Mitochondrial dysfunction in neurological disorders: Exploring mitochondrial transplantation

Pedro Norat, Sauson Soldozy, Jennifer D Sokolowski, Catherine M Gorick, Jeyan S Kumar, Youngrok Chae, Kaan Yağmurlu, Francesco Prada, Melanie Walker, Michael R Levitt, Richard J Price, Petr Tvrdik, M Yashar S Kalani, Pedro Norat, Sauson Soldozy, Jennifer D Sokolowski, Catherine M Gorick, Jeyan S Kumar, Youngrok Chae, Kaan Yağmurlu, Francesco Prada, Melanie Walker, Michael R Levitt, Richard J Price, Petr Tvrdik, M Yashar S Kalani

Abstract

Mitochondria are fundamental for metabolic homeostasis in all multicellular eukaryotes. In the nervous system, mitochondria-generated adenosine triphosphate (ATP) is required to establish appropriate electrochemical gradients and reliable synaptic transmission. Notably, several mitochondrial defects have been identified in central nervous system disorders. Membrane leakage and electrolyte imbalances, pro-apoptotic pathway activation, and mitophagy are among the mechanisms implicated in the pathogenesis of neurodegenerative diseases, such as Alzheimer's, Parkinson's, and Huntington's disease, as well as ischemic stroke. In this review, we summarize mitochondrial pathways that contribute to disease progression. Further, we discuss pathological states that damaged mitochondria impose on normal nervous system processes and explore new therapeutic approaches to mitochondrial diseases.

Conflict of interest statement

M.R.L. owns stocks of eLoupes, Inc., and Cerebrotech Medical Systems, Inc. The rest of the authors declare that there are no competing interests.

Figures

Fig. 1. Mitochondrial damage contributes to a…
Fig. 1. Mitochondrial damage contributes to a wide range of pathologies.
The main pathways leading to mitochondria-associated cellular dysfunction include (1) calcium overload in the matrix and mPTP pore opening, (2) cytochrome c release and activation of apoptosis, and (3) defects in mitochondrial fission and fusion affecting mitochondrial longevity. APAF1 apoptotic protease activating factor 1, ATP adenosine triphosphate, CypD cyclophilin D, cytc cytochrome c, Drp1 dynamin-related protein 1, IAPs inhibitor of apoptosis proteins, Mfn mitofusin, mPTP mitochondrial permeability transition pore, OPA optic atrophy mitochondrial fusion protein, ROS reactive oxygen species, Smac/DIABLO pro-apoptotic mitochondrial protein.
Fig. 2. Stereotactic injection facilitates efficient delivery…
Fig. 2. Stereotactic injection facilitates efficient delivery of mitochondria into the brain parenchyma.
Injected brains were harvested, cryo-sectioned and stained with nuclear stain DAPI (blue). ac Control brain injected with PBS, df Experimental brain injected with exogenous mitochondria labeled with MitoTracker Red CMXRos (Invitrogen). Both mice were injected directly into the striatum and striatal sections were imaged with confocal microscopy. The images were acquired at various distances from the injection site: a, d 200 μm, b, e 400 μm, and c, f 800 μm. Mitochondria were found to diffuse in the parenchyma and detected over 1 mm from the injection site. Scale bar, 25 μm.
Fig. 3. Intra-arterial injection can deliver mitochondria…
Fig. 3. Intra-arterial injection can deliver mitochondria in the ischemic brain parenchyma.
Mice were stroked with middle cerebral artery occlusion, injected intraarterially and their brains were cryo-sectioned and stained with DAPI (blue). a and b Stroked brain injected with exogenous mitochondria pre-labeled with MitoTracker (red), c Stroked brain injected with PBS. Both animals were injected in the common carotid artery following middle cerebral artery occlusion. Images were acquired in the striatum with confocal microscopy. Mitochondria crossed the blood–brain barrier and localized throughout the parenchyma. Occasionally, mitochondria were found deposited in the blood vessels (white arrow). b Higher magnification of a mitochondrial deposit shown in a. a Scale bar, 50 μm. b, c Scale bar, 25 μm.

References

    1. Frey TG, Mannella CA. The internal structure of mitochondria. Trends Biochem. Sci. 2000;25:319–324.
    1. Picard M, Taivassalo T, Gouspillou G, Hepple RT. Mitochondria: isolation, structure and function. J. Physiol. 2011;589:4413–4421.
    1. Rose J, et al. Mitochondrial dysfunction in glial cells: Implications for neuronal homeostasis and survival. Toxicology. 2017;391:109–115.
    1. Liu F, Lu J, Manaenko A, Tang J, Hu Q. Mitochondria in ischemic stroke: new insight and implications. Aging Dis. 2018;9:924–937.
    1. Briston T, et al. Mitochondrial permeability transition pore: sensitivity to opening and mechanistic dependence on substrate availability. Sci. Rep. 2017;7:10492.
    1. Halestrap AP, Richardson AP. The mitochondrial permeability transition: a current perspective on its identity and role in ischaemia/reperfusion injury. J. Mol. Cell. Cardiol. 2015;78:129–141.
    1. Mylonas C, Kouretas D. Lipid peroxidation and tissue damage. In Vivo. 1999;13:295–309.
    1. de Vasconcelos NM, Van Opdenbosch N, Van Gorp H, Parthoens E, Lamkanfi M. Single-cell analysis of pyroptosis dynamics reveals conserved GSDMD-mediated subcellular events that precede plasma membrane rupture. Cell Death Differ. 2019;26:146–161.
    1. Dixon SJ, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149:1060–1072.
    1. Fatokun AA, Dawson VL, Dawson TM. Parthanatos: mitochondrial-linked mechanisms and therapeutic opportunities. Br. J. Pharm. 2014;171:2000–2016.
    1. Gao M, et al. Role of mitochondria in ferroptosis. Mol. Cell. 2019;73:354–363 e353.
    1. Marshall KD, Baines CP. Necroptosis: is there a role for mitochondria? Front. Physiol. 2014;5:323.
    1. DeGregorio-Rocasolano N, Marti-Sistac O, Gasull T. Deciphering the iron side of stroke: neurodegeneration at the crossroads between iron dyshomeostasis, excitotoxicity, and ferroptosis. Front. Neurosci. 2019;13:85.
    1. Do Van B, et al. Ferroptosis, a newly characterized form of cell death in Parkinson’s disease that is regulated by PKC. Neurobiol. Dis. 2016;94:169–178.
    1. Lane DJR, Ayton S, Bush AI. Iron and Alzheimer’s disease: an update on emerging mechanisms. J. Alzheimers Dis. 2018;64:S379–S395.
    1. Xie BS, et al. Inhibition of ferroptosis attenuates tissue damage and improves long-term outcomes after traumatic brain injury in mice. CNS Neurosci. Ther. 2019;25:465–475.
    1. Lai Y, et al. STYK1/NOK correlates with ferroptosis in non-small cell lung carcinoma. Biochem. Biophys. Res. Commun. 2019;519:659–666.
    1. DeHart DN, et al. Opening of voltage dependent anion channels promotes reactive oxygen species generation, mitochondrial dysfunction and cell death in cancer cells. Biochem. Pharm. 2018;148:155–162.
    1. Krainz T, et al. A mitochondrial-targeted nitroxide is a potent inhibitor of ferroptosis. ACS Cent. Sci. 2016;2:653–659.
    1. Yang WS, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156:317–331.
    1. Azevedo RSS, et al. In situ immune response and mechanisms of cell damage in central nervous system of fatal cases microcephaly by Zika virus. Sci. Rep. 2018;8:1.
    1. Slee EA, Adrain C, Martin SJ. Executioner caspase-3, -6, and -7 perform distinct, non-redundant roles during the demolition phase of apoptosis. J. Biol. Chem. 2001;276:7320–7326.
    1. van der Bliek AM, Shen Q, Kawajiri S. Mechanisms of mitochondrial fission and fusion. Cold Spring Harb. Perspect. Biol. 2013;5:a011072.
    1. Meyer JN, Leuthner TC, Luz AL. Mitochondrial fusion, fission, and mitochondrial toxicity. Toxicology. 2017;391:42–53.
    1. Querfurth HW, LaFerla FM. Alzheimer’s disease. N. Engl. J. Med. 2010;362:329–344.
    1. Patergnani S, et al. Mitochondria in multiple sclerosis: molecular mechanisms of pathogenesis. Int. Rev. Cell Mol. Biol. 2017;328:49–103.
    1. Swerdlow RH, Khan SM. The Alzheimer’s disease mitochondrial cascade hypothesis: an update. Exp. Neurol. 2009;218:308–315.
    1. Du H, et al. Cyclophilin D deficiency attenuates mitochondrial and neuronal perturbation and ameliorates learning and memory in Alzheimer’s disease. Nat. Med. 2008;14:1097–1105.
    1. Rao VK, Carlson EA, Yan SS. Mitochondrial permeability transition pore is a potential drug target for neurodegeneration. Biochim. Biophys. Acta. 2014;1842:1267–1272.
    1. Du H, ShiDu Yan S. Unlocking the door to neuronal woes in Alzheimer’s disease: abeta and mitochondrial permeability transition pore. Pharmaceuticals. 2010;3:1936–1948.
    1. Tysnes OB, Storstein A. Epidemiology of Parkinson’s disease. J. Neural Transm. 2017;124:901–905.
    1. Winkler-Stuck K, et al. Re-evaluation of the dysfunction of mitochondrial respiratory chain in skeletal muscle of patients with Parkinson’s disease. J. Neural Transm. 2005;112:499–518.
    1. Gu M, Cooper JM, Taanman JW, Schapira AH. Mitochondrial DNA transmission of the mitochondrial defect in Parkinson’s disease. Ann. Neurol. 1998;44:177–186.
    1. Ekstrand MI, et al. Progressive parkinsonism in mice with respiratory-chain-deficient dopamine neurons. Proc. Natl Acad. Sci. USA. 2007;104:1325–1330.
    1. Klein C, Westenberger A. Genetics of Parkinson’s disease. Cold Spring Harb. Perspect. Med. 2012;2:a008888.
    1. Lucking CB, et al. Association between early-onset Parkinson’s disease and mutations in the parkin gene. N. Engl. J. Med. 2000;342:1560–1567.
    1. Gandhi S, et al. PINK1 protein in normal human brain and Parkinson’s disease. Brain. 2006;129:1720–1731.
    1. Wood-Kaczmar A, et al. PINK1 is necessary for long term survival and mitochondrial function in human dopaminergic neurons. PLoS ONE. 2008;3:e2455.
    1. Mallach A, et al. Post mortem examination of Parkinson’s disease brains suggests decline in mitochondrial biomass, reversed by deep brain stimulation of subthalamic nucleus. FASEB J. 2019;33:6957–6961.
    1. Quintanilla RA, Johnson GV. Role of mitochondrial dysfunction in the pathogenesis of Huntington’s disease. Brain Res. Bull. 2009;80:242–247.
    1. Lin J, et al. Defects in adaptive energy metabolism with CNS-linked hyperactivity in PGC-1alpha null mice. Cell. 2004;119:121–135.
    1. Milakovic T, Johnson GV. Mitochondrial respiration and ATP production are significantly impaired in striatal cells expressing mutant huntingtin. J. Biol. Chem. 2005;280:30773–30782.
    1. Choo YS, Johnson GV, MacDonald M, Detloff PJ, Lesort M. Mutant huntingtin directly increases susceptibility of mitochondria to the calcium-induced permeability transition and cytochrome c release. Hum. Mol. Genet. 2004;13:1407–1420.
    1. Tellez-Nagel I, Johnson AB, Terry RD. Studies on brain biopsies of patients with Huntington’s chorea. J. Neuropathol. Exp. Neurol. 1974;33:308–332.
    1. Song W, et al. Mutant huntingtin binds the mitochondrial fission GTPase dynamin-related protein-1 and increases its enzymatic activity. Nat. Med. 2011;17:377–382.
    1. Weydt P, et al. Thermoregulatory and metabolic defects in Huntington’s disease transgenic mice implicate PGC-1alpha in Huntington’s disease neurodegeneration. Cell Metab. 2006;4:349–362.
    1. Writing Group M, et al. Heart disease and stroke statistics-2016 update: a report from the American Heart Association. Circulation. 2016;133:e38–e360.
    1. Hofmeijer J, van Putten MJ. Ischemic cerebral damage: an appraisal of synaptic failure. Stroke. 2012;43:607–615.
    1. Dharmasaroja PA. Fluid intake related to brain edema in acute middle cerebral artery infarction. Transl. Stroke Res. 2016;7:49–53.
    1. Saita S, et al. Distinct types of protease systems are involved in homeostasis regulation of mitochondrial morphology via balanced fusion and fission. Genes Cells. 2016;21:408–424.
    1. Sims NR, Muyderman H. Mitochondria, oxidative metabolism and cell death in stroke. Biochim. Biophys. Acta. 2010;1802:80–91.
    1. Bereiter-Hahn J, Voth M. Dynamics of mitochondria in living cells: shape changes, dislocations, fusion, and fission of mitochondria. Microsc. Res. Tech. 1994;27:198–219.
    1. Wang J, et al. Mdivi-1 prevents apoptosis induced by ischemia-reperfusion injury in primary hippocampal cells via inhibition of reactive oxygen species-activated mitochondrial pathway. J. Stroke Cerebrovasc. Dis. 2014;23:1491–1499.
    1. Cowan DB, et al. Intracoronary delivery of mitochondria to the ischemic heart for cardioprotection. PLoS ONE. 2016;11:e0160889.
    1. McCully JD, et al. Injection of isolated mitochondria during early reperfusion for cardioprotection. Am. J. Physiol. Heart Circ. Physiol. 2009;296:H94–H105.
    1. McCully JD, Levitsky S, Del Nido PJ, Cowan DB. Mitochondrial transplantation for therapeutic use. Clin. Transl. Med. 2016;5:16.
    1. Masuzawa A, et al. Transplantation of autologously derived mitochondria protects the heart from ischemia-reperfusion injury. Am. J. Physiol. Heart Circ. Physiol. 2013;304:H966–H982.
    1. 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.
    1. Emani SM, McCully JD. Mitochondrial transplantation: applications for pediatric patients with congenital heart disease. Transl. Pediatr. 2018;7:169–175.
    1. Zhang Z, et al. Muscle-derived autologous mitochondrial transplantation: a novel strategy for treating cerebral ischemic injury. Behav. Brain Res. 2019;356:322–331.
    1. Liu K, et al. Mesenchymal stem cells rescue injured endothelial cells in an in vitro ischemia-reperfusion model via tunneling nanotube like structure-mediated mitochondrial transfer. Microvasc. Res. 2014;92:10–18.
    1. Pan BT, Johnstone RM. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor. Cell. 1983;33:967–978.
    1. Kramer-Albers EM, et al. Oligodendrocytes secrete exosomes containing major myelin and stress-protective proteins: trophic support for axons? Proteom. Clin. Appl. 2007;1:1446–1461.
    1. Simpson RJ, Jensen SS, Lim JW. Proteomic profiling of exosomes: current perspectives. Proteomics. 2008;8:4083–4099.
    1. Puhm F, et al. Mitochondria are a subset of extracellular vesicles released by activated monocytes and induce Type I IFN and TNF responses in endothelial cells. Circ. Res. 2019;125:43–52.
    1. Hayakawa K, et al. Transfer of mitochondria from astrocytes to neurons after stroke. Nature. 2016;535:551–555.
    1. Rustom A, Saffrich R, Markovic I, Walther P, Gerdes HH. Nanotubular highways for intercellular organelle transport. Science. 2004;303:1007–1010.
    1. Hayakawa K, et al. Protective effects of endothelial progenitor cell-derived extracellular mitochondria in brain endothelium. Stem Cells. 2018;36:1404–1410.
    1. Kaneko Y, et al. Cell therapy for stroke: emphasis on optimizing safety and efficacy profile of endothelial progenitor cells. Curr. Pharm. Des. 2012;18:3731–3734.
    1. Preble JM, et al. Rapid isolation and purification of mitochondria for transplantation by tissue dissociation and differential filtration. J. Vis. Exp. 2014;91:e51682.
    1. Poot M, et al. Analysis of mitochondrial morphology and function with novel fixable fluorescent stains. J. Histochem. Cytochem. 1996;44:1363–1372.
    1. Shi X, Zhao M, Fu C, Fu A. Intravenous administration of mitochondria for treating experimental Parkinson’s disease. Mitochondrion. 2017;34:91–100.
    1. Sun C, et al. Endocytosis-mediated mitochondrial transplantation: transferring normal human astrocytic mitochondria into glioma cells rescues aerobic respiration and enhances radiosensitivity. Theranostics. 2019;9:3595–3607.
    1. Lippert T, Borlongan CV. Prophylactic treatment of hyperbaric oxygen treatment mitigates inflammatory response via mitochondria transfer. CNS Neurosci. Ther. 2019;25:815–823.
    1. Joshi AU, et al. Fragmented mitochondria released from microglia trigger A1 astrocytic response and propagate inflammatory neurodegeneration. Nat. Neurosci. 2019;22:1635–1648.
    1. Robicsek O, et al. Isolated mitochondria transfer improves neuronal differentiation of schizophrenia-derived induced pluripotent stem cells and rescues deficits in a rat model of the disorder. Schizophr. Bull. 2018;44:432–442.
    1. Yao Y, et al. Connexin 43-mediated mitochondrial transfer of iPSC-MSCs alleviates asthma inflammation. Stem Cell Rep. 2018;11:1120–1135.
    1. Partikian A, Olveczky B, Swaminathan R, Li Y, Verkman AS. Rapid diffusion of green fluorescent protein in the mitochondrial matrix. J. Cell Biol. 1998;140:821–829.
    1. Gao L, Zhang Z, Lu J, Pei G. Mitochondria are dynamically transferring between human neural cells and Alexander disease-associated GFAP mutations impair the astrocytic transfer. Front. Cell. Neurosci. 2019;13:316.
    1. Gollihue JL, et al. Effects of mitochondrial transplantation on bioenergetics, cellular incorporation, and functional recovery after spinal cord injury. J. Neurotrauma. 2018;35:1800–1818.
    1. Kitani T, et al. Direct human mitochondrial transfer: a novel concept based on the endosymbiotic theory. Transpl. Proc. 2014;46:1233–1236.
    1. Rocca CJ, et al. Transplantation of wild-type mouse hematopoietic stem and progenitor cells ameliorates deficits in a mouse model of Friedreich’s ataxia. Sci. Transl. Med. 2017;9:eaaj2347.
    1. Laker RC, et al. A novel MitoTimer reporter gene for mitochondrial content, structure, stress, and damage in vivo. J. Biol. Chem. 2014;289:12005–12015.
    1. Wilson RJ, et al. Conditional MitoTimer reporter mice for assessment of mitochondrial structure, oxidative stress, and mitophagy. Mitochondrion. 2019;44:20–26.
    1. Gollihue JL, Rabchevsky AG. Prospects for therapeutic mitochondrial transplantation. Mitochondrion. 2017;35:70–79.
    1. Islam MN, et al. Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat. Med. 2012;18:759–765.
    1. Spees JL, Olson SD, Whitney MJ, Prockop DJ. Mitochondrial transfer between cells can rescue aerobic respiration. Proc. Natl Acad. Sci. USA. 2006;103:1283–1288.
    1. Paliwal S, Chaudhuri R, Agrawal A, Mohanty S. Regenerative abilities of mesenchymal stem cells through mitochondrial transfer. J. Biomed. Sci. 2018;25:31.
    1. Huang PJ, et al. Transferring xenogenic mitochondria provides neural protection against ischemic stress in ischemic rat brains. Cell Transpl. 2016;25:913–927.
    1. Hayakawa K, et al. Extracellular mitochondria for therapy and diagnosis in acute central nervous system injury. JAMA Neurol. 2018;75:119–122.
    1. Pletjushkina OY, et al. Effect of oxidative stress on dynamics of mitochondrial reticulum. Biochim. Biophys. Acta. 2006;1757:518–524.
    1. Chandel NS. Evolution of mitochondria as signaling organelles. Cell Metab. 2015;22:204–206.
    1. Nisoli E, Clementi E, Moncada S, Carruba MO. Mitochondrial biogenesis as a cellular signaling framework. Biochem. Pharm. 2004;67:1–15.
    1. Scholkmann F. Long range physical cell-to-cell signalling via mitochondria inside membrane nanotubes: a hypothesis. Theor. Biol. Med. Model. 2016;13:16.

Source: PubMed

3
Iratkozz fel