Abstract
Background-
Transplantation of autologous mitochondria into ischemic tissue may mitigate injury caused by ischemia and reperfusion.
Methods-
Using murine stroke models of middle cerebral artery occlusion, we sought to evaluate feasibility of delivery of viable mitochondria to ischemic brain parenchyma. We evaluated the effects of concurrent focused ultrasound activation of microbubbles, which serves to open the blood-brain barrier, on efficacy of delivery of mitochondria.
Results-
Following intra-arterial delivery, mitochondria distribute through the stroked hemisphere and integrate into neural and glial cells in the brain parenchyma. Consistent with functional integration in the ischemic tissue, the transplanted mitochondria elevate concentration of adenosine triphosphate in the stroked hemisphere, reduce infarct volume and increase cell viability. Additional of focused ultrasound leads to improved blood brain barrier opening without hemorrhagic complications.
Conclusions-
Our results have implications for the development of interventional strategies after ischemic stroke and suggest a novel potential modality of therapy after mechanical thrombectomy.
Graphical Abstract
INTRODUCTION
Stroke is a leading cause of death and severe disability in the United States1. Thus far, clinical trials of neuroprotective agents have been unsuccessful, and studies preventing stroke-related injury in animal models have not translated well into clinical settings. Current treatment options for stroke patients are limited to restoring blood flow via intravenous administration of thrombolytics (tissue plasminogen activator, tPA) or endovascular thrombectomy 2, 3. However, reperfusion intervention itself has been shown to trigger secondary injury pathways in the ischemic tissue4. Therefore, new therapeutic strategies addressing the ischemic and reperfusion-induced injury are needed to improve post-stroke outcomes.
Mitochondrial function is fundamental for metabolic homeostasis in all multicellular eukaryotes. In the nervous system, mitochondria-generated adenosine triphosphate (ATP) is essential for establishing electrochemical gradients across the plasma membrane and reliable synaptic transmission. Within minutes after arterial occlusion, brain mitochondria begin to lose electrochemical proton gradients, causing cessation of ATP synthesis and overproduction of reactive oxygen species (ROS). Prolonged depletion of ATP leads to plasma membrane depolarization and eventually apoptotic cell death5, 6.
Mitochondrial transplantation as a therapeutic approach was first explored in cardiovascular disease. Pioneered by McCully et al.7–9, mitochondria were harvested from autologous skeletal muscle and transplanted through coronary vasculature into the ischemic zone of a rabbit’s heart, improving post-ischemic myocardial functional recovery 8. In a clinical trial, Emani et al. transplanted mitochondria into human pediatric patients who sustained a myocardial ischemic event following cardiac surgery and found promising preliminary results 10. Recent studies have also suggested that neurons can incorporate mitochondria released in the extracellular space by other cells, such as astrocytes 11. Mitochondrial re-uptake can preserve neuronal viability and improve recovery after stroke 11–13. Exploiting this mechanism, Zhang et al. demonstrated that transplantation of muscle-derived autologous mitochondria in the lateral ventricle of the stroked brain reduced infarct size 14. In a more recent study, placenta-derived mitochondria delivered intravenously were shown to decrease infarct size after transient focal ischemia in mice 13.
While the evidence for beneficial effects of mitochondrial supplementation after ischemia is mounting, optimal routes of delivery to affected brain tissue have not been investigated in detail.
METHODS
Details of the methods used in this manuscript can be found in the supplemental materials available online.
Mouse model establishment
All experiments were approved by the Institutional Animal Care and Use Committee of the University of Virginia. Adult male or female C57BL/6J mice were purchased from the Jackson Laboratory (Bar Harbor, Maine, USA). All mice were housed in a controlled environment on 12 hours light/dark cycles and fed a standard chow. Only adult animals (8 to 10 weeks) were used in this study and animals from different cages were selected for each experimental group to ensure randomization. Littermates of the same sex were randomly assigned in a 1:1 ratio to experimental groups. All analysis was pooled, and sex was not used for sub-analysis of data.
Mitochondrial isolation and staining
Mitochondrial isolation was performed as previously described 18, and summarized in the supplemental methods.
Intra-arterial delivery of mitochondria
One hour after MCA occlusion, the mice were anesthetized and set up for reperfusion as described previously. Immediately prior to the withdrawal of the occluding filament, a micro-clip was placed on the ICA artery to avoid retrograde bleeding. The existing arteriotomy in the CCA was then used to insert an intra-arterial catheter (SAI infusion technologies, REF: MAC-01) filled with mitochondria or buffer solution. The catheter was secured in the CCA with a silk tie. Using a connected Hamilton syringe, slow infusion of 200 μl of the fresh mitochondrial suspension was performed at a rate of 100 μl per minute over two minutes. The catheter was then removed from the ICA and the CCA was permanently tied. The skin was closed, and analgesics were provided as above.
Stereotactic delivery of mitochondria
Mice were anaesthetized with isoflurane; the scalp was shaved and the animal was positioned in a stereotaxic frame (Kopf Instruments). Using aseptic techniques, a midline incision was made with small surgical scissors. The skin was removed and bregma and lambda were exposed. A small burr hole was drilled over the target area with a hand-held drill. A 5 μl Hamilton syringe filled with mitochondrial suspension was attached to the stereotaxic apparatus moved to the stereotactic target in the striatum. Two μl of mitochondria suspension were slowly deposited using Hamilton syringe. The skin was sutured, the animal was treated with analgesics and allowed to recover in a heated cage.
RESULTS
Rapid isolation of viable mitochondria
Mitochondria were isolated from the gastrocnemius muscle of C57Bl/6 mice using the method of Preble et al. 18, and their morphology and viability was assessed with electron microscopy, flow cytometry and bioluminometric ATP assays. Transmission electron microscopy confirmed organelle integrity, presence of cristae in the matrix and the mean area of isolated mitochondria (Figure 1A). Next, isolated mitochondria were labeled with fluorescent dye (MitoTracker Red CMXRos, Thermo Fisher), and their size was analyzed with flow cytometry. The peak size of isolated mitochondria was smaller than the 6 μm-diameter reference beads (Figure 1B), in line with previously reported dimensions and with the measurements we performed for our isolated mitochondria (Figure 1A) 21. In addition, MitoTracker CMXRos accumulation is dependent upon membrane potential 22, therefore bright fluorescence observed in our preparations indicated sustained electrochemical proton gradients in these mitochondria (Figure 1C). To follow the fate of isolated mitochondria in the ischemic brain independent of synthetic dye labeling, in a subset of experiments we took advantage of a transgenic line in which mitochondria are genetically labelled with a DsRed variant termed MitoTimer 17. Using fluorescence microscopy, mitochondria isolated from these mice appeared bright red, with a similar size distribution as MitoTracker-labelled organelles (Figure 1D). Finally, we evaluated the ATP content in isolated mitochondria to further confirm they were metabolically active. The mean ATP concentration in the fresh mitochondrial suspension was 0.20 ± 0.04 pM, while no ATP was detectable in solution buffer alone (Figure 1E).
Intra-arterially infused mitochondria can permeate a disrupted blood-brain barrier
To determine how mitochondria distribute in the brain following intra-arterial infusion, with or without ultrasound treatment, we performed a series of experiments in healthy and ischemic brains. C57Bl/6 mice were subjected to transient proximal middle cerebral artery occlusion (tpMCAo) by inserting an occluding filament in the common carotid artery (CCA), as previously described 23. After 60 min, the ischemic hemisphere was re-perfused by withdrawing the occluding filament, and fresh mitochondrial suspension was infused through the same arteriotomy in the CCA, while the control mice received mitochondrial infusion after exposing the CCA without occlusion. To test whether we could increase permeability of the blood-brain barrier, we sonicated intra-arterially injected MBs with pulsed focused ultrasound in the ischemic brain immediately prior to mitochondrial infusion. Finally, a direct stereotactic injection in the ischemic brain was also performed as a control group. In all cases, the animals were sacrificed 2 hours after mitochondrial transplantation and brain sections were analyzed with confocal microscopy.
To measure delivery of mitochondria, we quantified the density of mitochondria signal in the vessels and in the parenchyma of comparable areas in dorsolateral striatum (Figure 2). We used CD31 staining to identify vessels to calculate intravascular density and used the area outside the vessels to define parenchymal density (Figure 2A, 2B, 2C, 2D). A parenchymal-to-intravascular signal density ratio was calculated as a measure of mitochondrial extravasation (Figure 2E). Mitochondrial infusion in the normal brains with intact blood-brain barrier resulted in low extravasation, and the ratio of parenchymal to intravascular mitochondrial signal was 0.33 ± 0.09 (Figure 2A). In the brains subjected to tpMCAo, this ratio increased to 6.96 ± 4.50, consistent with partial disruption of the blood-brain barrier (Figure 2C). However, this increase was not statistically significant. In another set of mice subjected to tpMCAo, blood-brain barrier permeability was further increased by stereotactic FUS (see Methods). In these animals, infused mitochondria reached a much higher concentration in the extravascular parenchyma; the parenchymal to intravascular ratio was 20.20 ± 7.57 (Figure 2E). Finally, for comparison, we performed direct stereotactic injections of mitochondrial suspension in the striatum. This resulted in robust delivery of mitochondria along the site of delivery but showed limited diffusion beyond the injection perimeter (Figure 2G).
To further characterize the synergistic effects of ischemic stroke and FUS on permeabilization of the blood-brain barrier, we performed intra-arterial injections of Evans blue dye for each experimental paradigm. The animals were sacrificed 4 hours after injection and the brains were stained with 2,3,5-triphenyltetrazolium chloride (TTC) to delineate infarcted tissue (Figure S1A–S1D). Evans blue signal was quantified using a flatbed scanner, analyzed in ImageJ and expressed volumetrically. In the control brains, the mean volume of Evans blue extravasation was 0.048 ± 0.083 mm3. Application of stereotactic FUS to normal brains significantly increased Evans blue extravasation (7.914 ± 1.888 mm3). As expected, tpMCAo also substantially elevated the volume of Evans blue extravasation, to 10.64 ± 4.729 mm3. However, when we applied stereotactically guided FUS to brains post-tpMCAo, the volume of extravasated dye increased dramatically to 38.45 ± 16.30 mm3, significantly higher than either of the treatments alone (p < 0.05) (Figure S1E). This data reiterates that ischemic injury weakens the blood-brain barrier and that additional permeabilization can be achieved by activated MBs with pulsed FUS in the ischemic region.
Limited FUS does not cause hemorrhage in post-stroke brain
Given the significant improvement in delivery of mitochondria with the use of FUS, we asked if the use of FUS during acute ischemia could cause any additional injury, such as hemorrhage. First, we obtained T1- and T2-weighted Gd-DTPA-enhanced MRI scans in 4 mice undergoing acute phase of ischemic tpMCAo stroke. Next, we applied MRI-guided FUS delivery to 3 different focal points distributed over cortical and striatal areas in the ischemic side of the brain (Figure 3A–3C). Following FUS exposure, all animals were re-imaged using an analogous Gd-DTPA MRI sequence. Also, after FUS we acquired gradient-echo (GRE) T2*-weighted MRI sequences to detect brain hemorrhage 20. Evaluation of pre- and post-FUS T1 MRI sequences demonstrated the anticipated enhancement of mean grayscale intensity value over the areas targeted with FUS (pre-FUS, 72.30 ± 17.19; post-FUS, 112.5 ± 35.35; p = 0.0229) (Figure 3D). Hence, the results of MRI imaging afford additional support for synergistic effects of ischemia and focused ultrasound on focal permeabilization of the blood-brain barrier. Also, the GRE sequences did not detect any cerebral macro-hemorrhages in these mice (Figure 3E). Finally, histological staining was used to inspect for microhemorrhages. We serially sectioned infarcted regions of all brains receiving 3 targeted FUS deliveries and found no indication of vascular damage or bleeding (Figure 3F). Thus, limited use of focused ultrasound can be a safe and effective enhancement for mitochondrial delivery in the stroked brain.
Neurons and other cells in the brain incorporate infused mitochondria
To follow the fate of mitochondria after extravasation, we studied coronal sections from mitochondria-infused brains with immunohistochemistry and confocal imaging. Specific antibodies for neurons (anti-NeuN, anti-MAP2), astrocytes (anti-GFAP) and microglia (anti-Iba1) were used to colocalize mitochondrial fluorescence signal with these cell types in the brain parenchyma. Mitochondria were seen in the extracellular space and vascular endothelia. However, under all tested conditions with compromised blood-brain barrier, transplanted mitochondria were also found in close apposition, as well as incorporated into all investigated cell types (Figure 4). We did not notice any preferred integration among the cell types investigated. To further investigate the mitochondria incorporation to cells, we analyzed 728 neurons in the penumbra area of 4 stroked brains using Z stacks and quantified the percentage of neuron’s co-localization with mitochondria (Figure S2A). We found that 85% of neurons, in the penumbra area, incorporated the transplanted mitochondria. To strengthen the evidence of intracellular localization, we also performed 3D volume rendering of the confocal Z-stacks with the Imaris software package. We have identified fluorescent puncta, consistent with mitochondria, which had been internalized inside the cytoplasm of the brain cells, including neurons (Figure S2B and C).
Because MitoTracker is a synthetic dye which can possibly dissociate from mitochondria under certain conditions, and inadvertently label endogenous mitochondria, we also used genetically labeled mitochondria expressing a DsRed variant MitoTimer to corroborate our results. Unlike MitoTracker, which is a membrane-bound synthetic dye that remains significantly fluorescent even in fragmented and degraded mitochondria, MitoTimer is a soluble mitochondrial matrix protein. If MitoTimer leaks out from the damaged mitochondria, the protein becomes diluted in the cytosol and degraded by the proteasome. Alternatively, if a mitochondrion is degraded via mitophagy, the MitoTimer protein is completely degraded by the lysosomal enzymes. We have previously shown that all MitoTimer signals are Cox4 positive, i.e. of mitochondrial origin 16, supporting the claim that MitoTimer is a reliable marker of intact mitochondria (Figure S3).
Transplanted mitochondria increase ATP concentration in the stroked hemisphere
We evaluated for metabolic rescue by testing whether mitochondrial infusion augmented ATP concentrations in the brain (Figure 5). Using the paradigm of tpMCAo, we induced stroke in C57Bl/6 mice and delivered freshly isolated mitochondria. Two hours later, we flash-froze the brains and determined total ATP concentrations per hemisphere with bioluminometry. A predicted drop in endogenous ATP levels was measured in the ischemic hemi-dissected brains (from 10 ± 6 pmol/kg to 3 ± 2 pmol/kg) (Figure 5A). FUS-enhanced intra-arterial infusion as well as direct stereotactic injection of mitochondrial suspension increased total ATP to control levels. Surprisingly, the highest total ATP levels were reached after intra-arterial infusion without FUS facilitation (40 ± 6 pmol/kg), significantly greater than other groups (Figure 5A). Thus, mitochondrial transfusions appear to improve baseline ATP content in the infarcted brain, providing exogenous energy support to the ischemic tissue.
Mitochondrial transplantation improves cell viability and reduces infarct size
To address whether mitochondrial transplantation decreases infarct size, we used flow cytometry to investigate the basis of infarct size reduction at a single-cell level. Mice received tpMCAo and subsequent intraarterial infusion of freshly isolated mitochondria. Two hours after the procedures, the brains were dissociated to single-cell suspensions, stained for cell viability using Live/Dead-Aqua (Invitrogen) and analyzed with flow cytometry. The gating strategy and robustness of viability-based discrimination is shown in Figure 5B. We found that the percentage of viable cells in cell suspensions from ischemic brains decreased from 68.40 ± 1.70% to 32.47 ± 13.95% (p = 0.04, n = 3). Intra-arterial infusion mitochondria alone significantly rescued cell viability in the ischemic brains, (56.63 ± 1.30%), although remaining below the normal brain values (Figure 5C).
Finally, we used a permanent distal middle cerebral artery occlusion (pdMCAo) stroke model, which creates a more reproducible ischemic injury than transient proximal occlusion. After distal MCA electrocoagulation (see Methods), fresh mitochondrial suspension was infused through catheterized ipsilateral common carotid artery in the experimental mice, while the control group received the suspending buffer only. Twenty-four hours after infusion, the brains were extracted, sliced, and stained for infarct evaluation (Figure 5D and 5E). Importantly, mitochondrial infusions brought about a 34% reduction of the infarct volume (9.919 ± 2.154 mm3 to 6.523 ± 1.659 mm3, p = 0.012, n = 6) (Figure 5F).
DISCUSSION
We provide evidence that mitochondrial transplantation may be beneficial as therapeutic intervention for ischemic stroke. First, we show that healthy muscle-derived mitochondria can be isolated in a clinically relevant timescale. Second, we demonstrate that intra-arterial injections of mitochondria result in perfusion of extravascular spaces in a minimally invasive manner, and that this outflow can we further enhanced by FUS. Third, mitochondria from the interstitial spaces become integrated by multiple cell types in the ischemic region. Fourth, mitochondrial transplantations elevate ATP concentrations in the brain parenchyma which might provide a basis for a partial metabolic rescue of ischemic tissue. Finally, cerebral infarction volume is reduced, and cell survival is increased after mitochondrial infusions.
Mitochondrial dysfunction is a major contributor to neurological disease 24, 25. Mitochondria are particularly critical to neuronal survival during ischemic stroke and supplementing mitochondrial function could reverse cell death and protect the ischemic penumbra from further injury. Given the narrow treatment window for ischemic stroke has few medical options, autologous mitochondrial transplantation offers an attractive paradigm for therapeutic intervention in cerebral infarction.
With respect to cerebral ischemic injury, Zhang et al. transplanted mitochondria by stereotactic injections in the lateral ventricle of stroked rat brains 14. Their data revealed reduction in brain infarct volume, increased neurogenesis and reversed neurological deficits. The study also found reduced astrogliosis, cellular oxidative stress and apoptosis. While these results are very encouraging, direct intracerebroventricular injections into lateral ventricles are inherently invasive, which would add additional peri-procedural time and risks to stroke patients. Furthermore, lateral ventricles occupy only about 1% of the whole adult human brain 29, rendering this route of delivery less favorable from a clinical perspective. Furthermore, we have previously noted that stereotactic intraparenchymal injection is unlikely to be useful in the setting of stroke as the range of delivery is limited compared to intra-arterial delivery 25.
Our results favor the intra-arterial approach to mitochondrial transplantations. Whereas the normal blood-brain barrier is not highly permeable to infused mitochondria, the endothelial barrier coincidentally opens during the acute phase of ischemic stroke (Figure 2, Figure 4, Figure S1, S2, S3.). These results are also supported by a recent study that showed benefit with intravenous delivery of mitochondria after stroke 13. The advantage of arterial delivery over the intravenous approach includes potentially higher concentrations of mitochondria delivered specifically to the reperfused brain, and less non-specific delivery and diffusion into other tissues. Either way, delivery via blood vessels ensures homogeneous distribution of labeled mitochondria in the parenchyma, clearly superior to stereotactic injections (Figure 2). The intra-arterial route also robustly supplements brain tissue with exogenous ATP (Figure 5A) which may assist with metabolic recovery in the ischemic areas.
Our data also indicate that even greater permeabilization of the ischemic blood-brain barrier can be attained with judicious use of FUS. In the laboratory setting, blood-brain barrier permeability is consistently increased through mechanical effects of FUS on injected microbubbles 31–34. Utilizing FUS, we were able to achieve enhanced delivery of macromolecules to the distal ischemic penumbra (Figure 2, Figure S1). Achieving enhanced delivery through use of focused ultrasound seems appealing and we show that effective, yet safe and hemorrhage-free conditions can be established in an animal stroke model (Figure 3). However, we acknowledge that our functional tests yielded lower overall ATP levels and cell viability after FUS-assisted intra-arterial infusions (Figure 5A and 5C); ultimately, further investigation of this inconsistency is warranted, but we speculate that the sonication associated with the FUS-assisted delivery may result in the decrease in ATP levels and cellular viability.
Molecular mechanisms responsible for incorporating extracellular mitochondria in the surviving cells of ischemic penumbra are unclear. Several processes have been proposed to mediate the internalization, including endocytosis, formation of nanotubes, gap junctions or connexin bridges 35. A recent study found that mitochondrial transfer from healthy cells to injured neurons occurs naturally in vivo 11. Ischemia-induced release of mitochondria from astrocytes involves CD38-cADPR pathway and mitochondrial transfer into neurons appear to be dependent on integrin-mediated Src/Syk signaling 11. However, other cell types in the central nervous system have been shown to be able to integrate mitochondria from the extracellular space, including astrocytes 11, 36, microglia and endothelial cells 37. With two different mitochondrial labeling strategies, using synthetic and genetic fluorophores, we have observed mitochondria integrating in several different cell types with no obvious preference (Figure 4 and Figure S3).
The precise mechanism by which infusion of mitochondria mediates a benefit is not definitively addressed by our study. There could be other paracrine effects of mitochondria that are contributing. For example, mitochondrial constituents could be modulating inflammation or other protective cascades 38. It is noteworthy, however, that Ramirez-Barbieri and co-authors investigated the immune and damage-associated molecular patterns (DAMPs)-associated response after injections of allogeneic mitochondria in a transplant rejection system of fully MHC-mismatched skin allografts, and found no direct or indirect, acute or chronic alloreactivity or DAMPs associated reactions to single or serial injections of allogeneic mitochondria 39. The interested reader is pointed to our recent review (Norat et al. Ref 25) on potential mechanisms of mitochondrial-based neuroprotection. There remain main outstanding questions as to how these cellular powerhouses are responsible for the phenotypic changes noted after injury.
This study, as many others have done in the past, pooled results from male and female subjects. The role of sex differences in recovery from injury is an area of intense discourse, with notable examples of differences after traumatic brain injury, for example. The pooling of results is a potential limitation of this study.
We propose that autologous mitochondrial transplantation is extremely well-poised to complement and augment the revascularization treatment for maximum benefit to stroke patients. While the role for FUS in enhancing delivery of mitochondria in this context remains unclear, we do show that safe and hemorrhage-free conditions for FUS can be established in an animal stroke model.
Supplementary Material
Clinical Perspective.
The authors present a novel strategy of autologous mitochondrial harvest and transplantation after thrombectomy for large vessel occlusion using a murine model of cerebral ischemia and reperfusion that mimics current clinical paradigms.
They demonstrate that mitochondria can be harvested in a clinically viable time and that they can be delivered to the brain and integrate into the parenchyma. Upon integration, the mitochondria elevate concentration of ATP, reduce infarct volume, and increase cell viability.
This novel strategy lends itself to clinical translation because the cellular components are autologously harvested and minimally modified.
ACKNOWLEDGEMENTS
We gratefully acknowledge resources at the Center for Brain Immunology and Glia (BIG) at the University of Virginia School of Medicine. We thank G. Wilson Miller and William Garrison for assistance with MRI imaging and Stacey Criswell for help with transmission electron microscopy. We also thank Kalil Alves de Lima and Aminata Coulibaly for advice and helpful suggestions on flow cytometry. Image analysis support by Daniel Miranda (Bitplane) is acknowledged. This study was supported by National Institutes of Health (Grants R01EB020147 and R21EB024323 to R.J.P.). C.M.G. is supported by the American Heart Association Fellowship (18PRE34030022). M.R.L. is supported by R01NS105692 and P.T. is supported by R21NS116431.
Abbreviations:
- FUS
Focused Ultrasound
- MB
Microbubbles
- TTC
2,3,5-Triphenyltetrazolium chloride
- MCA
Middle Cerebral Artery
- tpMCAo
Transient proximal middle cerebral artery occlusion
- pdMCAo
Permanent distal middle cerebral artery occlusion
Footnotes
DECLARATION OF INTERESTS AND DISCLOSURES
The authors declare no competing financial interests or disclosures.
References:
- 1.Benjamin EJ, Muntner P, Alonso A, Bittencourt MS, Callaway CW, Carson AP, Chamberlain AM, Chang AR, Cheng S, Das SR, et al. Heart disease and stroke statistics-2019 update: A report from the american heart association. Circulation. 2019;139:e56–e528 [DOI] [PubMed] [Google Scholar]
- 2.Bansal S, Sangha KS, Khatri P. Drug treatment of acute ischemic stroke. Am J Cardiovasc Drugs. 2013;13:57–69 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Powers WJ, Rabinstein AA, Ackerson T, Adeoye OM, Bambakidis NC, Becker K, Biller J, Brown M, Demaerschalk BM, Hoh B, et al. 2018 guidelines for the early management of patients with acute ischemic stroke: A guideline for healthcare professionals from the american heart association/american stroke association. Stroke. 2018;49:e46–e110 [DOI] [PubMed] [Google Scholar]
- 4.Eltzschig HK, Eckle T. Ischemia and reperfusion--from mechanism to translation. Nat Med. 2011;17:1391–1401 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Hofmeijer J, van Putten MJ. Ischemic cerebral damage: An appraisal of synaptic failure. Stroke. 2012;43:607–615 [DOI] [PubMed] [Google Scholar]
- 6.Lee JM, Grabb MC, Zipfel GJ, Choi DW. Brain tissue responses to ischemia. J Clin Invest. 2000;106:723–731 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Cowan DB, Yao R, Akurathi V, Snay ER, Thedsanamoorthy JK, Zurakowski D, Ericsson M, Friehs I, Wu Y, Levitsky S, et al. Intracoronary delivery of mitochondria to the ischemic heart for cardioprotection. PLoS One. 2016;11:e0160889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.McCully JD, Cowan DB, Pacak CA, Toumpoulis IK, Dayalan H, Levitsky S. Injection of isolated mitochondria during early reperfusion for cardioprotection. Am J Physiol Heart Circ Physiol. 2009;296:H94–H105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.McCully JD, Levitsky S, Del Nido PJ, Cowan DB. Mitochondrial transplantation for therapeutic use. Clin Transl Med. 2016;5:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Emani SM, Piekarski BL, Harrild D, Del Nido PJ, McCully JD. Autologous mitochondrial transplantation for dysfunction after ischemia-reperfusion injury. J Thorac Cardiovasc Surg. 2017;154:286–289 [DOI] [PubMed] [Google Scholar]
- 11.Hayakawa K, Esposito E, Wang X, Terasaki Y, Liu Y, Xing C, Ji X, Lo EH. Transfer of mitochondria from astrocytes to neurons after stroke. Nature. 2016;535:551–555 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Hayakawa K, Bruzzese M, Chou SH, Ning M, Ji X, Lo EH. Extracellular mitochondria for therapy and diagnosis in acute central nervous system injury. JAMA Neurol. 2018;75:119–122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Nakamura Y, Lo EH, Hayakawa K. Placental mitochondria therapy for cerebral ischemia-reperfusion injury in mice. Stroke. 2020;51:3142–3146 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zhang Z, Ma Z, Yan C, Pu K, Wu M, Bai J, Li Y, Wang Q. Muscle-derived autologous mitochondrial transplantation: A novel strategy for treating cerebral ischemic injury. Behav Brain Res. 2019;356:322–331 [DOI] [PubMed] [Google Scholar]
- 15.Al Amir Dache Z, Otandault A, Tanos R, Pastor B, Meddeb R, Sanchez C, Arena G, Lasorsa L, Bennett A, Grange T, et al. Blood contains circulating cell-free respiratory competent mitochondria. FASEB J. 2020;34:3616–3630 [DOI] [PubMed] [Google Scholar]
- 16.Laker RC, Xu P, Ryall KA, Sujkowski A, Kenwood BM, Chain KH, Zhang M, Royal MA, Hoehn KL, Driscoll M, et al. A novel mitotimer reporter gene for mitochondrial content, structure, stress, and damage in vivo. J Biol Chem. 2014;289:12005–12015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wilson RJ, Drake JC, Cui D, Zhang M, Perry HM, Kashatus JA, Kusminski CM, Scherer PE, Kashatus DF, Okusa MD, et al. Conditional mitotimer reporter mice for assessment of mitochondrial structure, oxidative stress, and mitophagy. Mitochondrion. 2019;44:20–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Preble JM, Pacak CA, Kondo H, MacKay AA, Cowan DB, McCully JD. Rapid isolation and purification of mitochondria for transplantation by tissue dissociation and differential filtration. J Vis Exp. 2014:e51682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Benedek A, Moricz K, Juranyi Z, Gigler G, Levay G, Harsing LG Jr., Matyus P, Szenasi G, Albert M Use of ttc staining for the evaluation of tissue injury in the early phases of reperfusion after focal cerebral ischemia in rats. Brain Res. 2006;1116:159–165 [DOI] [PubMed] [Google Scholar]
- 20.Lin DD, Filippi CG, Steever AB, Zimmerman RD. Detection of intracranial hemorrhage: Comparison between gradient-echo images and b(0) images obtained from diffusion-weighted echo-planar sequences. AJNR Am J Neuroradiol. 2001;22:1275–1281 [PMC free article] [PubMed] [Google Scholar]
- 21.Picard M, White K, Turnbull DM. Mitochondrial morphology, topology, and membrane interactions in skeletal muscle: A quantitative three-dimensional electron microscopy study. J Appl Physiol (1985). 2013;114:161–171 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Poot M, Zhang YZ, Kramer JA, Wells KS, Jones LJ, Hanzel DK, Lugade AG, Singer VL, Haugland RP. Analysis of mitochondrial morphology and function with novel fixable fluorescent stains. J Histochem Cytochem. 1996;44:1363–1372 [DOI] [PubMed] [Google Scholar]
- 23.Morris GP, Wright AL, Tan RP, Gladbach A, Ittner LM, Vissel B. A comparative study of variables influencing ischemic injury in the longa and koizumi methods of intraluminal filament middle cerebral artery occlusion in mice. PLoS One. 2016;11:e0148503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Lin MT, Beal MF. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature. 2006;443:787–795 [DOI] [PubMed] [Google Scholar]
- 25.Norat P, Soldozy S, Sokolowski JD, Gorick CM, Kumar JS, Chae Y, Yagmurlu K, Prada F, Walker M, Levitt MR, et al. Mitochondrial dysfunction in neurological disorders: Exploring mitochondrial transplantation. NPJ Regen Med. 2020;5:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Masuzawa A, Black KM, Pacak CA, Ericsson M, Barnett RJ, Drumm C, Seth P, Bloch DB, Levitsky S, Cowan DB, et al. Transplantation of autologously derived mitochondria protects the heart from ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 2013;304:H966–982 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Lin HC, Liu SY, Lai HS, Lai IR. Isolated mitochondria infusion mitigates ischemia-reperfusion injury of the liver in rats. Shock. 2013;39:304–310 [DOI] [PubMed] [Google Scholar]
- 28.Zhu L, Zhang J, Zhou J, Lu Y, Huang S, Xiao R, Yu X, Zeng X, Liu B, Liu F, et al. Mitochondrial transplantation attenuates hypoxic pulmonary hypertension. Oncotarget. 2016;7:48925–48940 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Coletti AM, Singh D, Kumar S, Shafin TN, Briody PJ, Babbitt BF, Pan D, Norton ES, Brown EC, Kahle KT, et al. Characterization of the ventricular-subventricular stem cell niche during human brain development. Development. 2018;145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ghanouni P, Pauly KB, Elias WJ, Henderson J, Sheehan J, Monteith S, Wintermark M. Transcranial mri-guided focused ultrasound: A review of the technologic and neurologic applications. AJR Am J Roentgenol. 2015;205:150–159 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Curley CT, Sheybani ND, Bullock TN, Price RJ. Focused ultrasound immunotherapy for central nervous system pathologies: Challenges and opportunities. Theranostics. 2017;7:3608–3623 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mead BP, Mastorakos P, Suk JS, Klibanov AL, Hanes J, Price RJ. Targeted gene transfer to the brain via the delivery of brain-penetrating DNA nanoparticles with focused ultrasound. J Control Release. 2016;223:109–117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Nance E, Timbie K, Miller GW, Song J, Louttit C, Klibanov AL, Shih TY, Swaminathan G, Tamargo RJ, Woodworth GF, et al. Non-invasive delivery of stealth, brain-penetrating nanoparticles across the blood-brain barrier using mri-guided focused ultrasound. J Control Release. 2014;189:123–132 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Timbie KF, Mead BP, Price RJ. Drug and gene delivery across the blood-brain barrier with focused ultrasound. J Control Release. 2015;219:61–75 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Gollihue JL, Rabchevsky AG. Prospects for therapeutic mitochondrial transplantation. Mitochondrion. 2017;35:70–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Davis CH, Kim KY, Bushong EA, Mills EA, Boassa D, Shih T, Kinebuchi M, Phan S, Zhou Y, Bihlmeyer NA, et al. Transcellular degradation of axonal mitochondria. Proc Natl Acad Sci U S A. 2014;111:9633–9638 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Gollihue JL, Patel SP, Mashburn C, Eldahan KC, Sullivan PG, Rabchevsky AG. Optimization of mitochondrial isolation techniques for intraspinal transplantation procedures. J Neurosci Methods. 2017;287:1–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Miliotis S, Nicolalde B, Ortega M, Yepez J, Caicedo A. Forms of extracellular mitochondria and their impact in health. Mitochondrion. 2019;48:16–30 [DOI] [PubMed] [Google Scholar]
- 39.Ramirez-Barbieri G, Moskowitzova K, Shin B, Blitzer D, Orfany A, Guariento A, Iken K, Friehs I, Zurakowski D, Del Nido PJ, et al. Alloreactivity and allorecognition of syngeneic and allogeneic mitochondria. Mitochondrion. 2019;46:103–115 [DOI] [PubMed] [Google Scholar]
- 40.McCully JD, Cowan DB, Emani SM, Del Nido PJ. Mitochondrial transplantation: From animal models to clinical use in humans. Mitochondrion. 2017;34:127–134 [DOI] [PubMed] [Google Scholar]
- 41.Hasan TF, Todnem N, Gopal N, Miller DA, Sandhu SS, Huang JF, Tawk RG. Endovascular thrombectomy for acute ischemic stroke. Curr Cardiol Rep. 2019;21:112. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.