration. Int J Mol Sci 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van Cauwenberghe C, Van Broeckhoven C, Sleegers K, 2016. The genetic landscape of Alzheimer disease: clinical implications and perspectives. Genet Med 18, 421–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van den Berg A, Mols J, Han J, 2008. RISC-target interaction: cleavage and translational suppression. Biochim Biophys Acta 1779, 668–677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vendramin R, Marine JC, Leucci E, 2017. Non-coding RNAs: the dark side of nuclear-mitochondrial communication. EMBO J 36, 1123–1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verschueren KHG, Blanchet C, Felix J, Dansercoer A, De Vos D, Bloch Y, Van Beeumen J, Svergun D, Gutsche I, Savvides SN, Verstraete K, 2019. Structure of ATP citrate lyase and the origin of citrate synthase in the Krebs cycle. Nature 568, 571–575. [DOI] [PubMed] [Google Scholar]
- Vidoni S, Zanna C, Rugolo M, Sarzi E, Lenaers G, 2013. Why mitochondria must fuse to maintain their genome integrity. Antioxid Redox Signal 19, 379–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vijayan M, Kumar S, Yin X, Zafer D, Chanana V, Cengiz P, Reddy PH, 2018. Identification of novel circulatory microRNA signatures linked to patients with ischemic stroke. Hum Mol Genet 27, 2318–2329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wahid F, Shehzad A, Khan T, Kim YY, 2010. MicroRNAs: synthesis, mechanism, function, and recent clinical trials. Biochim Biophys Acta 1803, 1231–1243. [DOI] [PubMed] [Google Scholar]
- Wallace DC, 2005. A mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: a dawn for evolutionary medicine. Annu Rev Genet 39, 359–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wallace DC, 2010. Mitochondrial DNA mutations in disease and aging. Environ Mol Mutagen 51, 440–450. [DOI] [PubMed] [Google Scholar]
- Wang K, Long B, Jiao JQ, Wang JX, Liu JP, Li Q, Li PF, 2012. miR-484 regulates mitochondrial network through targeting Fis1. Nat Commun 3, 781. [DOI] [PubMed] [Google Scholar]
- Wang Q, Ge X, Zhang J, Chen L, 2020a. Effect of lncRNA WT1-AS regulating WT1 on oxidative stress injury and apoptosis of neurons in Alzheimer’s disease via inhibition of the miR-375/SIX4 axis. Aging (Albany NY) 12, 23974–23995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang W, Zhao F, Ma X, Perry G, Zhu X, 2020b. Mitochondria dysfunction in the pathogenesis of Alzheimer’s disease: recent advances. Mol Neurodegener 15, 30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang X, Schwarz TL, 2009. The mechanism of Ca2+ -dependent regulation of kinesin-mediated mitochondrial motility. Cell 136, 163–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y, Veremeyko T, Wong AH, El Fatimy R, Wei Z, Cai W, Krichevsky AM, 2017. Downregulation of miR-132/212 impairs S-nitrosylation balance and induces tau phosphorylation in Alzheimer’s disease. Neurobiol Aging 51, 156–166. [DOI] [PubMed] [Google Scholar]
- Ward A, Crean S, Mercaldi CJ, Collins JM, Boyd D, Cook MN, Arrighi HM, 2012. Prevalence of apolipoprotein E4 genotype and homozygotes (APOE e4/4) among patients diagnosed with Alzheimer’s disease: a systematic review and meta-analysis. Neuroepidemiology 38, 1–17. [DOI] [PubMed] [Google Scholar]
- Watts ME, Williams SM, Nithianantharajah J, Claudianos C, 2018. Hypoxia-Induced MicroRNA-210 Targets Neurodegenerative Pathways. Noncoding RNA 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wayman GA, Davare M, Ando H, Fortin D, Varlamova O, Cheng HY, Marks D, Obrietan K, Soderling TR, Goodman RH, Impey S, 2008. An activity-regulated microRNA controls dendritic plasticity by down-regulating p250GAP. Proc Natl Acad Sci U S A 105, 9093–9098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei YH, Lee HC, 2002. Oxidative stress, mitochondrial DNA mutation, and impairment of antioxidant enzymes in aging. Exp Biol Med (Maywood) 227, 671–682. [DOI] [PubMed] [Google Scholar]
- Weidling IW, Swerdlow RH, 2020. Mitochondria in Alzheimer’s disease and their potential role in Alzheimer’s proteostasis. Exp Neurol 330, 113321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weinberg RB, Mufson EJ, Counts SE, 2015. Evidence for a neuroprotective microRNA pathway in amnestic mild cognitive impairment. Front Neurosci 9, 430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams JA, Zhao K, Jin S, Ding WX, 2017. New methods for monitoring mitochondrial biogenesis and mitophagy in vitro and in vivo. Exp Biol Med (Maywood) 242, 781–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wingo TS, Yang J, Fan W, Min Canon S, Gerasimov ES, Lori A, Logsdon B, Yao B, Seyfried NT, Lah JJ, Levey AI, Boyle PA, Schneider JA, De Jager PL, Bennett DA, Wingo AP, 2020. Brain microRNAs associated with late-life depressive symptoms are also associated with cognitive trajectory and dementia. NPJ Genom Med 5, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wong HK, Veremeyko T, Patel N, Lemere CA, Walsh DM, Esau C, Vanderburg C, Krichevsky AM, 2013. De-repression of FOXO3a death axis by microRNA-132 and −212 causes neuronal apoptosis in Alzheimer’s disease. Hum Mol Genet 22, 3077–3092. [DOI] [PubMed] [Google Scholar]
- Wong W, 2020. Economic burden of Alzheimer disease and managed care considerations. Am J Manag Care 26, S177–S183. [DOI] [PubMed] [Google Scholar]
- Woo HN, Park S, Kim HL, Jung MK, Pack CG, Park J, Cho Y, Jo DG, Kim DK, Mook-Jung I, Kim SW, Lee H, 2021. miR-351-5p/Miro2 axis contributes to hippocampal neural progenitor cell death via unbalanced mitochondrial fission. Mol Ther Nucleic Acids 23, 643–656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xie S, Niu W, Xu F, Wang Y, Hu S, Niu C, 2020. Differential expression and significance of miRNAs in plasma extracellular vesicles of patients with Parkinson’s disease. Int J Neurosci, 1–16. [DOI] [PubMed] [Google Scholar]
- Xing RX, Li LG, Liu XW, Tian BX, Cheng Y, 2020. Down regulation of miR-218, miR-124, and miR-144 relates to Parkinson’s disease via activating NF-kappaB signaling. Kaohsiung J Med Sci 36, 786–792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiong H, Chen S, Lai L, Yang H, Xu Y, Pang J, Su Z, Lin H, Zheng Y, 2019. Modulation of miR-34a/SIRT1 signaling protects cochlear hair cells against oxidative stress and delays age-related hearing loss through coordinated regulation of mitophagy and mitochondrial biogenesis. Neurobiol Aging 79, 30–42. [DOI] [PubMed] [Google Scholar]
- Yamakuchi M, Ferlito M, Lowenstein CJ, 2008. miR-34a repression of SIRT1 regulates apoptosis. Proc Natl Acad Sci U S A 105, 13421–13426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yan C, Duanmu X, Zeng L, Liu B, Song Z, 2019. Mitochondrial DNA: Distribution, Mutations, and Elimination. Cells 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yan W, Kang Y, Ji X, Li S, Li Y, Zhang G, Cui H, Shi G, 2017. Testosterone Upregulates the Expression of Mitochondrial ND1 and ND4 and Alleviates the Oxidative Damage to the Nigrostriatal Dopaminergic System in Orchiectomized Rats. Oxid Med Cell Longev 2017, 1202459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yetkin-Arik B, Vogels IMC, Nowak-Sliwinska P, Weiss A, Houtkooper RH, Van Noorden CJF, Klaassen I, Schlingemann RO, 2019. The role of glycolysis and mitochondrial respiration in the formation and functioning of endothelial tip cells during angiogenesis. Sci Rep 9, 12608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Youle RJ, van der Bliek AM, 2012. Mitochondrial fission, fusion, and stress. Science 337, 1062–1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang C, Nestorova G, Rissman RA, Feng J, 2013. Detection and quantification of 8-hydroxy-2’-deoxyguanosine in Alzheimer’s transgenic mouse urine using capillary electrophoresis. Electrophoresis 34, 2268–2274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang L, Fang Y, Zhao X, Zheng Y, Ma Y, Li S, Huang Z, Li L, 2021. miR-204 silencing reduces mitochondrial autophagy and ROS production in a murine AD model via the TRPML1-activated STAT3 pathway. Mol Ther Nucleic Acids 24, 822–831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang R, Zhou H, Jiang L, Mao Y, Cui X, Xie B, Cui D, Wang H, Zhang Q, Xu S, 2016. MiR-195 dependent roles of mitofusin2 in the mitochondrial dysfunction of hippocampal neurons in SAMP8 mice. Brain Res 1652, 135–143. [DOI] [PubMed] [Google Scholar]
- Zhang X, Zuo X, Yang B, Li Z, Xue Y, Zhou Y, Huang J, Zhao X, Zhou J, Yan Y, Zhang H, Guo P, Sun H, Guo L, Zhang Y, Fu XD, 2014. MicroRNA directly enhances mitochondrial translation during muscle differentiation. Cell 158, 607–619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao L, Wang Z, 2019. MicroRNAs: Game Changers in the Regulation of alpha-Synuclein in Parkinson’s Disease. Parkinsons Dis 2019, 1743183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao RZ, Jiang S, Zhang L, Yu ZB, 2019. Mitochondrial electron transport chain, ROS generation and uncoupling (Review). Int J Mol Med 44, 3–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao XL, Wang WA, Tan JX, Huang JK, Zhang X, Zhang BZ, Wang YH, Yang Cheng HY, Zhu HL, Sun XJ, Huang FD, 2010. Expression of beta-amyloid induced age-dependent presynaptic and axonal changes in Drosophila. J Neurosci 30, 1512–1522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao Y, Jaber VR, LeBeauf A, Sharfman NM, Lukiw WJ, 2019. microRNA-34a (miRNA-34a) mediated down-regulation of the post-synaptic cytoskeletal element SHANK3 in sporadic Alzheimer’s disease (AD). Front Neurol 10, 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou L, McInnes J, Wierda K, Holt M, Herrmann AG, Jackson RJ, Wang YC, Swerts J, Beyens J, Miskiewicz K, Vilain S, Dewachter I, Moechars D, De Strooper B, Spires-Jones TL, De Wit J, Verstreken P, 2017. Tau association with synaptic vesicles causes presynaptic dysfunction. Nat Commun 8, 15295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zilocchi M, Broderick K, Phanse S, Aly KA, Babu M, 2020. Mitochondria under the spotlight: On the implications of mitochondrial dysfunction and its connectivity to neuropsychiatric disorders. Comput Struct Biotechnol J 18, 2535–2546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zorova LD, Popkov VA, Plotnikov EY, Silachev DN, Pevzner IB, Jankauskas SS, Babenko VA, Zorov SD, Balakireva AV, Juhaszova M, Sollott SJ, Zorov DB, 2018. Mitochondrial membrane potential. Anal Biochem 552, 50–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
