Abstract
Aging is a multifaceted process influenced by many elements. During cell division, the repetitive DNA sequences at the ends of chromosomes called telomeres protect them from degradation. Telomeres shorten alongside each cell division, eventually contributing to cellular senescence and aging. Telomerase as an enzyme has a role in the maintenance of telomere length. Reduced function of telomerase is linked to acceleration of aging and age-related diseases. By affecting cellular function, mutations in particular genes can cause aging. Genes involved in DNA repair, cellular metabolism, and inflammation play the key roles in this process. Accumulated mutations result in cellular dysfunction and age-related diseases over time. Epigenetic changes are the modifications that impact gene expression without altering the DNA sequence. Lifestyle factors (diet, exercise, stress) and environmental influences (toxins, trauma) can cause epigenetic alterations. DNA methylation as well as histone modifications are examples of epigenetic alterations. They influence how cells work and are essential to the aging process. Understanding these molecular mechanisms is essential for developing interventions to promote healthy aging and prevent age-related diseases. This paper explores the potential of CRISPR/Cas9 as a gene-editing tool to target these mechanisms and mitigate age-related conditions, ultimately enhancing longevity and quality of life.
Keywords: Molecular and cellular mechanism, Aging, Gene editing, CRISPR-Cas9
Introduction
Aging refers to the natural biological process characterized by the gradual decline in physiological functions and heightened vulnerability to age-related ailments. Ample research has focused on the molecular and cellular mechanisms associated with aging, uncovering the involved factors like telomere shortening, reduced telomerase activity, gene mutations, and epigenetic alterations [1]. Telomeres act as the protective caps at the ends of chromosomes, and when they shorten with each cell division, cellular senescence and aging occur. Telomerase is an enzyme that aids in maintaining the telomere length, and its diminished function has been associated with accelerated aging and age-related illnesses [2–4].
Mutations in genes involved in DNA repair, cellular metabolism, and inflammation can also contribute to the aging process via impairing cellular function and promoting the accumulation of damage over time. Furthermore, the aging process can be affected by epigenetic changes like alterations in DNA methylation and histone modifications through changing the gene expression patterns and cellular function [5]. Understanding how aging works at the molecular and cellular levels, such as telomere shortening, decreased telomerase function, gene mutations, and epigenetic changes, is important for creating ways to support healthy aging and avoid diseases related to aging. The present paper strives to unravel the complex interplay of these factors and identify potential targets for therapeutic interventions to enhance longevity and quality of life in aging populations [5]. Today, clustered regularly interspaced short palindromic repeats/CRISPR-associated protein-9 (CRISPR/Cas9) seems to be a powerful gene-editing tool that has garnered significant attention in scientific investigation. The current study tries to explore how telomerase can be leveraged to mitigate age-related diseases.
CRISPR/Cas9 and aging
Scientists have made significant progress in comprehending the molecular mechanisms of aging by CRISPR/Cas9 application, which has changed genetics and molecular biology [6]. The process of aging involves the deterioration of cellular functions, leading to health issues such as heart disease, cancer, and Alzheimer’s disease. In one notable study, mice suffered from Hutchinson–Gilford progeria syndrome, a rare genetic disorder that accelerates aging. The mice exhibited signs such as DNA damage, cardiac dysfunction, and a dramatically shortened lifespan [7, 8]. The researchers developed a novel CRISPR/Cas9 genome-editing therapy to conceal accelerated aging in these mice. Specifically, they targeted the LMNA gene, which produces two similar proteins: lamin A and lamin C. Progeria shifts the production toward a toxic form called progerin. Using CRISPR/Cas9, researchers were able to reduce the harmful effects and better understand the aging process by focusing on both lamin A and progerin [8]. Another study on aging used genome-wide CRISPR/Cas9 screening to identify several new senescence-promoting genes in humans [9]. Of note, cellular senescence, characterized by permanent growth arrest, plays a critical role in aging. [10]. It should be mentioned that this discovery opened up fresh therapeutic avenues for addressing aging-related pathologies (Fig. 1) [9].
Fig. 1.
The Evolution of Discoveries in Aging Biology and Epigenetics. From the earliest discoveries on cell division limits and the role of telomerase to cutting-edge technologies like deep learning and epigenome restoration, this image provides a comprehensive overview of key advancements in understanding the aging process. Looking ahead, the coming years could bring a revolution in biology and therapies related to longevity
CRISPR-Cas9: overview, mechanism, and significance
CRISPR-Cas9 is a revolutionary gene-editing tool that allows scientists to accurately change the DNA of living things, opening up many possibilities for treating genetic diseases, enhancing crop yields, and possibly changing the genes of an embryo [11]. CRISPR-Cas9 has several benefits compared to older gene-editing methods like TALENs and ZFNs, mainly because it is easier to use, more flexible, and more accurate. It is due to its simplicity, adaptability, and enhanced precision [12].
Mechanism
The CRISPR-Cas9 method is derived from a naturally occurring defense mechanism employed by bacteria to detect and remove viral DNA infiltrating their cells [11]. The mechanism of CRISPR-Cas9 genome editing involves several factors and steps (Fig. 2), including 1. Guide RNA (gRNA) synthesis: The initial step involves creating an RNA molecule with a short ‘guide’ sequence that binds to a specific DNA sequence in a genome. The RNA molecule also creates a binding connection with the Cas9 enzyme [13]. 2. Complex formation: The gRNA combines with the Cas9 enzyme to create a complex and facilitates the targeted binding of the Cas9 enzyme to the specific genomic locus that corresponds to its nucleotide sequence [14]. 3. DNA cleavage: Upon binding to the target DNA sequence, the Cas9-sgRNA complex starts a cleavage event by the Cas9 nuclease, leading to the simultaneous cutting of both strands of the DNA at a precise location within the target sequence. This event leads to the creation of a double-strand break (DSB) in the DNA molecule.[15] 4. DNA repair: The Cas9 nuclease repairs the double-stranded break (DSB), which is carried out by the cellular machinery of the host organism. The two primary repair mechanisms consist of non-homologous end joining (NHEJ) and homology-directed repair (HDR). Non-homologous end joining (NHEJ) is the most common and effective cellular method for DNA damage repair. This mechanism shows increased activity in cells; nonetheless, it is characterized by a tendency to introduce errors. Explicitly, NHEJ may result in small, stochastic insertions or deletions (indels) at the site of DNA cleavage. As a result, such changes in DNA can contribute to the occurrence of frame-shift mutations or premature stop codons. However, HDR is a more accurate mechanism in which a homologous DNA template is utilized to repair the DSB, resulting in a more precise restoration of the target DNA sequence [16].
Fig. 2.
Mechanism of CRISPR/Cas9
Significance
CRISPR-Cas9 has significant applications in many sectors, including medicine, agriculture, and basic biological investigation.
Medicine
The utilization of CRISPR-Cas9 in the medical field has promise for treating genetic disorders through correcting DNA mutations. This approach can be applied to treat various medical conditions, including genetic disorders such as cystic fibrosis and sickle cell disease, as well as complex illnesses like cancer and HIV [14, 17].
Agriculture
CRISPR-Cas9 can be employed in agriculture so as to increase crop yields, improve nutritional composition, and boost resistance against pests and diseases. This technology may address the urgent need for sustainable and resilient food systems in light of the growing world population and climate change effects [18].
Basic biological research
The application of CRISPR-Cas9 has considerably enhanced our capacity to engage in fundamental biological investigations. Researchers now have easier access to manipulate model organism genomes to explore gene function, provide disease models, and investigate genetic interactions. The use of this technology has significantly sped up our understanding of genetics and cellular biology [19].
Telomeres and telomerase editing
Telomeres are the protective structures considered as the heterochromatin repetitive sequences, consisting of TTAGGG units at the end of chromosomes, which are covered by six protein complexes known as Shelterin. Shelterin, along with telomeres, plays a role in both binding and telomere length protection. In eukaryotic cells, the special structures at the ends of the chromosomes that prevent the separation of terminal bases and the shortening of chromosomes and end joining of chromosomes are called telomerase, a ribonucleoprotein consisting of a reverse transcriptase (TERT) and an RNA subunit (TERC), playing a role as a template to increase telomere length. Telomerase is responsible for maintaining the telomere length in some cells, such as stem cells, lymphocytes, and proliferating cells; however, the telomere length is different owing to the different activity of telomerase [20, 21]. It is well-established that alterations in telomere length, resulting in telomere damage, lead to cellular disorders, including aging and various diseases [22]. CRISPR/Cas9 is now a valuable tool for investigating the impact of mutations in living cancer cells and for modeling cellular aging. Researchers can employ the CRISPR/Cas9 method to study telomere damage, specifically at the DNA double-strand break and single-nucleotide levels (Fig. 3). For instance, researchers apply it to induce DNA double-strand breaks (DSBs), activate the repair system, and completely remove telomeres [23, 24]. Besides, a new class of CRISPR-based tools efficiently corrects single-nucleotide changes in cell lines, animal models, and perhaps the clinic [25]. Research shows that when telomeres shorten and DNA gets damaged in key tissues, it leads to problems in those areas and can predict the development of age-related diseases, such as cancer, in other tissues [26–28].
Fig. 3.
Telomeres and Telomerase Editing by CRISPR/Cas9
The most common TERT promoter mutation in malignant melanoma, which is associated with increased mRNA and protein expression, telomerase activity, telomere length, and very poor prognosis, highlights the critical role of telomerase in cancer progression. Investigation of this malignancy has shown that the frequency of mutations in the TERT promoter is higher than the occurrence of all known mutations in melanoma [29]. By manipulating the hTERT promoter, expression can be amplified or reduced, a phenomenon likely occurring in cancer cells or cells undergoing aging and death [30, 31]. Using CRISPR and dual CKM and TERT promoters, inhibition of malignant osteosarcoma cells and induction of apoptosis in cancer cells were observed [32]. These findings suggest that with structural modifications to telomerase, it is possible to design control mechanisms for this enzyme, some of which may be implemented within living organisms.
Adding a halo protein tag to the N-terminal end of TERT or introducing a single DNA substitution mutation in the hTERT promoter can impact telomerase expression, as observed in urothelial cancer cells where telomerase levels are reduced [33, 34]. Epigenetic modification, such as m6A, is a key regulator of telomerase activity and can be modulated using CRISPR genome editing. For instance, if the m6A regulator hnRNP affects hTERC, it could activate telomerase and delay stem cell aging. Similarly, if ALKBH5 acts as a methylation regulator influencing telomerase RNA (hTER), it could be pivotal in controlling telomerase activity and cellular function [35–37].
Alternative lengthening of telomeres (ALT) is a cellular mechanism that allows some cancers to extend their telomeres. Deletion of the TERT or TERC subunits of the telomerase enzyme using CRISPR can activate the ALT mechanism, leading to increased telomere length. Similarly, deleting hTERT also results in telomere elongation [30, 38–40]. Reprogramming fibroblast cells with hTERT is being used to create new models for studying Werner syndrome [30, 41]. Mutations in the TERT gene promoter are the most common clonal oncogenic mutations in glioblastoma, one of the most lethal cancers, making telomerase a promising therapeutic target. When TERT is inactivated, glioblastoma cells lose their ability to replicate due to telomere shortening and chromatin bridge formation over time. CRISPR technology can be employed to eliminate TERT expression in glioblastoma cell lines with TERT promoter mutations [42].
Epigenetics—editing
During genomic editing, there is a comprehensive alteration in the nucleotide sequence configuration, specifically targeting the encoded regions [43]. This procedure may result in the formation of new antigens due to genetic mutations [30]. However, epigenetic editing is a form of manipulation through a non-cutting process, and it has a persistent and reversible nature [43]. Instead of altering the fundamental DNA codes, it targets the sections of DNA responsible for controlling gene transcription and the way that DNA sequences interact with proteins [43]. This process can change the molecular makeup of the chromatin structure, and the way DNA is packaged [43]. Epigenetic editing can also reactivate genes that are generally only expressed in particular organs with distinct immune traits [30].
Epigenetic editing can be performed via four main mechanisms, including DNA methylation, post-histone translational modification, chromatin remodeling regulation, and regulation of noncoding RNA [30, 44]. DNA methylation is a regulatory mechanism that plays a key role in cell differentiation, tissue development, and homeostasis [44]. It is associated with physiological and pathological processes like gene expression, chromosomal inactivation, aging, and disease in tissues [44]. During the methylation process, DNA covalent modifications are made in the cytosine of the nucleotide sequence of CPG within the promoter region, which suppresses transcription [43, 45, 46]. The DNA methyltransferases (DNMTs) are catalyzed into 5-methyl cytosine (5MC) by S-adenosyl methionine (SAM) as a methyl donor. Of note, a group of proteins known as epigenetic writers, epigenetic readers, and epigenetic erasers regulates the methylation process. Alterations across the genome are made by the ‘writer’ that is identified by the ‘reader,’ and if these changes are not needed, the ‘eraser’ removes them [45].
There are four important types of DNMTs proteins: NMT3A, DNMT3B, DNMT1, and DNMT3L. NMT3A and DNMT3B belong to the group of de novo methyltransferases [44, 45]. They are involved in the methylation of CPG dinucleotide islands during the embryonic period [44]. DNMT1 preserves the methylation pattern by adding methyl groups to hemimethylated CPG during chromosome replication [44, 45]. Unlike other types, DNMT3L lacks enzymatic function [43]. The main function of DNA methyltransferases in epigenetic processes is to act as fusion proteins in the methylation of off-target DNA. By targeting it, the target gene can be repressed by inducing de novo 5mc [46]. If a mutation occurs in DNMT, it impacts both catalytic and multimerization activity through the activation of protein kinase and reduces the amount of off-target 5MC and constrains methylation near the target sites [46]. The post-translational modification of the histone is one of the mechanisms of epigenetic editing and is considered a covalent modification that mainly includes methylation, acetylation, ADP-ribosylation, phosphorylation, ubiquitylation, SUMOylation, and glycosylation [47].
It should be mentioned that stone acetylation is a process of epigenetic modification, affecting the function of proteins. The neutralization of unmodified lysine residual loads is carried out through the actions of two enzymes, histone acetyl transferase (HAT), and histone deacetylases (HDAC) [45]. Through doing so, they reduce the electrostatic interaction between negatively charged DNA and histone [48]. Subsequently, the structure of chromatin can be alternately opened and closed, leading to the relaxation of the chromatin [49]. As a result, the transcription rate increases [45]. All aspects of this process potentially can affect the composition of proteins and their interactions with cofactors, substrates, and macromolecules [45, 50]. Besides, it should be noted that mutations in the HAT enzyme contribute to its diminished function, which has been reported in numerous ailments like cancer [45]. In the histone methylation process, methyl groups are added to specific amino acids in histone proteins [51]. This process acts as a transcription suppressor and gene silencer [46, 51]. The residues of lysine and arginine in the tail of the histone can be the target of histone methylation [45, 46]. In contrast to histone acetylation, in histone methylation, the DNA and histone physical interaction does not result in neutralizing the histone load [45]. Subsequently, histone methylation provides a more complex chromatin structure compared with histone acetylation [45, 46]. Nonetheless, it can generate a more durable and stable post-translational correction. Histone methylation is catalyzed by histone methyltransferases (HMTs); however, it can be removed by histone demethylase (HDMs) enzymes, allowing for dynamic regulation of gene expression and chromatin remodeling [51]. It is worth mentioning that zinc finger nuclease (ZFN), transcription activator-like effector nuclease (TALEN), and CRISPR/Cas9-based RNA-guided DNA endonuclease as the targeted genome-editing tools are effective for modeling human disease through the creation of isogenic cells and transgenic animals [52] (Fig. 4) (Table 1). These efficient programmable chimeric nucleases are much faster and more cost-effective in comparison to the traditional homologous recombination-based mutagenic approach [53]. These tools investigate the role of epigenetic modulators in gene activity in native tissues and cell-type-specific chromatin states [54]. Among the mentioned tools, CRISPR-based genome-editing tools are a more usable and popular system for gene-editing and gene transcription adjustment. This is due to its high versatility with chemical modification, efficiency, multiplexity, simplicity in goal design, and cost-effectiveness [53].
Fig. 4.
Genome-editing tools are based on RNA-guided endonuclease, DNA based on zinc finger nuclease (ZFN), transcription-like activating nuclease (TALEN)
Table 1.
Overview of Targeted Gene-Editing Approaches in Various Age-related Disorders
| Disorders | Target Sites | Model (cell line) | Model of action | Delivery Method | Advantages | Disadvantage | Refs |
|---|---|---|---|---|---|---|---|
| Amyotrophic Lateral Sclerosis (ALS) | SOD1 gene | Patient-derived induced pluripotent stem cells (iPSCs) | Suppress the expression | Adeno-associated virus (AAV) vectors or lipid nanoparticles | Enhancements in motor function and an extended lifespan | May require complex delivery methods, and off-target effects could be a concern | [95, 96] |
| C9orf72 gene | iPSCs | Mutation | Viral vectors, typically AAV or lentivirus | Reinstatement of typical gene expression and enhanced the viability of motor neurons in a cellular environment | Potential challenges in achieving efficient and precise gene editing in clinical settings | [97] | |
| scAAV9-hIGF1 | Mice | Inhibition of the NF-κB signaling pathway | Adeno-associated virus serum type 9 (scAAV9-hIGF1) | Delay the initiation and decelerate the advancement of the ailment | Risks associated with viral vector delivery, including immune response and off-target effects | [98] | |
| Colorectal Cancer (CRC) | KRAS | HT29, WIDR, HCT116, LS174T, and HEK293T; SW480 and A549; and CFPAC‐1 | Knock out | Two‐vector lentivirus system |
GRB7‐PLK1 has a critical axis for RTK tolerance. PLK1 and thus a suitable target for synergizing MEK inhibitors in CRC patients with KRAS mutations |
Potential resistance development and off-target effects, leading to unintended gene alterations | [99] |
| Klotho | Caco‐2 | Knock out | Lentiviral vectors for delivering CRISPR/Cas9 to target cells | By causing apoptosis, Klotho gene overexpression in Caco‐2 cells by CRISPR/Cas9 inhibits cell growth | Limited to specific cancer types; potential off-target effects and challenges in gene delivery | [100] | |
| uPAR | CRL1619, CCCL247 | Knock out | Okayama‐Berg vector |
Knockout of the uPAR gene Leads to tumor growth inhibition, EGFR downregulation, and an increase in stemness markers |
May require complex delivery methods, and the long-term effects of gene knockout are still unknown | [101] | |
| Breast Cancer | MYC gene | Animal models | Target and suppress oncogenes | Lentiviruses, adeno-associated viruses, non-viruses like nanoparticles and lipids | Beneficial effects of CRISPR/Cas9 in cancer treatment | Off-target | [102, 103] |
| Ovarian Cancer | EGFL6 exons | SKOV3 | Delete | Lentiviral vectors | The removal of EGFL6 significantly reduced the migration, invasion, and growth of SKOV3 cells, while also promoting the death of tumor cells | Potential off-target effects and challenges in achieving precise gene editing in a clinical setting | [104] |
| BMI1, CXCR2, MTF1, miR-21, and BIRC5 | ovarian cancer cells | Knock out | Lentiviral CRISPR/Cas9 nickase vector | targets multiple genes associated with cancer progression, offering a comprehensive approach to treatment | Complexities in delivery and potential off-target effects could limit its effectiveness | [105–113] | |
| Prostate Cancer | Androgen receptor (AR) gene | LNCaP, VCaP, and PC-3 prostate cancer cell lines | Disruption of AR gene expression | AAV or lentiviral vectors | Slow down prostate cancer growth | Potential development of resistance and off-target effects could pose challenges in treatment | [114] |
| ER-β gene | Mice | Knock out | Lentiviral vectors | Regulated the prostate tumor size and acted as a tumor-suppressor gene | Delivery challenges and off-target effects may limit its clinical application | [114, 115] | |
| Erythropoietin-producing hepatocyte receptor A2 (EphA2) | DU145 and PC-3 tumor cells | CRISPR/Cas9 targeting of EphA2 to inhibit cell migration, invasion, and proliferation, potentially reducing metastatic potential | Calcium phosphate-based nanomedicine | Uses a novel nanomedicine approach for targeted delivery, potentially increasing treatment efficacy | Risks associated with nanomedicine delivery, including toxicity and off-target effects, are still under investigation | [116] |
The CRISPR/Cas9 system consists of two important components [55]: the guide component, responsible for specificity, which includes a single-stranded RNA molecule called sgRNA, and an effective component. sgRNA targets a specific DNA sequence in a genomic area and binds to the effective component via a scaffold RNA welding component. Cas protein is considered the most effective component, and the most common is Cas9 [55].
Promoters and insulators serve as barriers to inhibit interactions between enhancers and promoters, and they also hinder the spread of heterochromatin by neutralizing its effects [56]. In the process of gene activation using the CRISPR/Cas9 strategy, an effector domain (ED) can be fused to dCas9 (dCas9-ED) and precisely targeted to prevent insulator and promoter activity. This approach enables targeted DNA methylation, histone methylation or demethylation, and histone acetylation or deacetylation in mammalian cells, allowing for precise control of gene expression [53].
The efficiency of the CRISPR editing system depends on sgRNA and directs Cas9 protein for genome cutting [57]. dCas9-ED with sgRNA-mediated can transcribe local epigenetic localized states such as the histone sequence 5mC and modify transcription activity.
Due to reversible and flexible nature of epigenetic modification, it has the potential to be utilized as an anticancer treatment and enables the readjustments of the cancer epigenome [44]. Based on several studies on the genomic sequence of cancer cells, it has been clearly revealed that a high rate of changes in epigenetic regulatory genes occurs [30, 52]. These alterations affect all cancer hallmarks, such as the interaction between the immune system and tumor cells [58]. Hence, epigenetic editing mechanisms, in particular targeting epigenetic regulators, importantly provide an interaction between immune cells and tumor cells and enhance immune responses against tumors [54]. Of note, epigenetic regulatory mechanisms play a critical role in the interactions between cancer cells and immune cells in the tumor microenvironment through a number of specific proteins, including 1. ‘Writers’ add epigenetic marks to DNA. 2. ‘Erasers’ remove epigenetic marks from DNA. 3. ‘Readers’ are to identify specific epigenetic marks that act as mediators in DNA. 4. ‘Remodelers’ to moderate chromatin status [30].
In the cancer-immunity cycle, epigenetic mechanisms can influence cytokine production, leading to a dysregulated antigen presentation machinery in tumor cells. This dysregulation may prevent tumor cells from becoming invisible to T cells, thereby affecting immune recognition [30]. In addition, dysregulation results in gene reactivation with limited development and the creation of tumor differentiation antigens [58]. By regulating programmed death-ligand 1 (PD-L1) expression and binding to T cells, the proliferation of T cells, and the production and activity of cytokines can be suppressed, leading to T cell exhaustion [30].
The epigenetic machinery in immune cells plays a role in determining the fate of lymphocyte cells during lymphocyte development. Furthermore, it impacts the occurrence of functional and phenotypic changes when the adaptive immune system is activated. Alterations in epigenetic regulators play a direct role in the development of hematologic malignancies [58]. Chromatin organization is involved in T cell exhaustion. T cell exhaustion is defined via altered transcriptional and epigenetic states, reduced proliferative capacity and production of effector cytokines, sustained expression of inhibitory receptors, and impaired metabolic activity [59]. Exhausted T cells originated from infection have many open chromatin regions, providing a very different structure compared to effective lymphocytes [30]. Chromatin changes caused by T cell exhaustion can be reversible at the first stage; however, at the next stage they enter a fixed state, which cannot be reversed, leading to resisting changes and reprogramming [30].
During tumor development, the epigenome undergoes several changes, such as a decrease in DNA methylation across the genome and an increase in methylation, especially in the CpG islands of tumor-suppressor genes (TSGs). Epigenetic alterations can be considered as the drivers of tumorigenesis onset, especially in children’s brain tumors, which are defined as an aberrant epigenetic pattern [44, 54]. Epigenetic alterations can be considered as the drivers of tumorigenesis onset, especially in children’s brain tumors, which are defined as an aberrant epigenetic pattern [44].
The role of epigenetic disorders in human disease etiology and the aging process has been revealed. Epigenetic regulation disorder is recognized as a significant biological characteristic of aging and lifespan [56]. Aberrant transcription regulation caused by incorrect epigenetic patterns can accelerate aging and reduce life expectancy. Environmental factors can trigger heritable changes in the epigenome, leading to faster aging and the development of age-related diseases [53]. Genome and epigenomic engineering mechanisms involved in aging and age-associated diseases in humans reduce hemostatic processes over time and increase degenerative conditions, leading to an enhanced risk of many chronic diseases [44]. The contribution of genetic and epigenetic components in animal model lifespan has been recognized by applying the CRISPR-Cas9 technology. Thus, it is possible to quickly explore their roles in health and longevity with the functional investigation of genome-mapped genetic variants of the human genome [11]. Genome and epigenome-editing technology seem to be promising in age-related disease treatment. Therefore, targeted editing of genes associated with aging offers a new therapy for multiple diseases [53].
General mechanisms of aging in mammals include loss of histone associated with chromatin regeneration, imbalance in activation and suppression of histone changes, localized or general alterations in DNA methylation, change of transcription, nuclear reorganization, and subtractive or gradual changes in heterochromatin [44, 53]. It should be mentioned that the depletion of histone methyltransferase SUV39H1, which specifically methylates lysine nine in histone H3 and performs as a transcriptional suppressant, has been shown to improve DNA repair capacity, expand life expectancy, and delay the aging process in animal models [53].
Based on numerous lines of evidence, several afflictions have been shown to be related to epigenetic changes. Those alterations are linked to aging, cancer, and neurodegenerative disorders, such as amyotrophic lateral sclerosis (ALS) [60]. In other words, mechanisms involving aberrant DNA methylation could be relevant to human ALS pathobiology and therapeutic targeting. Abnormal DNA methylation patterns that originate from environmental factors play a role in the beginning or evolution of neurodegenerative diseases like ALS [61]. DNA methylation is involved in proliferation and differentiation of neural stem cells, synaptic plasticity, neuronal reparation, learning, and memory [62]. According to pathway analysis, genes with different methylation in sporadic ALS were particularly involved in calcium homeostasis, neurotransmission, and oxidative stress [63].
The etiology of colorectal cancer (CRC) is primarily associated with a number of genetic and epigenetic abnormalities within normal colonic epithelial cells, coupled with the reshaping of the tumor microenvironment in the surrounding stroma [60]. Based on prior investigations, epigenetic regulation is a key factor in acquiring underlying inherent drug resistance in CRC patients. It has been shown that unusual expression of the CRC epigenome contributes to resistance against conventional drugs, including cetuximab, 5-fluorouracil, and oxaliplatin. Epigenetic alterations often occur in the early progression of diseases and have a key role in nearly all cancer-associated pathways. Therefore, these alterations are prime candidates for cancer detection, diagnosis, and prognostic biomarkers. It is worth noting that to acquire the early CRC screening, classic CRC biomarkers, including mutations in KRAS and BRAF, methylation modifications of BMP3, NDRG4, and SEPT9, high expression micro-RNAs like miR-92a and miR-144, and the presence of F. nucleatum, are used in liquid biopsies [64–67].
The progression of breast cancer (BC) is a complicated and multistep procedure. This process includes interactions between genetic and epigenetic changes, influenced by various internal and external factors. Factors such as the cell’s internal microenvironment, availability of nutrients, cellular stress, and exposure to endocrine disruptors or carcinogens in the external environment can all play a role in BC pathogenesis. The two main players in epigenetic alterations in BC are the methylation of DNA and the modification of histone proteins. Overall, important genes that control cell growth, death, movement, and invasion are affected by the epigenetic changes associated with the progression of breast cancer [68]. Several BC genes show CpG island hypermethylation or aberrant promoter methylation, leading to silencing of tumor-suppressor genes involved in estrogen signaling, apoptosis, cell cycle, and DNA repair, such as HOXA5, TMS1, p16, RASSF1A, BRCA, E-cadherin, GSTP1, and p21/CIP1/WAF1 [69–75]. Histone methylation occurs on lysine and arginine side chains, adding methyl groups without changing protein charge. Methyltransferases and demethylases dynamically control this process [76]. Methylation affects gene regulation, activation, and repression. Various BC subtypes show differences in histone modifications, correlating with tumor characteristics and metastasis. Histone marks like H3K4 and H4K20 play key roles in cancer progression. Epigenetic changes impact gene expression and therapeutic targets in BC treatment [77, 78]. The enhancer of zeste homolog 2 (EZH2) is a key histone methyltransferase in BC, upregulated and promoting EMT [79]. Lysine methyltransferase 2 (KMT2) family members facilitate growth by activating estrogen-dependent genes [68]. The histone methylase disruptor silencing 1-like (DOT1L) is a potential therapeutic target for invasive BC [80]. Lysine-specific histone demethylase 1 (LSD1) negatively regulates cell growth genes [81]. Several demethylases (KDMs) are involved in BC progression and metastasis [82]. Plant homeodomain finger proteins 8 (PHF8) and PHF20L1 have oncogenic roles in BC metastasis [83, 84]. Noncoding RNA, like long noncoding RNAs (lncRNAs) and micro-RNAs (miRs), plays a crucial role in regulating gene expression in breast cancer. Micro-RNAs, especially the let-7 family, miR-148a & miR-152, miR-125b, miR-126, miR-31, miR-663, miR-148, miR-9-1, miR-152, and miR-124a3, have been studied a lot for how they affect gene activity in breast cancer [85–89].
Ovarian cancer (OC) also utilizes epigenetic modifications, such as DNA methylation and histone deacetylation, to manipulate gene expression for cell division and metastasis. One significant example of hypermethylation is reported the silencing of BRCA genes [90]. In addition, two gene sets have been proposed to be related to OC. First gene set have been proposed to be associated with frequency of histotype-specific DNA methylation including BRCA1, PTEN, CDKN2A, MLH1, RASSF1A, and CDH1. Second gene set focused on chromatin remodeling such as ARID1A, SPOP, and KMT2D. Targeting these processes with pharmaceutical agents may reverse unusual changes in cancer cells, inhibiting growth, invasion, metastasis, and chemotherapy resistance [91].
Epigenetic abnormalities are intricately linked to the development and progression of prostate cancer (PC). Similar to cancers mentioned above, aberrant DNA methylation, histone modifications, and noncoding ribonucleic acids play a role in the initiation and progression of PC [92]. The hypermethylated genes are involved in DNA repair, cell cycle, growth suppression, apoptosis, and cell adhesion, such as GSTP1, MGMT, CDKN2, RASSF1, APC, RARβ, DCR1, DCR2, XAF1, TMS1, CDH1, and CD44 [93]. It should be mentioned that PC also harbors genomic hypomethylation. However, the late phase of PC tumorigenesis, such as metastasis, often reports DNA hypomethylation more frequently than the early stage [94]. Hypomethylated genes in PC include PLAU, HPSE, and CYP1B1. Furthermore, changes in histone modifications, another form of epigenetic modification, significantly influence PC. Researchers have highlighted methylation, acetylation, phosphorylation, and ubiquitination among these modifications [93]. Hypomethylated genes in PC include PLAU, HPSE, and CYP1B1. Furthermore, changes in histone modifications, another form of epigenetic modification, significantly influence PC. Among these modifications, methylation, acetylation, phosphorylation, and ubiquitination have been highlighted [90]. Furthermore, researchers have proposed a relationship between two gene sets and OC. The first gene set has been proposed to be associated with the frequency of histotype-specific DNA methylation, including BRCA1, PTEN, CDKN2A, MLH1, RASSF1A, and CDH1. The second gene set focused on chromatin remodeling, such as ARID1A, SPOP, and KMT2D. Targeting these processes with pharmaceutical agents may reverse unusual changes in cancer cells, inhibiting growth, invasion, metastasis, and chemotherapy resistance [91].
Epigenetic abnormalities are intricately linked to the development and progression of prostate cancer (PC). Similar to cancers mentioned above, aberrant DNA methylation, histone modifications, and noncoding ribonucleic acids play a role in the initiation and progression of PC [92]. The hypermethylated genes are involved in DNA repair, cell cycle, growth suppression, apoptosis, and cell adhesion, such as GSTP1, MGMT, CDKN2, RASSF1, APC, RARβ, DCR1, DCR2, XAF1, TMS1, CDH1, and CD44 [93]. It should be mentioned that PC also harbors genomic hypomethylation. Nonetheless, DNA hypomethylation is more frequently reported in the late phase, such as metastasis, rather than the early stage of tumorigenesis in PC [94]. Hypomethylated genes in PC include PLAU, HPSE, and CYP1B1. Moreover, alterations in histone modifications as another epigenetic change play a key role in PC. Among these modifications, methylation, acetylation, phosphorylation, and ubiquitination have been highlighted [93]. Understanding how epigenetic regulatory mechanisms work will help us gain a better insight into PC and identify new epigenetic biomarkers that can indicate the risk of developing cancer, predict how patients will respond to drugs, or determine their likelihood of a poor prognosis.
CRISPR-Cas9 therapeutic role in age-related diseases
CRISPR-Cas9 gene therapy in amyotrophic lateral sclerosis (ALS)
Amyotrophic lateral sclerosis (ALS), colloquially referred to as Lou Gehrig’s disease, is a degenerative neurological condition characterized by the progressive impairment of motor neurons located in the brain and spinal cord [117]. At present, ALS lacks a definitive cure, and the available treatment modalities remain constrained [118]. Nevertheless, recent advancements in gene therapy applying the CRISPR-Cas9 approaches have exhibited potential in the treatment of this debilitating ailmen[119]. As the CRISPR-Cas9 system is a highly effective mechanism to manipulate the DNA sequence of cells, this method is able to modify patient-derived induced pluripotent stem cells (iPSCs). IPSCs can be further developed into motor neurons for the purposes of disease modeling and drug screening [120]. Superoxide dismutase 1 (SOD1) is the gene being studied in relation to ALS, as it has been connected to the development of this disease. A particular subset of familial ALS instances exhibit changes in the SOD1 gene. Furthermore, the abnormal accumulation of SOD1 protein has been identified in both familial and sporadic cases of ALS [121].
Several investigations have shown that the effective application of CRISPR-Cas9 technology in rectifying a mutation found in the SOD1 gene leads to notable enhancements in motor function and an extended lifespan [117, 120, 122]. Another potential approach includes using CRISPR-Cas9 technology to target and suppress the production of harmful proteins, such as TDP-43 and FUS, known to play a role in causing ALS. Several studies have revealed that this approach is practical and effective in cellular and animal models of ALS. Currently, clinical trials are being carried out to evaluate how safe and effective this method is for human subjects [120]. Another potential approach includes using CRISPR-Cas9 technology to target and suppress the production of harmful proteins, such as TDP-43 and FUS, known to play a role in causing ALS. Several studies have revealed that this approach is practical and effective in cellular and animal models of ALS. Currently, clinical trials are being carried out to evaluate how safe and effective this method is for human subject [123]. In other studies, the delivery of self-complementary adeno-associated virus serum type 9 encoding the human insulin-like growth factor 1 (scAAV9-hIGF1) was administered to the subarachnoid space of mice with ALS utilizing the CRISPR-Cas9 system. The results showed that using scAAV9-hIGF1 significantly helped to slow down the start and progression of the disease by blocking the NF-κB signaling pathway [124].
CRISPR-Cas9 sys gene therapy in colorectal cancer
Targeting the CRISPR-associated protein-9 nuclease (Cas9) within a living cell enables the highly efficient genome editing of CRISPR/Cas9 technology [126, 127]. The CRISPR/Cas9 method has been applied to in vitro CRC by inhibiting some molecular pathway [100, 101, 128]. The estimation showed that CRC is the second leading cause of deaths among cancers and the third most common malignancy worldwide [129–132]. Telomerase reverse transcriptase (TERT and hTERT in humans) is the catalytic portion of telomerase which is known as the limiting factor of this enzyme [133]. Classified as a ribonucleoprotein, telomerase consists of both RNA and proteins. Specifically, it consists of two molecules of telomerase reverse transcriptase, telomere RNA, and dyskerin [134–136]. Each of these subunits plays an important role in the biological function of telomerase. Nevertheless, it is critical to note that the catalytic component, known as telomerase reverse transcriptase (TERT or hTERT in humans), is typically the limiting factor when it comes to telomerase activity and the elongation of telomeres [133].
The transforming growth factor b (TGF- β) pathways are context-dependent signal transduction cascades that can act as both a tumor promoter and tumor suppressor [137]. TGF-β1 acts as a controller of proliferation and differentiation in many tissues [138]. Besides, higher TGF-b1 protein expression is related to advanced tumor stages and metastasis in colorectal carcinoma [139]. By using CRISPR/Cas9 technology, previous studies indicated that TGF-β1 gene expression has a direct correlation with tumor progression in non-small cell carcinoma (NSCLC), gastric cancer, prostate cancer, and also colorectal cancer [140–143].
Vaults are barrel-shaped ribonucleoprotein (RNP) particles in many eukaryotic cells, with their role being unclear still [144]. These structures contain MVP (major vault protein), TEP1, vPARP, and noncoding RNAs [145, 146]. It has been demonstrated that they participate in developing drug resistance in malignant cells [147]. By using CRISPR/Cas9, current studies have demonstrated that MVPs are involved in regulating migration and proliferation of cancer cells and are highly expressed in tumor metastasis in colorectal adenocarcinoma; they are also essential components for HAP1 cells to survive [148]. CST, which stands for the ssDNA-binding trimetric complex, is composed of the CTC1, STN1, and TEN1 subunits [149, 150]. This complex plays a crucial role in various aspects of telomere replication [150, 151]. Via applying CRISPR/Cas9 technology, recent studies utilized have demonstrated that CTC1-STN1 actively terminates telomerase activity to prevent excessive extension of the G-overhang [152]. In addition, it has been described that STN1-TEN1 is essential for C-strand synthesis during telomere replication [152].
CRISPR-Cas9 gene therapy in breast cancer
BC is the world’s second most prevalent cancer diagnosis. Its heterogenic nature is due to the involvement of various gene mutations [102, 153]. While traditional BC therapies like radiation and chemotherapy can be beneficial, they often cause adverse effects and fail to target-specific mutations. In contrast, CRISPR-Cas9 gene therapy can help target-specific gene mutations in BC [154]. CRISPR-Cas9 offers exceptional accuracy, efficiency, affordability, and reduced risk, which makes it a better option than previous gene therapies such as ZFN and TALEN [153–156]. CRISPR-Cas9 technology could repair cancer-causing mutations in animal models as a first step toward translational applications. This method enables scientists to determine the responsible gene in various cancers, including BC. This is achieved by creating and deploying guide RNAs (gRNAs) that specifically target-specific sections of the gene [157]. Targeting and suppressing oncogenes with CRISPR-Cas9 can slow down the progression and metastasis of BC [158]. The majority of clinical trials that use CRISPR-Cas9 rely on genetically engineered T cells and cancer immunotherapy [159]. Moreover, viruses such as lentiviruses and adeno-associated viruses and non-viruses like nanoparticles and lipids can deliver CRISPR/Cas9 components to target tissues in vivo. Gold nanomaterials offer several benefits that make them useful in theranostics, delivery vehicles, imaging, and photothermal agents [102, 160, 161]. BC CRISPR-Cas9 gene therapy may target the MYC gene (a type of oncogene). Schuijers et al. used CRISPR-Cas9 to downregulate the MYC gene, which increases 30–50% in high-grade BC. Although there are several beneficial effects of CRISPR/Cas9 in cancer treatment, it has also been associated with higher carcinogenesis [162]. CRISPR-Cas9 stimulates both genotoxicity and immunotoxicity, with genotoxicity linked to off-target outcomes. These issues can lead to unintended mutations that may harm humans [163]. Furthermore, tumor heterogeneity is considered an issue in treatment because tumors are typically made up of distinct subclones [45].
CRISPR-Cas9 gene therapy in ovarian cancer
The seventh most prevalent disease among women worldwide is OC. This variation increases the genetic susceptibility to OC and can be inherited as a result of rare, moderately to highly penetrant mutations. A woman’s lifetime risk of acquiring OC is 1–2%, and there is a 1 in 100 probabilities of dying from it. Every year, OC causes 239,000 new cases and 152,000 fatalities worldwide [164, 165]. Additionally, ovarian malignancies typically spread to distal organs such as the liver, colon, and omentum through peritoneal dissemination from the main tumor site [166]. CRISPR-Cas9, a promising gene-editing technique, offers the potential to treat OC through gene-editing procedures [167]. CRISPR knockouts of various oncogenes implicated in the OC etiology, including BIRC5, BMI1, MTF1, CXCR2, and miR-21, have been described in studies, demonstrating the promise of the CRISPR-Cas9 approach to efficiently treat OC [168].
OC progresses and metastasizes due to tumor angiogenesis [169] and exhibits high levels of epidermal growth factor-like domain multiple (EGFL6) protein expression, suggesting that this protein is crucial for encouraging tumor angiogenesis [170]. EGFL6 is a member of the EGFL (epidermal growth factor-like) family of proteins [171]. Embryos and most tumor tissues express EGFL6 extensively. EGFL6 enhanced the OC stem cells’ asymmetric division, phosphorylated ERK, and supported the proliferation and spread of OC. Additionally, Noh et al. found that the TWIST transcription factor regulated EGFL6 and helped the growth of new blood vessels in OC by managing integrin/Tie2/AKT [172]. In one study, EGFL6 was knocked out using CRISPR-Cas9 by transfecting a guide RNA (gRNA) that was specifically engineered to target EGFL6 exons into an OC cell line. The EGFL6 gene was removed from the ovarian cancer cell line SKOV3 using the CRISPR/Cas9 system after a guide RNA was designed to specifically target the EGFL6 exons. The elimination of EGFL6 dramatically decreased the migration, invasion, and proliferation of SKOV3 cells and facilitated the death of tumor cells. This study suggests that EGFL6 might regulate the FGF-2/PDGFB signaling pathway, which could influence blood vessel formation, movement, and growth of ovarian cancer cells [170].
DNMT1 is a key biological target for cancer epigenetic treatment [173]. It is an oncogene that inhibits tumor-suppressor genes when it is overexpressed and is essential for cancer stem cell maintenance. It is associated with carcinogenesis and OC relapse, tumorigenesis, and resistance. Frequently, it has been shown that patients with poor prognoses had elevated levels of DNMT1. Thus, DNMT1 has been considered a viable molecular target for OC therapy [106]. Targeting DNMT1 with CRISPR-Cas9 was found to suppress DNMT1 expression and slow tumor growth, indicating the promise of DNMT1 as an OC therapeutic target. In a recent study, for the delivery of CRISPR plasmid DNA co-expressing Cas9 and single-guide RNA addressing the OC-related DNMT1 gene, a folate receptor-targeted liposome (F-LP) was employed. Cas9 plasmids were effectively delivered to cancer cells using cationic liposomes to edit the targeted genome in vivo. Nonetheless, additional research on non-specific anticancer effects is required [174].
Patients with OC typically have a poor prognosis or low survival rate because BIRC5 is highly expressed in these cells. The gene BIRC5 encodes the survivin protein, which is an inhibitor of apoptosis family members [175]. Its expression is frequently linked to tumor spread and chemoresistance. In contrast to expression in normal adult tissues, survivin is highly expressed in a wide range of malignancies, including OC [176]. It has also been demonstrated that survivin has a role in tumor spread and chemoresistance in OC [177]. To modify BRIC5 expression, one may use the lentiviral CRISPR/Cas9 nickase vector or the tiny pharmacological inhibitor YM155. This approach would prevent EMT, significantly reduce cell growth and invasion, and initiate apoptosis. Loss of BIRIC5 expression decreased TGF-pathway activity [178].
In a study, the efficiency of the CRISPR-Cas9 genome-editing technique in knocking down the expression of the ABCB1 gene in OC cells resistant to adriamycin was examined [179]. Multidrug resistance (MDR) is an important challenge to effective OC chemotherapy [180]. The current study examined how well CRISPR-Cas9 can help overcome multiple drug resistance (MDR) in adriamycin-resistant ovarian cancer cells. A typical phenomenon in cancer cells is the overexpression of ATP-binding cassette (ABC) drug transporters, which is linked to chemo-resistant phenotypes and multiple drug resistance (MDR) in solid tumors [179]. Up-regulation of ABCB1/P-gp in OC is highly correlated with a poor response to chemotherapy [113]. The ABCB1 gene was downregulated using the CRISPR-Cas9 gene-editing technology, which led to a sharp decrease in ABCB1 gene expression and a rise in the doxorubicin sensitivity of cells transfected with sgRNAs [179].
CRISPR-Cas9 vectors are effective methods for altering noncoding RNAs, such as miRNA genes, in addition to gene expression [181]. According to one study, lentiviral CRISPR-Cas9 vectors are extremely successful at disrupting miRNA expression [182]. miR-21 had oncogenic properties in OC [183]. Pre-miR-21 sequence alteration decreased cell invasion, migration, and proliferation while increasing the sensitivity to chemotherapeutic drugs. E-cadherin, a hallmark of epithelial cells, was upregulated, and mesenchymal markers were downregulated in both cells when the epithelial-to-mesenchymal transition (EMT) was blocked by miR-21 disruption [182].
PARP inhibitors have significantly improved epithelial OC treatment [184]. Particularly for those with recombination repair abnormalities or BRCA1/2 mutations [185]. Adult patients with advanced OC who have a BRCA mutation can take olaparib as maintenance therapy. It is a PARP inhibitor, meaning that it prevents the DNA repair enzyme poly ADP-ribose polymerase (PARP) from working [186]. It works against breast, ovarian, and prostate cancers as well as malignancies in those with inherited BRCA1 or BRCA2 mutations. After the discovery of 93 genes, the CRISPR-Cas9 mutagenesis test was used to predict the olaparib response. The study identified the genes NBN, MUS81, BRCA2, RAD51B, and ATM as predictive markers. Along with three PARPi and carboplatin, the study looked at nine important candidate genes, and the results showed similar dropout rates that were not affected by the genes or the drugs used. Across EOC cell lines, CDK12 is frequently vulnerable to olaparib-induced survival and proliferation, underscoring its potential as a therapeutic target [187, 188].
A different investigation was conducted on poly (ADP-ribose) polymerase-1 (PARP-1) using the CRISPR-Cas9 technology, which had an important impact on controlling the cell cycle, responding to DNA damage, and apoptosis, as a possible therapeutic target. Cancer cells die as a result of PARP-1 inhibition. Inhibiting PARP-1 using CRISP-Cas9 resulted in SKOV3 cell (OC cell line) apoptosis and limited cancer growth [189]. These investigations demonstrate the CRISPR-Cas9 system’s therapeutic promise for treating OC. However, there are restrictions that limit CRISPR-Cas9 biomedical applications and the deployment of ovarian cancer gene therapy. Examples include CRISPR-Cas9’s effect on nontargeting normal cells and off-target DNA damage [190].
CRISPR-Cas9 gene therapy in prostate cancer
PC is the second most common cancer diagnosis made in men and the fifth leading cause of death globally [191]. PC is initially treated by inhibiting androgen receptor signaling, but eventually progresses to castration-resistant PC (CRPC). Next-generation androgen receptor (AR) signaling inhibitors have improved survival for patients with CRPC, but resistance remains an issue. Scientists can use CRISPR/Cas9 to target the AR gene, which plays a major role in the growth and survival of PC cells. By editing this gene, the growth and metastasis of cancer cells can be reduced. Inhibiting the AR function could help slow down PC growth [115, 192]. The CRISPR/Cas system disrupted the AR at specific sites and inhibited the growth of androgen-sensitive PC cells by inducing cellular apoptosis [193].
The use of CRISPR/Cas technology to target the AR gene and the combination of AR inhibition with PRMT1 inhibition may provide promising therapeutic strategies for the treatment of advanced PC [194]. Also, by using CRISPR/Cas9, knockout of the ER-β gene in the mouse genome effectively regulated the prostate tumor size and acted as a tumor-suppressor gene [115]. In another research, researchers found that a CRISPR-Cas9-based genome-editing calcium phosphate-based nanomedicine was developed to target erythropoietin-producing hepatocyte receptor A2 (EphA2), a receptor presented in tumor cells, for PC therapy [116]. The results show that removing IL30 led to a decrease in cancer-causing genes and an increase in genes that help prevent tumors, which slowed down tumor growth and spread in pancreatic cancer models. The researchers suggest that CRISPR/Cas9-mediated targeting of Interleukin-30 (IL30) could be a promising clinical approach for curbing PC progression [195].
An additional prospective strategy showed that CRISPR-Cas9 can be effectively delivered to cancer cells using graphene quantum dots as carriers. This approach targets the TP53 gene mutation, which is commonly overexpressed in many types of cancer. The delivery of CRISPR RNP and a gene repair template leads to successful restoration of the healthy TP53 gene, promoting cellular repair pathways and causing cancer cell apoptosis [196]. They found that the use of nanocarriers for CRISPR/Cas9 delivery holds great potential for targeted gene therapy in PC treatment. The aptamer-liposome-CRISPR/Cas9 chimera provides promising evidence for the clinical development of this technology [197]. Findings suggest that the aptamer-liposome-CRISPR/Cas9 chimera has enormous potential as a targeted gene therapy for PC. The use of an RNA aptamer as a ligand allows for specific binding to cancer cells, while the cationic liposomes enable efficient delivery of therapeutic CRISPR/Cas9 [198]. These findings suggest that metal–organic frameworks (MOFs) could be a promising platform for targeted gene therapy in PC, and the use of ZIF-C through RNAi and CRISPR/Cas9 technologies could provide more precise and effective treatment options. The use of the addition of a green tea phytochemical coating may also enhance the delivery of therapeutic agents to cancer cells [199].
The activation of the activating protein-1 (AP-1) transcription factor has been associated with the onset and progression of cancer, particularly in prostate cancer, where the proto-oncogenes JUN and FOS play a pivotal role. CRISPR/Cas9-mediated knockout of nuclear paraspeckle assembly transcript 1 (NEAT1) in AGS cells caused S phase cell cycle arrest and apoptosis and upregulated FAS level [200]. Besides focusing on TP53 mutations, CRISPR/Cas9 can also help find new genes that influence how PC cells respond to PARP inhibitors [201]. These studies demonstrate the power of CRISPR/Cas9 technology in identifying genes involved in the response to PARP inhibitors in PC cells. By targeting both known and unknown genes, researchers have identified potential biomarkers for PARP inhibition and new therapeutic targets for overcoming resistance [202]. PC modeling has led to the development of tools to examine therapies for this type of cancer. Cultured somatic rat models for prostate cancer have been produced, and nanomedicine can target cancer cells with reduced side effects and increased efficacy. PEG-targeted liposomal loaded nanomedicine is being investigated for potential treatment of PC, along with CRISPR/Cas9 gene editing [203]. The application of CRISPR/Cas9 technology in genetic screening has helped uncover the mechanisms behind cancer progression and drug resistance. This process has led to the discovery of new potential genes that can be targeted for therapy [204].
Concluding remarks and future perspectives
Aging is an intricate biological phenomenon marked by a progressive decline in cellular and physiological functions, rendering individuals increasingly susceptible to a spectrum of age-related diseases. This manuscript has meticulously explored the molecular underpinnings of aging, spotlighting key contributors such as telomere shortening, diminished telomerase activity, genetic mutations, and epigenetic alterations. These elements collectively precipitate cellular senescence—a state of permanent growth arrest that serves as a cornerstone for age-related pathologies. Within this context, the advent of CRISPR-Cas9 gene-editing technology emerges as a transformative tool, offering unprecedented precision in targeting and modulating these molecular drivers to mitigate cellular senescence and its associated diseases.
At the heart of aging lies the erosion of telomeres, the protective caps at chromosome ends, which shorten with each cell division. This process, exacerbated by reduced telomerase activity, accelerates cellular aging and heightens disease risk. Concurrently, mutations in genes governing DNA repair, metabolism, and inflammation accumulate over time, impairing cellular integrity. Epigenetic changes, including DNA methylation and histone modifications, further compound this decline by altering gene expression without modifying the DNA sequence itself. CRISPR-Cas9 addresses these challenges head-on, enabling precise interventions at both genetic and epigenetic levels. For instance, studies highlighted in this manuscript demonstrate its efficacy in targeting the LMNA gene in progeria mouse models, reducing toxic progerin production, and shedding light on aging pathways. Similarly, genome-wide CRISPR-Cas9 screens have unveiled novel senescence-promoting genes, broadening the therapeutic landscape.
The versatility of CRISPR-Cas9 extends beyond genetic correction to epigenetic reprogramming. By editing telomere-related genes, such as the TERT promoter in cancer cells, or modulating telomerase activity, CRISPR-Cas9 offers insights into telomere dynamics and their role in aging and malignancy. Its application in epigenetic editing—targeting DNA methylation or histone modifications—provides a reversible means to restore youthful gene expression patterns, holding promise for reversing age-related epigenetic drift. This dual capability underscores CRISPR-Cas9’s potential as a multifaceted tool in aging research and therapy. The manuscript further delineates CRISPR-Cas9’s therapeutic applications across a range of age-related diseases. In ALS, it targets genes like SOD1 and C9orf72, correcting mutations or suppressing toxic protein production, resulting in improved motor function and survival in preclinical models. In colorectal cancer, it disrupts pathways like KRAS or enhances tumor-suppressor genes, such as Klotho, inhibiting tumor progression. Breast cancer benefits from CRISPR-Cas9’s ability to silence oncogenes like MYC, while in ovarian cancer, it knocks out angiogenesis-promoting genes like EGFL6 or multidrug resistance genes like ABCB1, enhancing chemotherapy sensitivity. Prostate cancer research leverages CRISPR-Cas9 to target the androgen receptor (AR) gene, slowing cancer growth and offering novel therapeutic avenues. These examples collectively illustrate CRISPR-Cas9’s capacity to address both the hallmarks of aging and the specific molecular aberrations driving age-related diseases.
Despite its transformative potential, CRISPR-Cas9 is not without challenges. Off-target effects remain a critical concern, as unintended genetic modifications could compromise safety in therapeutic settings. Delivery mechanisms—whether viral vectors, nanoparticles, or liposomes—require optimization to ensure efficient and tissue-specific targeting. Ethical considerations, particularly regarding germline editing, demand rigorous scrutiny to balance innovation with societal implications. These hurdles necessitate ongoing refinement to translate CRISPR-Cas9 from bench to bedside effectively. Looking ahead, the future of CRISPR-Cas9 in aging and disease mitigation is bright yet contingent on continued innovation. Integrating this technology with AI could enhance precision by predicting off-target effects and optimizing guide RNA design. Advances in delivery systems, such as next-generation nanoparticles or bioengineered vectors, promise to improve efficacy and safety. Also, using CRISPR-Cas9 for more age-related issues—like other brain diseases besides ALS or heart problems—could make it more useful. Collaborative efforts to address ethical frameworks will further ensure its responsible deployment.
In summary, CRISPR-Cas9 stands at the forefront of a paradigm shift in combating cellular senescence and age-related diseases. By targeting the molecular roots of aging—telomere attrition, genetic mutations, and epigenetic dysregulation—it offers a pathway to extend lifespan and enhance health span. As research progresses, the synergy of CRISPR-Cas9 with cutting-edge technologies and a more profound understanding of aging biology holds the potential to revolutionize therapeutic strategies, fostering healthier aging and improving quality of life for aging populations worldwide. This manuscript underscores the promise and complexity of this endeavor, paving the way for a future where aging is not merely endured but actively managed.
Authors’ contributions
A.A., M.SH., R.D., and S.A. contributed to the hypothesis, investigating, gathering data, and writing the main text of the manuscript. P.L., A.HK., Z.Z., and H.GH. contributed to investigating, writing, and reviewing the final draft of the manuscript. H.GH., H.F., and, H.R. contributed to data gathering and designing figures and tables. as well as content and grammatical editing. S.T. and Q.B. contributed to the hypothesis, scientific, and structural editing, supervision, and verifying the manuscript before submission.
Funding
This research received no grant from any funding agency, commercial or not-for-profit sectors.
Availability of data and material
No datasets were generated or analysed during the current study.
Declarations
Ethics approval and consent to participate
Not applicable.
Competing interests
The authors declare no competing interests.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Alireza Azani, Malihe Sharafi and Reyhaneh Doachi contributed equally to this work.
Contributor Information
Safa Tahmasebi, Email: Safa.tahmasebi@sbmu.ac.ir.
Qumars Behfar, Email: q_behfar@yahoo.com, Email: Safa.tahmasebi@sbmu.ac.ir.
References
- 1.Tenchov R, Sasso JM, Wang X, Zhou QA. Aging hallmarks and progression and age-related diseases: a landscape view of research advancement. ACS Chem Neurosci. 2023;15(1):1–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Wang X, Ma Z, Cheng J, Lv Z. A genetic program theory of aging using an RNA population model. Ageing Res Rev. 2014;13:46–54. [DOI] [PubMed] [Google Scholar]
- 3.Miquel J. An update on the oxygen stress-mitochondrial mutation theory of aging: genetic and evolutionary implications. Exp Gerontol. 1998;33(1–2):113–26. [DOI] [PubMed] [Google Scholar]
- 4.Martin GM. Genetic engineering of mice to test the oxidative damage theory of aging. Ann N Y Acad Sci. 2005;1055:26–34. [DOI] [PubMed] [Google Scholar]
- 5.Proshkina EN, Solovev IA, Shaposhnikov MV, Moskalev AA. Key molecular mechanisms of aging, biomarkers, and potential interventions. Mol Biol. 2020;54(6):777–811. [DOI] [PubMed] [Google Scholar]
- 6.Asmamaw M, Zawdie B. Mechanism and applications of CRISPR/cas-9-mediated genome editing. Biolog Targets Therapy. 2021;15:353–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Harman D. The aging process. Proc Natl Acad Sci U S A. 1981;78(11):7124–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Beyret E, Liao H-K, Yamamoto M, Hernandez-Benitez R, Fu Y, Erikson G, et al. Single-dose CRISPR–Cas9 therapy extends lifespan of mice with Hutchinson-Gilford progeria syndrome. Nat Med. 2019;25(3):419–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wang W, Zheng Y, Sun S, Li W, Song M, Ji Q, et al. A genome-wide CRISPR-based screen identifies KAT7 as a driver of cellular senescence. Sci Transl Med. 2021. 10.1126/scitranslmed.abd2655. [DOI] [PubMed] [Google Scholar]
- 10.Wang W, Zheng Y, Sun S, Li W, Song M, Ji Q, et al. A genome-wide CRISPR-based screen identifies KAT7 as a driver of cellular senescence. Sci Translat Med. 2021;13(575):2655. [DOI] [PubMed] [Google Scholar]
- 11.Carneiro SP, Greco A, Chiesa E, Genta I, Merkel OM. Shaping the future from the small scale: dry powder inhalation of CRISPR-Cas9 lipid nanoparticles for the treatment of lung diseases. Expert Opin Drug Deliv. 2023;20(4):471–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Cox DBT, Platt RJ, Zhang F. Therapeutic genome editing: prospects and challenges. Nat Med. 2015;21(2):121–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Doudna JA, Charpentier E. The new frontier of genome engineering with CRISPR-Cas9. Science. 2014. 10.1126/science.1258096. [DOI] [PubMed] [Google Scholar]
- 14.Hsu PD, Lander ES, Zhang F. Development and applications of CRISPR-Cas9 for genome engineering. Cell. 2014;157(6):1262–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Jaganathan D, Ramasamy K, Sellamuthu G, Jayabalan S, Venkataraman G. CRISPR for crop improvement: an update review. Front Plant Sci. 2018;9:985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337(6096):816–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Knight SC, Xie L, Deng W, Guglielmi B, Witkowsky LB, Bosanac L, et al. Dynamics of CRISPR-Cas9 genome interrogation in living cells. Science. 2015;350(6262):823–6. [DOI] [PubMed] [Google Scholar]
- 18.O’Keeffe Ahern J, Lara-Sáez I, Zhou D, Murillas R, Bonafont J, Mencía Á, et al. Non-viral delivery of CRISPR–Cas9 complexes for targeted gene editing via a polymer delivery system. Gene Ther. 2022;29(3–4):157–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Shao M, Xu T-R, Chen C-S. The big bang of genome editing technology: development and application of the CRISPR/Cas9 system in disease animal models. Zoolog Res. 2016;37(4):191–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kim T, Tarangelo A. Telomeres and telomerase in cancer: overview and therapeutic potential. J Stud Res. 2022;11(3):1597. [Google Scholar]
- 21.Salimi-Jeda A, Badrzadeh F, Esghaei M, Abdoli A. The role of telomerase and viruses interaction in cancer development, and telomerase-dependent therapeutic approaches. Cancer Treat Res Commun. 2021;27:100323. [DOI] [PubMed] [Google Scholar]
- 22.Rossiello F, Jurk D, Passos JF, Dadda di Fagagna F. Telomere dysfunction in ageing and age-related diseases. Nature cell Biol. 2022;24(2):135–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Mao P, Liu J, Zhang Z, Zhang H, Liu H, Gao S, et al. Homologous recombination-dependent repair of telomeric DSBs in proliferating human cells. Nat Commun. 2016;7:12154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kim H, Ham S, Jo M, Lee GH, Lee YS, Shin JH, et al. CRISPR-Cas9 mediated telomere removal leads to mitochondrial stress and protein aggregation. Int J Mol Sci. 2017. 10.3390/ijms18102093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Davis DJ, Yeddula SGR. CRISPR Advancements for Human Health. Mo Med. 2024;121(2):170. [PMC free article] [PubMed] [Google Scholar]
- 26.Carneiro MC, Henriques CM, Nabais J, Ferreira T, Carvalho T, Ferreira MG. Short telomeres in key tissues initiate local and systemic aging in zebrafish. PLoS Genet. 2016;12(1):e1005798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bernadotte A, Mikhelson VM, Spivak IM. Markers of cellular senescence. Telomere shortening as a marker of cellular senescence. Aging (Albany NY). 2016;8(1):3–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kordyukova MY, Kalmykova AI. Nature and functions of telomeric transcripts. Biochemistry (Mosc). 2019;84(2):137–46. [DOI] [PubMed] [Google Scholar]
- 29.Guo Y, Chen Y, Zhang L, Ma L, Jiang K, Yao G, et al. TERT promoter mutations and telomerase in melanoma. J Oncol. 2022;2022:6300329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Brane AC, Tollefsbol TO. Targeting telomeres and telomerase: studies in aging and disease utilizing CRISPR/Cas9 technology. Cells. 2019. 10.3390/cells8020186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chiba K, Johnson JZ, Vogan JM, Wagner T, Boyle JM, Hockemeyer D. Cancer-associated TERT promoter mutations abrogate telomerase silencing. Elife. 2015;4:e07918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hu Y, Zhang H, Guo Z, Zhou J, Zhang W, Gong M, et al. CKM and TERT dual promoters drive CRISPR-dCas9 to specifically inhibit the malignant behavior of osteosarcoma cells. Cell Mol Biol Lett. 2023;28(1):52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Chiba K, Vogan JM, Wu RA, Gill MS, Zhang X, Collins K, et al. Endogenous telomerase reverse transcriptase n-terminal tagging affects human telomerase function at telomeres in vivo. Mol Cell Biol. 2017;37(3):e00541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Xi L, Schmidt JC, Zaug AJ, Ascarrunz DR, Cech TR. A novel two-step genome editing strategy with CRISPR-Cas9 provides new insights into telomerase action and TERT gene expression. Genome Biol. 2015;16:231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Yi M, Wang M, Xu Y, Cao Z, Ling Y, Zhang Z, et al. CRISPR-based m(6)A modification and its potential applications in telomerase regulation. Front Cell Dev Biol. 2023;11:1200734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Han S, Zhao BS, Myers SA, Carr SA, He C, Ting AY. RNA-protein interaction mapping via MS2- or Cas13-based APEX targeting. Proc Natl Acad Sci U S A. 2020;117(36):22068–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Xu C, Xie N, Su Y, Sun Z, Liang Y, Zhang N, et al. HnRNP F/H associate with hTERC and telomerase holoenzyme to modulate telomerase function and promote cell proliferation. Cell Death Differ. 2020;27(6):1998–2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hu Y, Shi G, Zhang L, Li F, Jiang Y, Jiang S, et al. Switch telomerase to ALT mechanism by inducing telomeric DNA damages and dysfunction of ATRX and DAXX. Sci Rep. 2016;6:32280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Min J, Wright WE, Shay JW. Alternative lengthening of telomeres can be maintained by preferential elongation of lagging strands. Nucleic Acids Res. 2017;45(5):2615–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Loe TK, Li JSZ, Zhang Y, Azeroglu B, Boddy MN, Denchi EL. Telomere length heterogeneity in ALT cells is maintained by PML-dependent localization of the BTR complex to telomeres. Genes Dev. 2020;34(9–10):650–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Wang S, Liu Z, Ye Y, Li B, Liu T, Zhang W, et al. Ectopic hTERT expression facilitates reprograming of fibroblasts derived from patients with Werner syndrome as a WS cellular model. Cell Death Dis. 2018;9(9):923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Aquilanti E, Kageler L, Watson J, Baird DM, Jones RE, Hodges M, et al. Telomerase inhibition is an effective therapeutic strategy in TERT promoter-mutant glioblastoma models with low tumor volume. Neuro Oncol. 2023;25(7):1275–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.<7.Genes Dev.-1998-Struhl-599–606.pdf>. [DOI] [PubMed]
- 44.Cao J, Yan Q. Cancer epigenetics, tumor immunity, and immunotherapy. Trends Cancer. 2020;6(7):580–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Chen M, Mao A, Xu M, Weng Q, Mao J, Ji J. CRISPR-Cas9 for cancer therapy: opportunities and challenges. Cancer Lett. 2019;447:48–55. [DOI] [PubMed] [Google Scholar]
- 46.Christensen DG, Xie X, Basisty N, Byrnes J, McSweeney S, Schilling B, et al. Post-translational protein acetylation: an elegant mechanism for bacteria to dynamically regulate metabolic functions. Front Microbiol. 2019;10:1604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Gjaltema RAF, Rots MG. Advances of epigenetic editing. Curr Opin Chem Biol. 2020;57:75–81. [DOI] [PubMed] [Google Scholar]
- 48.Gujral P, Mahajan V, Lissaman AC, Ponnampalam AP. Histone acetylation and the role of histone deacetylases in normal cyclic endometrium. Reprod Biol Endocrinol. 2020;18(1): 84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Hiranniramol K, Chen Y, Liu W, Wang X. Generalizable sgRNA design for improved CRISPR/Cas9 editing efficiency. Bioinformatics. 2020;36(9):2684–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Khalil AM. The genome editing revolution: review. J Genet Eng Biotechnol. 2020;18(1):68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Lau CH, Suh Y. Genome and epigenome editing in mechanistic studies of human aging and aging-related disease. Gerontology. 2017;63(2):103–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Lau C-H, Suh Y. Genome and epigenome editing in mechanistic studies of human aging and aging-related disease. Gerontology. 2017;63(2):103–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Nasir A, Bullo MMH, Ahmed Z, Imtiaz A, Yaqoob E, Jadoon M, et al. Nutrigenomics: epigenetics and cancer prevention: a comprehensive review. Crit Rev Food Sci Nutr. 2020;60(8):1375–87. [DOI] [PubMed] [Google Scholar]
- 54.Park JW, Han JW. Targeting epigenetics for cancer therapy. Arch Pharm Res. 2019;42(2):159–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Schrauben M, Dempster E, Lunnon K. Applying gene-editing technology to elucidate the functional consequence of genetic and epigenetic variation in Alzheimer’s disease. Brain Pathol. 2020;30(5):992–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Shams F, Bayat H, Mohammadian O, Mahboudi S, Vahidnezhad H, Soosanabadi M, et al. Advance trends in targeting homology-directed repair for accurate gene editing: an inclusive review of small molecules and modified CRISPR-Cas9 systems. Bioimpacts. 2022;12(4):371–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Werner CT, Gancarz AM, Dietz DM. Mechanisms Regulating Compulsive Drug Behaviors. Neural Mechanisms of Addiction2019. p. 137–55.
- 58.Yang J, Corces VG. Chromatin insulators: a role in nuclear organization and gene expression. Adv Cancer Res. 2011;110:43–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Watowich MB, Gilbert MR, Larion M. T cell exhaustion in malignant gliomas. Trends Cancer. 2023;9(4):270–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Cao Q, Tian Y, Deng Z, Yang F, Chen E. Epigenetic alteration in colorectal cancer: potential diagnostic and prognostic implications. Int J Mol Sci. 2024;25(6):3358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Reis AHDO, Figalo LB, Orsini M, Lemos B. The implications of DNA methylation for amyotrophic lateral sclerosis. An Acad Bras Cienc. 2023;95(suppl 2):e20230277. [DOI] [PubMed] [Google Scholar]
- 62.Fuso A. The ‘golden age’of DNA methylation in neurodegenerative diseases. Clin Chem Lab Med. 2013;51(3):523–34. [DOI] [PubMed] [Google Scholar]
- 63.Morahan JM, Yu B, Trent RJ, Pamphlett R. A genome-wide analysis of brain DNA methylation identifies new candidate genes for sporadic amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2009;10(5–6):418–29. [DOI] [PubMed] [Google Scholar]
- 64.Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell. 1990;61(5):759–67. [DOI] [PubMed] [Google Scholar]
- 65.Xiao W, Zhao H, Dong W, Li Q, Zhu J, Li G, et al. Quantitative detection of methylated NDRG4 gene as a candidate biomarker for diagnosis of colorectal cancer. Oncol Lett. 2015;9(3):1383–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Laugsand EA, Brenne SS, Skorpen F. DNA methylation markers detected in blood, stool, urine, and tissue in colorectal cancer: a systematic review of paired samples. Int J Colorectal Dis. 2021;36:239–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.He Q, Chen H-Y, Bai E-Q, Luo Y-X, Fu R-J, He Y-S, et al. Development of a multiplex methyllight assay for the detection of multigene methylation in human colorectal cancer. Cancer Genet Cytogenet. 2010;202(1):1–10. [DOI] [PubMed] [Google Scholar]
- 68.Thakur C, Qiu Y, Fu Y, Bi Z, Zhang W, Ji H, et al. Epigenetics and environment in breast cancer: new paradigms for anti-cancer therapies. Front Oncol. 2022;12:971288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Esteller M, Silva JM, Dominguez G, Bonilla F, Matias-Guiu X, Lerma E, et al. Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors. J Natl Cancer Inst. 2000;92(7):564–9. [DOI] [PubMed] [Google Scholar]
- 70.Radpour R, Barekati Z, Kohler C, Schumacher MM, Grussenmeyer T, Jenoe P, et al. Integrated epigenetics of human breast cancer: synoptic investigation of targeted genes, microRNAs and proteins upon demethylation treatment. PLoS ONE. 2011;6(11): e27355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Fujikane T, Nishikawa N, Toyota M, Suzuki H, Nojima M, Maruyama R, et al. Genomic screening for genes upregulated by demethylation revealed novel targets of epigenetic silencing in breast cancer. Breast Cancer Res Treat. 2010;122:699–710. [DOI] [PubMed] [Google Scholar]
- 72.Shargh SA, Sakizli M, Khalaj V, Movafagh A, Yazdi H, Hagigatjou E, et al. Downregulation of E-cadherin expression in breast cancer by promoter hypermethylation and its relation with progression and prognosis of tumor. Med Oncol. 2014;31:1–6. [DOI] [PubMed] [Google Scholar]
- 73.Mirza S, Sharma G, Prasad CP, Parshad R, Srivastava A, Gupta SD, et al. Promoter hypermethylation of TMS1, BRCA1, ERα and PRB in serum and tumor DNA of invasive ductal breast carcinoma patients. Life Sci. 2007;81(4):280–7. [DOI] [PubMed] [Google Scholar]
- 74.Esteller M, Corn PG, Baylin SB, Herman JG. A gene hypermethylation profile of human cancer. Can Res. 2001;61(8):3225–9. [PubMed] [Google Scholar]
- 75.Askari M, Sobti RC, Nikbakht M, Sharma SC. Aberrant promoter hypermethylation of p21 (WAF1/CIP1) gene and its impact on expression and role of polymorphism in the risk of breast cancer. Mol Cell Biochem. 2013;382:19–26. [DOI] [PubMed] [Google Scholar]
- 76.Barski A, Cuddapah S, Cui K, Roh T-Y, Schones DE, Wang Z, et al. High-resolution profiling of histone methylations in the human genome. Cell. 2007;129(4):823–37. [DOI] [PubMed] [Google Scholar]
- 77.Messier TL, Gordon JA, Boyd JR, Tye CE, Browne G, Stein JL, et al. Histone H3 lysine 4 acetylation and methylation dynamics define breast cancer subtypes. Oncotarget. 2016;7(5):5094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Jørgensen S, Schotta G, Sørensen CS. Histone H4 lysine 20 methylation: key player in epigenetic regulation of genomic integrity. Nucleic Acids Res. 2013;41(5):2797–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Gan L, Yang Y, Li Q, Feng Y, Liu T, Guo W. Epigenetic regulation of cancer progression by EZH2: From biological insights to therapeutic potential [Internet]. Biomark. Res. BioMed Central Ltd 2018. [DOI] [PMC free article] [PubMed]
- 80.Alexandrova E, Salvati A, Pecoraro G, Lamberti J, Melone V, Sellitto A, et al. Histone methyltransferase DOT1L as a promising epigenetic target for treatment of solid tumors. Front Genet. 2022;13:864612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Zheng Y, Zeng Y, Qiu R, Liu R, Huang W, Hou Y, et al. The homeotic protein SIX3 suppresses carcinogenesis and metastasis through recruiting the LSD1/NuRD (MTA3) complex. Theranostics. 2018;8(4):972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Berry WL, Janknecht R. KDM4/JMJD2 histone demethylases: epigenetic regulators in cancer cells. Cancer Res. 2013;73(10):2936–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Shao P, Liu Q, Maina PK, Cui J, Bair TB, Li T, et al. Histone demethylase PHF8 promotes epithelial to mesenchymal transition and breast tumorigenesis. Nucleic Acids Res. 2017;45(4):1687–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Sierra-Ramirez JA, Seseña-Mendez E, Godinez-Victoria M, Hernandez-Caballero ME. An insight into the promoter methylation of PHF20L1 and the gene association with metastasis in breast cancer. Adv Clin Exp Med. 2021;30(5):507–15. [DOI] [PubMed] [Google Scholar]
- 85.Xu Q, Jiang Y, Yin Y, Li Q, He J, Jing Y, et al. A regulatory circuit of miR-148a/152 and DNMT1 in modulating cell transformation and tumor angiogenesis through IGF-IR and IRS1. J Mol Cell Biol. 2013;5(1):3–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Zhang Y, Yan L-X, Wu Q-N, Du Z-M, Chen J, Liao D-Z, et al. miR-125b is methylated and functions as a tumor suppressor by regulating the ETS1 proto-oncogene in human invasive breast cancer. Cancer Res. 2011;71(10):3552–62. [DOI] [PubMed] [Google Scholar]
- 87.Augoff K, McCue B, Plow EF, Sossey-Alaoui K. Mir-31 and its host gene lncRNA LOC554202 are regulated by promoter hypermethylation in triple-negative breast cancer. Mol Cancer. 2012;11(1):5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Zhang Y, Yang P, Sun T, Li D, Xu X, Rui Y, et al. MiR-126 and miR-126* repress recruitment of mesenchymal stem cells and inflammatory monocytes to inhibit breast cancer metastasis. Nat Cell Biol. 2013;15(3):284–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Lehmann U, Hasemeier B, Christgen M, Müller M, Römermann D, Länger F, et al. Epigenetic inactivation of microRNA gene hsa-mir-9-1 in human breast cancer. J Pathol. 2008;214(1):17–24. [DOI] [PubMed] [Google Scholar]
- 90.Aviles D, Warshal D, Buchbinder M, Ostrovsky O. Influence of aberrant epigenetic changes and the tumor microenvironment in ovarian cancer metastasis. Exon Publications. 2022:77–89. [PubMed]
- 91.Natanzon Y, Goode EL, Cunningham JM, editors. Epigenetics in ovarian cancer. Seminars in cancer biology; 2018: Elsevier. [DOI] [PMC free article] [PubMed]
- 92.Kanwal R, Gupta S. Epigenetic modifications in cancer. Clin Genet. 2012;81(4):303–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Sugiura M, Sato H, Kanesaka M, Imamura Y, Sakamoto S, Ichikawa T, et al. Epigenetic modifications in prostate cancer. Int J Urol. 2021;28(2):140–9. [DOI] [PubMed] [Google Scholar]
- 94.Yegnasubramanian S, Haffner MC, Zhang Y, Gurel B, Cornish TC, Wu Z, et al. DNA hypomethylation arises later in prostate cancer progression than CpG island hypermethylation and contributes to metastatic tumor heterogeneity. Cancer Res. 2008;68(21):8954–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kim BW, Ryu J, Jeong YE, Kim J, Martin LJ. Human motor neurons with SOD1-G93A mutation generated from CRISPR/Cas9 gene-edited iPSCs develop pathological features of amyotrophic lateral sclerosis. Front Cell Neurosci. 2020;14:604171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Guan L, Han Y, Yang C, Lu S, Du J, Li H, et al. CRISPR-Cas9-mediated gene therapy in neurological disorders. Mol Neurobiol. 2022;59(2):968–82. [DOI] [PubMed] [Google Scholar]
- 97.Long C, Amoasii L, Bassel-Duby R, Olson EN. Genome editing of monogenic neuromuscular diseases: a systematic review. JAMA Neurol. 2016;73(11):1349–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Hu H, Lin H, Duan W, Cui C, Li Z, Liu Y, et al. Intrathecal injection of scAAV9-hIGF1 prolongs the survival of ALS model mice by inhibiting the NF-kB pathway. Neuroscience. 2018;381:1–10. [DOI] [PubMed] [Google Scholar]
- 99.Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell. 1990;61(5):759–67. [DOI] [PubMed] [Google Scholar]
- 100.Gunes S, Soykan MN, Sariboyaci AE, Uysal O, Sevimli TS. Enhancement of Apo2L/TRAIL signaling pathway receptors by the activation of Klotho gene with CRISPR/Cas9 in Caco-2 colon cancer cells. Med Oncol. 2021;38(12):146. [DOI] [PubMed] [Google Scholar]
- 101.Biagioni A, Chillà A, Del Rosso M, Fibbi G, Scavone F, Andreucci E, et al. CRISPR/Cas9 uPAR Gene Knockout Results in Tumor Growth Inhibition, EGFR Downregulation and Induction of Stemness Markers in Melanoma and Colon Carcinoma Cell Lines. Front Oncol. 2021;11:663225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Singh DD, Verma R, Tripathi SK, Sahu R, Trivedi P, Yadav DK. Breast cancer transcriptional regulatory network reprogramming by using the CRISPR/Cas9 system: an oncogenetics perspective. Curr Top Med Chem. 2021;21(31):2800–13. [DOI] [PubMed] [Google Scholar]
- 103.Zhang H, Qin C, An C, Zheng X, Wen S, Chen W, et al. Application of the CRISPR/Cas9-based gene editing technique in basic research, diagnosis, and therapy of cancer. Mol Cancer. 2021;20(1):126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Zhu W, Liu C, Lu T, Zhang Y, Zhang S, Chen Q, et al. Knockout of EGFL6 by CRISPR/Cas9 mediated inhibition of tumor angiogenesis in ovarian cancer. Front Oncol. 2020;10:1451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Cai Y, Tsai HC, Yen RC, Zhang YW, Kong X, Wang W, et al. Critical threshold levels of DNA methyltransferase 1 are required to maintain DNA methylation across the genome in human cancer cells. Genome Res. 2017;27(4):533–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Caobi A, Dutta RK, Garbinski LD, Esteban-Lopez M, Ceyhan Y, Andre M, et al. The impact of CRISPR-Cas9 on age-related disorders: from pathology to therapy. Aging Dis. 2020;11(4):895–915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.He ZY, Zhang YG, Yang YH, Ma CC, Wang P, Du W, et al. In vivo ovarian cancer gene therapy using CRISPR-Cas9. Hum Gene Ther. 2018;29(2):223–33. [DOI] [PubMed] [Google Scholar]
- 108.Filipchiuk C, Laganà AS, Beteli R, Ponce TG, Christofolini DM, Martins Trevisan C, et al. BIRC5/survivin expression as a non-invasive biomarker of endometriosis. Diagnostics. 2020. 10.3390/diagnostics10080533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Li D, Hu C, Li H. Survivin as a novel target protein for reducing the proliferation of cancer cells. Biomed Rep. 2018;8(5):399–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Zhao G, Wang Q, Wu Z, Tian X, Yan H, Wang B, et al. Ovarian primary and metastatic tumors suppressed by survivin knockout or a novel survivin inhibitor. Mol Cancer Ther. 2019;18(12):2233–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Zhao G, Wang Q, Gu Q, Qiang W, Wei JJ, Dong P, et al. Lentiviral CRISPR/Cas9 nickase vector mediated BIRC5 editing inhibits epithelial to mesenchymal transition in ovarian cancer cells. Oncotarget. 2017;8(55):94666–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Norouzi-Barough L, Sarookhani M, Salehi R, Sharifi M, Moghbelinejad S. CRISPR/Cas9, a new approach to successful knockdown of ABCB1/P-glycoprotein and reversal of chemosensitivity in human epithelial ovarian cancer cell line. Iran J Basic Med Sci. 2018;21(2):181–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Zhao M, Lei C, Yang Y, Bu X, Ma H, Gong H, et al. Abraxane, the nanoparticle formulation of paclitaxel can induce drug resistance by up-regulation of P-gp. PLoS ONE. 2015;10(7):e0131429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Wikström P, Stattin P, Franck-Lissbrant I, Damber JE, Bergh A. Transforming growth factor beta1 is associated with angiogenesis, metastasis, and poor clinical outcome in prostate cancer. Prostate. 1998;37(1):19–29. [DOI] [PubMed] [Google Scholar]
- 115.Hazafa A, Mumtaz M, Farooq MF, Bilal S, Chaudhry SN, Firdous M, et al. CRISPR/Cas9: a powerful genome editing technique for the treatment of cancer cells with present challenges and future directions. Life Sci. 2020;263:118525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Wei CG, Zhang R, Wei LY, Pan P, Zu H, Liu YZ, et al. Calcium phosphate-based nanomedicine mediated CRISPR/Cas9 delivery for prostate cancer therapy. Front Bioeng Biotechnol. 2022;10:1078342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Abati E, Bresolin N, Comi G, Corti S. Silence superoxide dismutase 1 (SOD1): a promising therapeutic target for amyotrophic lateral sclerosis (ALS). Expert Opin Ther Targets. 2020;24(4):295–310. [DOI] [PubMed] [Google Scholar]
- 118.De Plano LM, Calabrese G, Conoci S, Guglielmino SPP, Oddo S, Caccamo A. Applications of CRISPR-Cas9 in Alzheimer’s disease and related disorders. Int J Mol Sci. 2022;23(15):8714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Fang T, Je G, Pacut P, Keyhanian K, Gao J, Ghasemi M. Gene therapy in amyotrophic lateral sclerosis. Cells. 2022;11(13):2066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Guan L, Han Y, Yang C, Lu S, Du J, Li H, et al. CRISPR-Cas9-mediated gene therapy in neurological disorders. Mol Neurobiol. 2022;59(2):968–82. [DOI] [PubMed] [Google Scholar]
- 121.Hu H, Lin H, Duan W, Cui C, Li Z, Liu Y, et al. Intrathecal injection of scAAV9–hIGF1 prolongs the survival of ALS model mice by inhibiting the NF-kB pathway. Neuroscience. 2018;381:1–10. [DOI] [PubMed] [Google Scholar]
- 122.Karpe Y, Chen Z, Li X-J. Stem cell models and gene targeting for human motor neuron diseases. Pharmaceuticals (Basel). 2021;14(6):565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Long C, Amoasii L, Bassel-Duby R, Olson EN. Genome editing of monogenic neuromuscular diseases. JAMA Neurol. 2016;73(11):1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Filipi T, Hermanova Z, Tureckova J, Vanatko O, Anderova M. Glial cells—the strategic targets in amyotrophic lateral sclerosis treatment. J Clin Med. 2020;9(1):261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Sen T, Thummer RP. CRISPR and iPSCs: recent developments and future perspectives in neurodegenerative disease modelling, research, and therapeutics. Neurotox Res. 2022;40(5):1597–623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F. Genome engineering using the CRISPR-Cas9 system. Nat Protoc. 2013;8(11):2281–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Komor AC, Badran AH, Liu DR. CRISPR-based technologies for the manipulation of eukaryotic genomes. Cell. 2017;168(1–2):20–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Yu C, Luo D, Yu J, Zhang M, Zheng X, Xu G, et al. Genome-wide CRISPR-cas9 knockout screening identifies GRB7 as a driver for MEK inhibitor resistance in KRAS mutant colon cancer. Oncogene. 2022;41(2):191–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Goodarzi E, Beiranvand R, Naemi H, Momenabadi V, Khazaei Z. Worldwide incidence and mortality of colorectal cancer and human development index (HDI): an ecological study. World Cancer Res J. 2019;6:8. [Google Scholar]
- 130.Siegel RL, Torre LA, Soerjomataram I, Hayes RB, Bray F, Weber TK, et al. Global patterns and trends in colorectal cancer incidence in young adults. Gut. 2019;68(12):2179–85. [DOI] [PubMed] [Google Scholar]
- 131.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70(1):7–30. [DOI] [PubMed] [Google Scholar]
- 132.Rawla P, Sunkara T, Barsouk A. Epidemiology of colorectal cancer: incidence, mortality, survival, and risk factors. Gastroenterol Rev. 2019;14(2):89–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Li C, Wu M-Y, Liang Y-R, Wu X-Y. Correlation between expression of human telomerase subunits and telomerase activity in esophageal squamous cell carcinoma. World J Gastroenterol. 2003;9(11):2395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Nugent CI, Lundblad V. The telomerase reverse transcriptase: components and regulation. Genes Dev. 1998;12(8):1073–85. [DOI] [PubMed] [Google Scholar]
- 135.Nguyen THD, Tam J, Wu RA, Greber BJ, Toso D, Nogales E, et al. Cryo-EM structure of substrate-bound human telomerase holoenzyme. Nature. 2018;557(7704):190–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Cohen SB, Graham ME, Lovrecz GO, Bache N, Robinson PJ, Reddel RR. Protein composition of catalytically active human telomerase from immortal cells. Science. 2007;315(5820):1850–3. [DOI] [PubMed] [Google Scholar]
- 137.Korkut A, Zaidi S, Kanchi RS, Rao S, Gough NR, Schultz A, et al. A pan-cancer analysis reveals high-frequency genetic alterations in mediators of signaling by the TGF-β superfamily. Cell Syst. 2018;7(4):422-37.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Sporn MB, Roberts AB, Wakefield LM, Assoian RK. Transforming growth factor-β: biological function and chemical structure. Science. 1986;233(4763):532–4. [DOI] [PubMed] [Google Scholar]
- 139.Langenskiöld M, Holmdahl L, Falk P, Angenete E, Ivarsson ML. Increased TGF-beta1 protein expression in patients with advanced colorectal cancer. J Surg Oncol. 2008;97(5):409–15. [DOI] [PubMed] [Google Scholar]
- 140.Hasegawa Y, Takanashi S, Kanehira Y, Tsushima T, Imai T, Okumura K. Transforming growth factor-β1 level correlates with angiogenesis, tumor progression, and prognosis in patients with nonsmall cell lung carcinoma. Cancer. 2001;91(5):964–71. [PubMed] [Google Scholar]
- 141.Saito H, Tsujitani S, Oka S, Kondo A, Ikeguchi M, Maeta M, et al. The expression of transforming growth factor-β1 is significantly correlated with the expression of vascular endothelial growth factor and poor prognosis of patients with advanced gastric carcinoma. Cancer. 1999;86(8):1455–62. [DOI] [PubMed] [Google Scholar]
- 142.Wikström P, Stattin P, Franck-Lissbrant I, Damber JE, Bergh A. Transforming growth factor β1 is associated with angiogenesis, metastasis, and poor clinical outcome in prostate cancer. Prostate. 1998;37(1):19–29. [DOI] [PubMed] [Google Scholar]
- 143.Tsushima H, Kawata S, Tamura S, Ito N, Shirai Y, Kiso S, et al. High levels of transforming growth factor beta 1 in patients with colorectal cancer: association with disease progression. Gastroenterology. 1996;110(2):375–82. [DOI] [PubMed] [Google Scholar]
- 144.Rome L, Kedersha N, Chugani D. Unlocking vaults: organelles in search of a function. Trends Cell Biol. 1991;1(2–3):47–50. [DOI] [PubMed] [Google Scholar]
- 145.Kedersha NL, Miquel M-C, Bittner D, Rome LH. Vaults. II. Ribonucleoprotein structures are highly conserved among higher and lower eukaryotes. J Cell Biol. 1990;110(4):895–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Kedersha NL, Rome LH. Isolation and characterization of a novel ribonucleoprotein particle: large structures contain a single species of small RNA. J Cell Biol. 1986;103(3):699–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Wojtowicz K, Januchowski R, Nowicki M, Zabel M. vPARP adjusts MVP expression in drug-resistant cell lines in conjunction with MDR proteins. Anticancer Res. 2017;37(6):3015–23. [DOI] [PubMed] [Google Scholar]
- 148.Pietras P, Leśniczak-Staszak M, Kasprzak A, Andrzejewska M, Jopek K, Sowiński M, et al. MVP expression facilitates tumor cell proliferation and migration supporting the metastasis of colorectal cancer cells. Int J Mol Sci. 2021;22(22):12121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Feng X, Hua Z, Qian D, Zhou Q, Shi A, Wei W, et al. Cost-effectiveness and cost-benefit analysis of upper gastrointestinal cancer screening in Yangzhong City, Jiangsu Province, from 2009 to 2015. Zhonghua yu Fang yi xue za zhi [Chinese J Prevent Med]. 2019;53(11):1115–8. [DOI] [PubMed] [Google Scholar]
- 150.Wang F, Stewart JA, Kasbek C, Zhao Y, Wright WE, Price CM. Human CST has independent functions during telomere duplex replication and C-strand fill-in. Cell Rep. 2012;2(5):1096–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Feng X, Hsu S-J, Kasbek C, Chaiken M, Price CM. CTC1-mediated c-strand fill-in is an essential step in telomere length maintenance. Nucleic Acids Res. 2017;45(8):4281–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Feng X, Hsu S-J, Bhattacharjee A, Wang Y, Diao J, Price CM. CTC1-STN1 terminates telomerase while STN1-TEN1 enables C-strand synthesis during telomere replication in colon cancer cells. Nat Commun. 2018;9(1):2827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Ahmed M, Daoud GH, Mohamed A, Harati R. New insights into the therapeutic applications of CRISPR/Cas9 genome editing in breast cancer. Genes. 2021. 10.3390/genes12050723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Karn V, Sandhya S, Hsu W, Parashar D, Singh HN, Jha NK, et al. CRISPR/Cas9 system in breast cancer therapy: advancement, limitations and future scope. Cancer Cell Int. 2022;22(1):234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Janik E, Niemcewicz M, Ceremuga M, Krzowski L, Saluk-Bijak J, Bijak M. Various aspects of a gene editing system-CRISPR-Cas9. Int J Mol Sci. 2020. 10.3390/ijms21249604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Mitra S, Sarker J, Mojumder A, Shibbir TB, Das R, Emran TB, et al. Genome editing and cancer: how far has research moved forward on CRISPR/Cas9? Biomed Pharmacother. 2022;150:113011. [DOI] [PubMed] [Google Scholar]
- 157.Sabit H, Abdel-Ghany S, Tombuloglu H, Cevik E, Alqosaibi A, Almulhim F, et al. New insights on CRISPR/Cas9-based therapy for breast cancer. Genes Environ. 2021;43(1):15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Xiong S, Tan X, Wu X, Wan A, Zhang G, Wang C, et al. Molecular landscape and emerging therapeutic strategies in breast cancer brain metastasis. Ther Adv Med Oncol. 2023;15:17588359231165976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Gerlinger M, Rowan AJ, Horswell S, Math M, Larkin J, Endesfelder D, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366(10):883–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Lino CA, Harper JC, Carney JP, Timlin JA. Delivering CRISPR: a review of the challenges and approaches. Drug Deliv. 2018;25(1):1234–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Zhang H, Qin C, An C, Zheng X, Wen S, Chen W, et al. Application of the CRISPR/Cas9-based gene editing technique in basic research, diagnosis, and therapy of cancer. Mol Cancer. 2021;20:1–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Schuijers J, Manteiga JC, Weintraub AS, Day DS, Zamudio AV, Hnisz D, et al. Transcriptional dysregulation of MYC reveals common enhancer-docking mechanism. Cell Rep. 2018;23(2):349–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Yang H, Jaeger M, Walker A, Wei D, Leiker K, Weitao T. Break breast cancer addiction by CRISPR/Cas9 genome editing. J Cancer. 2018;9(2):219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Reid BM, Permuth JB, Sellers TA. Epidemiology of ovarian cancer: a review. Cancer Biol Med. 2017;14(1):9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Marliyya SZ. Risk factors for ovarian cancer. In: Ahmed L, Karine C-S, (eds.) Tumor progression and metastasis. Rijeka: IntechOpen; 2020;4.
- 166.Ahmed N, Stenvers KL. Getting to know ovarian cancer ascites: opportunities for targeted therapy-based translational research. Front Oncol. 2013;3: 256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Yang X, Zhang B. A review on CRISPR/Cas: a versatile tool for cancer screening, diagnosis, and clinic treatment. Funct Integr Genomics. 2023;23(2):182. [DOI] [PubMed] [Google Scholar]
- 168.Aoun Mehmood Sherazi S, Rafique F, Haris M, Arshad A, Qaiser H, Uzair M, et al. Applications of CRISPR Cas-9 in ovarian cancer research. Protein Pept Lett. 2023;30(8):653. [DOI] [PubMed] [Google Scholar]
- 169.Gavalas NG, Liontos M, Trachana SP, Bagratuni T, Arapinis C, Liacos C, et al. Angiogenesis-related pathways in the pathogenesis of ovarian cancer. Int J Mol Sci. 2013;14(8):15885–909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Zhu W, Liu C, Lu T, Zhang Y, Zhang S, Chen Q, et al. Knockout of EGFL6 by CRISPR/Cas9 mediated inhibition of tumor angiogenesis in ovarian cancer. Front Oncol. 2020;10:1451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Yeung G, Mulero JJ, Berntsen RP, Loeb DB, Drmanac R, Ford JE. Cloning of a novel epidermal growth factor repeat containing gene EGFL6: expressed in tumor and fetal tissues. Genomics. 1999;62(2):304–7. [DOI] [PubMed] [Google Scholar]
- 172.Noh K, Mangala LS, Han H-D, Zhang N, Pradeep S, Wu SY, et al. Differential effects of EGFL6 on tumor versus wound angiogenesis. Cell Rep. 2017;21(10):2785–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Cai Y, Tsai H-C, Yen R-WC, Zhang YW, Kong X, Wang W, et al. Critical threshold levels of DNA methyltransferase 1 are required to maintain DNA methylation across the genome in human cancer cells. Genome Res. 2017;27(4):533–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.He Z-Y, Zhang Y-G, Yang Y-H, Ma C-C, Wang P, Du W, et al. In vivo ovarian cancer gene therapy using CRISPR-Cas9. Hum Gene Ther. 2018;29(2):223–33. [DOI] [PubMed] [Google Scholar]
- 175.Filipchiuk C, Laganà AS, Beteli R, Ponce TG, Christofolini DM, Martins Trevisan C, et al. BIRC5/survivin expression as a non-invasive biomarker of endometriosis. Diagnostics. 2020;10(8):533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Li D, Hu C, Li H. Survivin as a novel target protein for reducing the proliferation of cancer cells. Biomed Rep. 2018;8(5):399–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Zhao G, Wang Q, Wu Z, Tian X, Yan H, Wang B, et al. Ovarian primary and metastatic tumors suppressed by survivin knockout or a novel survivin inhibitor. Mol Cancer Ther. 2019;18(12):2233–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Zhao G, Wang Q, Gu Q, Qiang W, Wei J-J, Dong P, et al. Lentiviral CRISPR/Cas9 nickase vector mediated BIRC5 editing inhibits epithelial to mesenchymal transition in ovarian cancer cells. Oncotarget. 2017;8(55):94666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Norouzi-Barough L, Sarookhani M, Salehi R, Sharifi M, Moghbelinejad S. CRISPR/Cas9, a new approach to successful knockdown of ABCB1/P-glycoprotein and reversal of chemosensitivity in human epithelial ovarian cancer cell line. Iran J Basic Med Sci. 2018;21(2):181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Ren F, Shen J, Shi H, Hornicek FJ, Kan Q, Duan Z. Novel mechanisms and approaches to overcome multidrug resistance in the treatment of ovarian cancer. Biochimica et Biophys Acta (BBA)-Rev Cancer. 2016;1866(2):266–75. [DOI] [PubMed] [Google Scholar]
- 181.Chang H, Yi B, Ma R, Zhang X, Zhao H, Xi Y. CRISPR/Cas9, a novel genomic tool to knock down microRNA in vitro and in vivo. Sci Rep. 2016;6(1):22312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Huo W, Zhao G, Yin J, Ouyang X, Wang Y, Yang C, et al. Lentiviral CRISPR/Cas9 vector mediated miR-21 gene editing inhibits the epithelial to mesenchymal transition in ovarian cancer cells. J Cancer. 2017;8(1):57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Chan JK, Blansit K, Kiet T, Sherman A, Wong G, Earle C, et al. The inhibition of miR-21 promotes apoptosis and chemosensitivity in ovarian cancer. Gynecol Oncol. 2014;132(3):739–44. [DOI] [PubMed] [Google Scholar]
- 184.Li L, Qiu C, Hou M, Wang X, Huang C, Zou J, et al. Ferroptosis in ovarian cancer: A novel therapeutic strategy. Front Oncol. 2021;11: 665945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Creeden JF, Nanavaty NS, Einloth KR, Gillman CE, Stanbery L, Hamouda DM, et al. Homologous recombination proficiency in ovarian and breast cancer patients. BMC Cancer. 2021;21(1):1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Moore K, Colombo N, Scambia G, Kim BG, Oaknin A, Friedlander M, et al. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med. 2018;379(26):2495–505. [DOI] [PubMed] [Google Scholar]
- 187.Dey G, Bharti R, Braley C, Alluri R, Esakov E, Crean-Tate K, et al. LCK facilitates DNA damage repair by stabilizing RAD51 and BRCA1 in the nucleus of chemoresistant ovarian cancer. J Ovarian Res. 2023;16(1):1–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Coelho R, Tozzi A, Disler M, Lombardo F, Fedier A, López MN, et al. Overlapping gene dependencies for PARP inhibitors and carboplatin response identified by functional CRISPR-Cas9 screening in ovarian cancer. Cell Death Dis. 2022;13(10):909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Kim SM, Yang Y, Oh SJ, Hong Y, Seo M, Jang M. Cancer-derived exosomes as a delivery platform of CRISPR/Cas9 confer cancer cell tropism-dependent targeting. J Control Release. 2017;266:8–16. [DOI] [PubMed] [Google Scholar]
- 190.Uddin F, Rudin CM, Sen T. CRISPR gene therapy: applications, limitations, and implications for the future. Front Oncol. 2020;10:1387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Rawla P. Epidemiology of prostate cancer. World J Oncol. 2019;10(2):63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Akram F, Haq IU, Sahreen S, Nasir N, Naseem W, Imitaz M, et al. CRISPR/Cas9: a revolutionary genome editing tool for human cancers treatment. Technol Cancer Res Treat. 2022;21:15330338221132078. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 193.Wei C, Wang F, Liu W, Zhao W, Yang Y, Li K, et al. CRISPR/Cas9 targeting of the androgen receptor suppresses the growth of LNCaP human prostate cancer cells. Mol Med Rep. 2018;17(2):2901–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Tang S, Sethunath V, Metaferia NY, Nogueira MF, Gallant DS, Garner ER, et al. A genome-scale CRISPR screen reveals PRMT1 as a critical regulator of androgen receptor signaling in prostate cancer. Cell Rep. 2022;38(8): 110417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Sorrentino C, D’Antonio L, Ciummo SL, Fieni C, Landuzzi L, Ruzzi F, et al. CRISPR/Cas9-mediated deletion of Interleukin-30 suppresses IGF1 and CXCL5 and boosts SOCS3 reducing prostate cancer growth and mortality. J Hematol Oncol. 2022;15(1):1–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Lee B, Gries K, Valimukhametova AR, McKinney RL, Gonzalez-Rodriguez R, Topkiran UC, et al. In vitro prostate cancer treatment via CRISPR-Cas9 gene editing facilitated by polyethyleneimine-derived graphene quantum dots. Adv Funct Mater. 2023. 10.1002/adfm.202305506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Gotama KT, Fahira A, Pandhita BAW. Targeted cellular delivery of CRISPR/Cas9 with RNA aptamer-cationic liposome as a potential therapy for metastatic prostate cancer. J Asian Med Students’ Assoc. 2019;7(1):28. [Google Scholar]
- 198.Zhen S, Takahashi Y, Narita S, Yang YC, Li X. Targeted delivery of CRISPR/Cas9 to prostate cancer by modified gRNA using a flexible aptamer-cationic liposome. Oncotarget. 2017;8(6):9375–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Poddar A, Pyreddy S, Carraro F, Dhakal S, Rassell A, Field MR, et al. ZIF-C for targeted RNA interference and CRISPR/Cas9 based gene editing in prostate cancer. Chem Commun (Camb). 2020;56(98):15406–9. [DOI] [PubMed] [Google Scholar]
- 200.Shojaei Baghini S, Gardanova ZR, Abadi SAH, Zaman BA, İlhan A, Shomali N, et al. CRISPR/Cas9 application in cancer therapy: a pioneering genome editing tool. Cell Mol Biol Lett. 2022;27(1):1–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Tsujino T, Takai T, Hinohara K, Gui F, Tsutsumi T, Bai X, et al. CRISPR screens reveal genetic determinants of PARP inhibitor sensitivity and resistance in prostate cancer. Nat Commun. 2023;14(1):252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Ipsen MB, Sørensen EMG, Thomsen EA, Weiss S, Haldrup J, Dalby A, et al. A genome-wide CRISPR-Cas9 knockout screen identifies novel PARP inhibitor resistance genes in prostate cancer. Oncogene. 2022;41(37):4271–81. [DOI] [PubMed] [Google Scholar]
- 203.Abdel-Megeed RM. CRISPR-Cas9 genome engineering as a novel prognostic tools for Prostate cancer: long non-coding RNAs the predictive diagnostic index of targeted liposomal-coated compounds in prostate cancer therapy. 2022 [DOI] [PMC free article] [PubMed]
- 204.Tsujino T, Komura K, Inamoto T, Azuma H. Crispr screen contributes to novel target discovery in prostate cancer. Int J Mol Sci. 2021;22(23):12777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Guo J, Huang X, Dou L, Yan M, Shen T, Tang W, et al. Aging and aging-related diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Target Ther. 2022;7(1):391. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No datasets were generated or analysed during the current study.




