Mitochondria on the move: Horizontal mitochondrial transfer in disease and health

Mammalian genes were long thought to be constrained within somatic cells in most cell types. This concept was challenged recently when cellular organelles including mitochondria were shown to move between mammalian cells in culture via cytoplasmic bridges. Recent research in animals indicates transfer of mitochondria in cancer and during lung injury in vivo, with considerable functional consequences. Since these pioneering discoveries, many studies have confirmed horizontal mitochondrial transfer (HMT) in vivo, and its functional characteristics and consequences have been described. Additional support for this phenomenon has come from phylogenetic studies. Apparently, mitochondrial trafficking between cells occurs more frequently than previously thought and contributes to diverse processes including bioenergetic crosstalk and homeostasis, disease treatment and recovery, and development of resistance to cancer therapy. Here we highlight current knowledge of HMT between cells, focusing primarily on in vivo systems, and contend that this process is not only (patho)physiologically relevant, but also can be exploited for the design of novel therapeutic approaches.

[1]  Noa Sher,et al.  Mitochondrial augmentation of hematopoietic stem cells in children with single large-scale mitochondrial DNA deletion syndromes , 2022, Science Translational Medicine.

[2]  P. Puigserver,et al.  Mechanisms of mitochondrial respiratory adaptation , 2022, Nature Reviews Molecular Cell Biology.

[3]  Mikko T. Huuskonen,et al.  Neuron-astrocyte transmitophagy is altered in Alzheimer's disease , 2022, Neurobiology of Disease.

[4]  Edward T Chouchani,et al.  Why succinate? Physiological regulation by a mitochondrial coenzyme Q sentinel , 2022, Nature Chemical Biology.

[5]  M. Broekman,et al.  Advances in local therapy for glioblastoma — taking the fight to the tumour , 2022, Nature Reviews Neurology.

[6]  P. Carmeliet,et al.  Mitochondrial respiration supports autophagy to provide stress resistance during quiescence , 2022, Autophagy.

[7]  Jonathan R. Brestoff,et al.  Ejection of damaged mitochondria and their removal by macrophages ensure efficient thermogenesis in brown adipose tissue. , 2022, Cell metabolism.

[8]  S. Mitalipov,et al.  Horizontal mtDNA transfer between cells is common during mouse development , 2022, iScience.

[9]  Daniel A. Colón-Ramos,et al.  Multiview confocal super-resolution microscopy , 2021, Nature.

[10]  P. Majumder,et al.  Intercellular nanotubes mediate mitochondrial trafficking between cancer and immune cells , 2021, Nature Nanotechnology.

[11]  J. Norman,et al.  PINK1 drives production of mtDNA-containing extracellular vesicles to promote invasiveness , 2021, The Journal of cell biology.

[12]  H. Nagase,et al.  Intercellular transfer of mitochondrial DNA carrying metastasis-enhancing pathogenic mutations from high- to low-metastatic tumor cells and stromal cells via extracellular vesicles , 2021, BMC Molecular and Cell Biology.

[13]  J. Olefsky,et al.  Exosomes as mediators of intercellular crosstalk in metabolism. , 2021, Cell metabolism.

[14]  Shunbang Yu,et al.  Migrasome biogenesis and functions , 2021, The FEBS journal.

[15]  Y. Tabata,et al.  Iron oxide nanoparticles augment the intercellular mitochondrial transfer–mediated therapy , 2021, Science advances.

[16]  Yoon‐Kyoung Cho,et al.  Programmed exosome fusion for energy generation in living cells , 2021, Nature Catalysis.

[17]  U. Chandran,et al.  Microvesicles transfer mitochondria and increase mitochondrial function in brain endothelial cells. , 2021, Journal of controlled release : official journal of the Controlled Release Society.

[18]  K. Man,et al.  Automated Optical Tweezers Manipulation to Transfer Mitochondria from Fetal to Adult MSCs to Improve Antiaging Gene Expressions. , 2021, Small.

[19]  P. Fischer-Posovszky,et al.  Extracellular vesicle-based interorgan transport of mitochondria from energetically stressed adipocytes. , 2021, Cell metabolism.

[20]  J. Neuzil,et al.  Mitochondrial Function as Related to Psychological Distress in Health Care Professionals , 2021, Psychosomatic medicine.

[21]  L. Moro,et al.  Mitochondria Can Cross Cell Boundaries: An Overview of the Biological Relevance, Pathophysiological Implications and Therapeutic Perspectives of Intercellular Mitochondrial Transfer , 2021, International journal of molecular sciences.

[22]  K. Bhat,et al.  Tunneling nanotubes, TNT, communicate glioblastoma with surrounding non-tumor astrocytes to adapt them to hypoxic and metabolic tumor conditions , 2021, Scientific Reports.

[23]  R. Linden,et al.  Mitotherapy: Unraveling a Promising Treatment for Disorders of the Central Nervous System and Other Systemic Conditions , 2021, Cells.

[24]  F. Uher,et al.  Stromal Cells Serve Drug Resistance for Multiple Myeloma via Mitochondrial Transfer: A Study on Primary Myeloma and Stromal Cells , 2021, Cancers.

[25]  Robert W. Taylor,et al.  Mitochondrial disease in adults: recent advances and future promise , 2021, The Lancet Neurology.

[26]  J. Neuzil,et al.  Miro proteins connect mitochondrial function and intercellular transport , 2021, Critical reviews in biochemistry and molecular biology.

[27]  D. Shukla,et al.  Role of Tunneling Nanotubes in Viral Infection, Neurodegenerative Disease, and Cancer , 2021, Frontiers in Immunology.

[28]  Xiaoyu Hu,et al.  Mitocytosis, a migrasome-mediated mitochondrial quality-control process , 2021, Cell.

[29]  G. Pigino Intraflagellar transport , 2021, Current Biology.

[30]  Qionghai Dai,et al.  Iterative tomography with digital adaptive optics permits hour-long intravital observation of 3D subcellular dynamics at millisecond scale , 2021, Cell.

[31]  Qiong Zhou,et al.  The Functions, Methods, and Mobility of Mitochondrial Transfer Between Cells , 2021, Frontiers in Oncology.

[32]  A. Rodríguez-Sinovas,et al.  Connexins in the Heart: Regulation, Function and Involvement in Cardiac Disease , 2021, International journal of molecular sciences.

[33]  C. Zurzolo Tunneling nanotubes: Reshaping connectivity. , 2021, Current opinion in cell biology.

[34]  A. Rizvanov,et al.  Mitochondria Donation by Mesenchymal Stem Cells: Current Understanding and Mitochondria Transplantation Strategies , 2021, Frontiers in Cell and Developmental Biology.

[35]  R. Norris Transfer of mitochondria and endosomes between cells by gap junction internalization , 2021, Traffic.

[36]  P. Sonveaux,et al.  Mitochondrial Transfer in Cancer: A Comprehensive Review , 2021, International journal of molecular sciences.

[37]  T. Larsen,et al.  Mitochondrial Transfer Improves Cardiomyocyte Bioenergetics and Viability in Male Rats Exposed to Pregestational Diabetes , 2021, International journal of molecular sciences.

[38]  Changqing Zhang,et al.  Intercellular mitochondrial transfer as a means of tissue revitalization , 2021, Signal Transduction and Targeted Therapy.

[39]  M. Teitell,et al.  Stable transplantation of human mitochondrial DNA by high-throughput, pressurized isolated mitochondrial delivery , 2021, eLife.

[40]  M. Germain,et al.  Selective packaging of mitochondrial proteins into extracellular vesicles prevents the release of mitochondrial DAMPs , 2020, Nature Communications.

[41]  Cuifang Li,et al.  Immunoregulatory Effects of Mitochondria Transferred by Extracellular Vesicles , 2021, Frontiers in Immunology.

[42]  J. Neuzil,et al.  Platelets Facilitate the Wound-Healing Capability of Mesenchymal Stem Cells by Mitochondrial Transfer and Metabolic Reprogramming. , 2020, Cell metabolism.

[43]  J. Gerst,et al.  RNA transfer through tunneling nanotubes. , 2020, Biochemical Society transactions.

[44]  C. Zurzolo,et al.  The Ways of Actin: Why Tunneling Nanotubes Are Unique Cell Protrusions. , 2020, Trends in cell biology.

[45]  Sandeep P. Dumbali,et al.  Mitochondrial Transfer and Regulators of Mesenchymal Stromal Cell Function and Therapeutic Efficacy , 2020, Frontiers in Cell and Developmental Biology.

[46]  Jonathan R. Brestoff,et al.  Intercellular Mitochondria Transfer to Macrophages Regulates White Adipose Tissue Homeostasis and Is Impaired in Obesity. , 2020, Cell metabolism.

[47]  C. Toulas,et al.  Patient-derived glioblastoma stem cells transfer mitochondria through tunneling nanotubes in tumor organoids , 2020, bioRxiv.

[48]  Menna E. Jones,et al.  Evolution and lineage dynamics of a transmissible cancer in Tasmanian devils , 2020, PLoS biology.

[49]  P. Chinnery,et al.  Extreme heterogeneity of human mitochondrial DNA from organelles to populations , 2020, Nature reviews. Genetics.

[50]  R. Rahbarghazi,et al.  Mitochondrial donation in translational medicine; from imagination to reality , 2020, Journal of translational medicine.

[51]  K. Golan,et al.  Bone marrow regeneration requires mitochondrial transfer from donor Cx43-expressing hematopoietic progenitors to stroma. , 2020, Blood.

[52]  S. Priori,et al.  A Network of Macrophages Supports Mitochondrial Homeostasis in the Heart , 2020, Cell.

[53]  Li Yu,et al.  Chemical screening identifies ROCK1 as a regulator of migrasome formation , 2020, Cell Discovery.

[54]  T. Jin,et al.  Mesenchymal Stem/Stromal Cell-Mediated Mitochondrial Transfer and the Therapeutic Potential in Treatment of Neurological Diseases , 2020, Stem cells international.

[55]  P. Chinnery,et al.  Recurrent horizontal transfer identifies mitochondrial positive selection in a transmissible cancer , 2020, Nature Communications.

[56]  Anthony Scimè,et al.  Mitochondrial Function in Muscle Stem Cell Fates , 2020, Frontiers in Cell and Developmental Biology.

[57]  D. Ormond,et al.  Mitochondrial transfer from mesenchymal stem cells improves neuronal metabolism after oxidant injury in vitro: The role of Miro1 , 2020, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism.

[58]  N. Bresolin,et al.  The Role of Mitochondria in Neurodegenerative Diseases: the Lesson from Alzheimer’s Disease and Parkinson’s Disease , 2020, Molecular Neurobiology.

[59]  Hannah W. Miller,et al.  Biting Off What Can Be Chewed: Trogocytosis in Health, Infection, and Disease , 2020, Infection and Immunity.

[60]  S. Weinberg,et al.  Mitochondrial ubiquinol oxidation is necessary for tumor growth , 2020, Nature.

[61]  H. Chinnery,et al.  Tunneling Nanotubes and the Eye: Intercellular Communication and Implications for Ocular Health and Disease , 2020, BioMed research international.

[62]  Mirko H. H. Schmidt,et al.  Tunneling Nanotubes and Tumor Microtubes in Cancer , 2020, Cancers.

[63]  M. Minczuk,et al.  Therapeutic Manipulation of mtDNA Heteroplasmy: A Shifting Perspective. , 2020, Trends in molecular medicine.

[64]  A. Kheradvar,et al.  Bioenergetics Consequences of Mitochondrial Transplantation in Cardiomyocytes , 2020, Journal of the American Heart Association.

[65]  A. Kavelaars,et al.  Astrocytes rescue neuronal health after cisplatin treatment through mitochondrial transfer , 2020, Acta Neuropathologica Communications.

[66]  J. Neuzil,et al.  Dihydroorotate dehydrogenase in oxidative phosphorylation and cancer. , 2020, Biochimica et biophysica acta. Molecular basis of disease.

[67]  S. Diez,et al.  Mitochondria-adaptor TRAK1 promotes kinesin-1 driven transport in crowded environments , 2020, bioRxiv.

[68]  Zizhen Zhao,et al.  Improvement of cognitive and motor performance with mitotherapy in aged mice , 2020, International journal of biological sciences.

[69]  Peter M. Jones,et al.  Optimizing beta cell function through mesenchymal stromal cell‐mediated mitochondria transfer , 2020, Stem cells.

[70]  P. D. del Nido,et al.  Preischemic autologous mitochondrial transplantation by intracoronary injection for myocardial protection. , 2020, The Journal of thoracic and cardiovascular surgery.

[71]  Iain G. Johnston,et al.  Regulation of Mother-to-Offspring Transmission of mtDNA Heteroplasmy , 2019, Cell metabolism.

[72]  P. D. del Nido,et al.  Mitochondrial transplantation for myocardial protection in diabetic hearts. , 2019, European journal of cardio-thoracic surgery : official journal of the European Association for Cardio-thoracic Surgery.

[73]  F. Di Palma,et al.  ROS-mediated PI3K activation drives mitochondrial transfer from stromal cells to hematopoietic stem cells in response to infection , 2019, Proceedings of the National Academy of Sciences.

[74]  P. D. del Nido,et al.  Mitochondrial Transplantation Enhances Murine Lung Viability and Recovery after Ischemia Reperfusion Injury. , 2019, American journal of physiology. Lung cellular and molecular physiology.

[75]  Zizhen Zhao,et al.  Healthy mitochondria inhibit the metastatic melanoma in lungs , 2019, International journal of biological sciences.

[76]  Nidhi Sharma,et al.  Mitochondrial DNA: Epigenetics and environment , 2019, Environmental and molecular mutagenesis.

[77]  E. Sahai,et al.  Activated Stromal Cells Transfer Mitochondria to Rescue Acute Lymphoblastic Leukaemia Cells from Oxidative Stress. , 2019, Blood.

[78]  A. Suomalainen Mitochondrial DNA Inheritance in Humans: Mix, Match, and Survival of the Fittest. , 2019, Cell metabolism.

[79]  M. Kozlov,et al.  Migrasome formation is mediated by assembly of micron-scale tetraspanin macrodomains , 2019, Nature Cell Biology.

[80]  Xiaoxin Yin,et al.  Mitochondrial transplantation attenuates lipopolysaccharide- induced depression-like behaviors , 2019, Progress in Neuro-Psychopharmacology and Biological Psychiatry.

[81]  Zhen Zhang,et al.  Mitochondria Are Dynamically Transferring Between Human Neural Cells and Alexander Disease-Associated GFAP Mutations Impair the Astrocytic Transfer , 2019, Front. Cell. Neurosci..

[82]  Matthew A. Kayala,et al.  Mitochondrial Akt Signaling Modulated Reprogramming of Somatic Cells , 2019, Scientific Reports.

[83]  Michael Zhuo Wang,et al.  Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis , 2019, Cells.

[84]  Linyi Chen,et al.  Current progress of mitochondrial transplantation that promotes neuronal regeneration , 2019, Translational Neurodegeneration.

[85]  Hong Zhang,et al.  Endocytosis-mediated mitochondrial transplantation: Transferring normal human astrocytic mitochondria into glioma cells rescues aerobic respiration and enhances radiosensitivity , 2019, Theranostics.

[86]  David L. Bennett,et al.  Germline selection shapes human mitochondrial DNA diversity , 2019, Science.

[87]  Yonghuai Feng,et al.  Human Bone Marrow Mesenchymal Stem Cells Rescue Endothelial Cells Experiencing Chemotherapy Stress by Mitochondrial Transfer Via Tunneling Nanotubes. , 2019, Stem cells and development.

[88]  K. Bowles,et al.  CD38-Driven Mitochondrial Trafficking Promotes Bioenergetic Plasticity in Multiple Myeloma. , 2019, Cancer research.

[89]  Chuanzhu Yan,et al.  Mesenchymal stem cells transfer mitochondria into cerebral microvasculature and promote recovery from ischemic stroke. , 2019, Microvascular research.

[90]  H. Tse,et al.  Donation of mitochondria by iPSC-derived mesenchymal stem cells protects retinal ganglion cells against mitochondrial complex I defect-induced degeneration , 2019, Theranostics.

[91]  C. Borlongan,et al.  Prophylactic treatment of hyperbaric oxygen treatment mitigates inflammatory response via mitochondria transfer , 2019, CNS neuroscience & therapeutics.

[92]  E. Charpentier,et al.  Phase I trial of isatuximab monotherapy in the treatment of refractory multiple myeloma , 2019, Blood Cancer Journal.

[93]  M. Fujimiya,et al.  Mitochondria transfer from mesenchymal stem cells structurally and functionally repairs renal proximal tubular epithelial cells in diabetic nephropathy in vivo , 2019, Scientific Reports.

[94]  Zhong Chen,et al.  Mitochondrial transport serves as a mitochondrial quality control strategy in axons: Implications for central nervous system disorders , 2019, CNS neuroscience & therapeutics.

[95]  Xiaoming Zhang,et al.  Mitochondrial Transfer from Bone Marrow Mesenchymal Stem Cells to Motor Neurons in Spinal Cord Injury Rats via Gap Junction , 2019, Theranostics.

[96]  E. Giannoni,et al.  Cancer-associated fibroblasts promote prostate cancer malignancy via metabolic rewiring and mitochondrial transfer , 2019, bioRxiv.

[97]  Yaoqi Zhou,et al.  Reactivation of Dihydroorotate Dehydrogenase-Driven Pyrimidine Biosynthesis Restores Tumor Growth of Respiration-Deficient Cancer Cells. , 2019, Cell metabolism.

[98]  H. Sampath,et al.  Mitochondrial DNA Integrity: Role in Health and Disease , 2019, Cells.

[99]  F. Prósper,et al.  The Mechanism of Action of the Anti-CD38 Monoclonal Antibody Isatuximab in Multiple Myeloma , 2019, Clinical Cancer Research.

[100]  E. Schon,et al.  Mitochondria, OxPhos, and neurodegeneration: cells are not just running out of gas. , 2019, The Journal of clinical investigation.

[101]  P. D. del Nido,et al.  Mitochondrial transplantation prolongs cold ischemia time in murine heart transplantation. , 2019, The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation.

[102]  Zhi Ma,et al.  Muscle-derived autologous mitochondrial transplantation: A novel strategy for treating cerebral ischemic injury , 2019, Behavioural Brain Research.

[103]  S. Weinberg,et al.  Mitochondrial complex III is necessary for endothelial cell proliferation during angiogenesis , 2018, Nature Metabolism.

[104]  Iain G. Johnston,et al.  Mitochondrial Heterogeneity , 2018, Front. Genet..

[105]  I. Johnston,et al.  Mitochondrial Network Fragmentation Modulates Mutant mtDNA Accumulation Independently of Absolute Fission-Fusion Rates. , 2018, Genetics.

[106]  S. Benhamron,et al.  Mitochondrial Transfer Ameliorates Cognitive Deficits, Neuronal Loss, and Gliosis in Alzheimer's Disease Mice. , 2019, Journal of Alzheimer's disease : JAD.

[107]  C. Sommer,et al.  Dietary salt promotes ischemic brain injury and is associated with parenchymal migrasome formation , 2018, PloS one.

[108]  A. Kavelaars,et al.  Mitochondrial transfer from mesenchymal stem cells to neural stem cells protects against the neurotoxic effects of cisplatin , 2018, Acta neuropathologica communications.

[109]  J. Do,et al.  Mitochondrial Dynamics in Stem Cells and Differentiation , 2018, International journal of molecular sciences.

[110]  H. Tse,et al.  Connexin 43-Mediated Mitochondrial Transfer of iPSC-MSCs Alleviates Asthma Inflammation , 2018, Stem cell reports.

[111]  G. Pigino,et al.  The cryo-EM structure of intraflagellar transport trains reveals how dynein is inactivated to ensure unidirectional anterograde movement in cilia , 2018, Nature Cell Biology.

[112]  D. Turnbull,et al.  Mitochondrial donation: from test tube to clinic , 2018, The Lancet.

[113]  M. Berridge,et al.  Intercellular Communication in Tumor Biology: A Role for Mitochondrial Transfer , 2018, Front. Oncol..

[114]  R. Donahue,et al.  Effects of Mitochondrial Transplantation on Bioenergetics, Cellular Incorporation, and Functional Recovery after Spinal Cord Injury. , 2018, Journal of neurotrauma.

[115]  Emiri T. Mandeville,et al.  Protective Effects of Endothelial Progenitor Cell‐Derived Extracellular Mitochondria in Brain Endothelium , 2018, Stem cells.

[116]  M. Haigis,et al.  The multifaceted contributions of mitochondria to cellular metabolism , 2018, Nature Cell Biology.

[117]  T. Dokland,et al.  Exosomal transfer of mitochondria from airway myeloid-derived regulatory cells to T cells☆ , 2018, Redox biology.

[118]  Q. Gao,et al.  Dependence on the Pyrimidine Biosynthetic Enzyme DHODH Is a Synthetic Lethal Vulnerability in Mutant KRAS-Driven Cancers. , 2018, Cell chemical biology.

[119]  Xianchao Sun,et al.  Treatment of acetaminophen‐induced liver injury with exogenous mitochondria in mice , 2018, Translational research : the journal of laboratory and clinical medicine.

[120]  Xin Sun,et al.  Global DNA methylation synergistically regulates the nuclear and mitochondrial genomes in glioblastoma cells , 2018, Nucleic acids research.

[121]  S. Emani,et al.  Mitochondrial transplantation: applications for pediatric patients with congenital heart disease. , 2018, Translational pediatrics.

[122]  R. Guo,et al.  Intercellular transfer of mitochondria rescues virus-induced cell death but facilitates cell-to-cell spreading of porcine reproductive and respiratory syndrome virus. , 2018, Virology.

[123]  A. Agrawal,et al.  Regenerative abilities of mesenchymal stem cells through mitochondrial transfer , 2018, Journal of Biomedical Science.

[124]  G. Sukhikh,et al.  Miro1 Enhances Mitochondria Transfer from Multipotent Mesenchymal Stem Cells (MMSC) to Neural Cells and Improves the Efficacy of Cell Recovery , 2018, Molecules.

[125]  K. Bieńkowska-Szewczyk,et al.  Tunneling Nanotubes as a Novel Route of Cell-to-Cell Spread of Herpesviruses , 2018, Journal of Virology.

[126]  I. Weiner,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 , 2018, Schizophrenia bulletin.

[127]  Ross A. Poché,et al.  The mito::mKate2 mouse: A far‐red fluorescent reporter mouse line for tracking mitochondrial dynamics in vivo , 2018, Genesis.

[128]  A. Xiang,et al.  Cell adhesion-mediated mitochondria transfer contributes to mesenchymal stem cell-induced chemoresistance on T cell acute lymphoblastic leukemia cells , 2018, Journal of Hematology & Oncology.

[129]  L. Lerman,et al.  Renal scattered tubular-like cells confer protective effects in the stenotic murine kidney mediated by release of extracellular vesicles , 2018, Scientific Reports.

[130]  I. Adcock,et al.  Mesenchymal stem cells alleviate oxidative stress–induced mitochondrial dysfunction in the airways , 2017, The Journal of allergy and clinical immunology.

[131]  K. He,et al.  Mitochondria are transported along microtubules in membrane nanotubes to rescue distressed cardiomyocytes from apoptosis , 2018, Cell Death & Disease.

[132]  M. Ingelsson,et al.  Human Astrocytes Transfer Aggregated Alpha-Synuclein via Tunneling Nanotubes , 2017, The Journal of Neuroscience.

[133]  J. Peyron,et al.  Mitochondrial Transfer in the Leukemia Microenvironment. , 2017, Trends in cancer.

[134]  E. Eugenin,et al.  Tunneling nanotubes (TNT) mediate long-range gap junctional communication: Implications for HIV cell to cell spread , 2017, Scientific Reports.

[135]  E. K. Cunningham,et al.  Mesenchymal Stromal Cells Modulate Macrophages in Clinically Relevant Lung Injury Models by Extracellular Vesicle Mitochondrial Transfer , 2017, American journal of respiratory and critical care medicine.

[136]  T. Schwarz,et al.  Mitostasis in Neurons: Maintaining Mitochondria in an Extended Cellular Architecture , 2017, Neuron.

[137]  M. D. Den Boer,et al.  Tunneling Nanotubes and Gap Junctions–Their Role in Long-Range Intercellular Communication during Development, Health, and Disease Conditions , 2017, Front. Mol. Neurosci..

[138]  D. Edwards,et al.  NADPH oxidase-2 derived superoxide drives mitochondrial transfer from bone marrow stromal cells to leukemic blasts. , 2017, Blood.

[139]  E. Lo,et al.  Extracellular Mitochondria in Cerebrospinal Fluid and Neurological Recovery After Subarachnoid Hemorrhage , 2017, Stroke.

[140]  F. Cabrera,et al.  Artificial Mitochondria Transfer: Current Challenges, Advances, and Future Applications , 2017, Stem cells international.

[141]  Alexander Sasha Rabchevsky,et al.  Prospects for therapeutic mitochondrial transplantation. , 2017, Mitochondrion.

[142]  C. Jorgensen,et al.  Cell Connections by Tunneling Nanotubes: Effects of Mitochondrial Trafficking on Target Cell Metabolism, Homeostasis, and Response to Therapy , 2017, Stem cells international.

[143]  J. Dubois-Randé,et al.  Mesenchymal stem cells sense mitochondria released from damaged cells as danger signals to activate their rescue properties , 2017, Cell Death and Differentiation.

[144]  Ailing Fu,et al.  Mitotherapy for Fatty Liver by Intravenous Administration of Exogenous Mitochondria in Male Mice , 2017, Front. Pharmacol..

[145]  Shang-Der Chen,et al.  Mitochondrial Transfer from Wharton's Jelly Mesenchymal Stem Cell to MERRF Cybrid Reduces Oxidative Stress and Improves Mitochondrial Bioenergetics , 2017, Oxidative medicine and cellular longevity.

[146]  S. Emani,et al.  Mitochondrial transplantation: From animal models to clinical use in humans. , 2017, Mitochondrion.

[147]  P. D. del Nido,et al.  Myocardial rescue with autologous mitochondrial transplantation in a porcine model of ischemia/reperfusion , 2017, The Journal of thoracic and cardiovascular surgery.

[148]  Alissa M. Weaver,et al.  Extracellular Vesicles: Unique Intercellular Delivery Vehicles. , 2017, Trends in cell biology.

[149]  P. Stopka,et al.  Horizontal transfer of whole mitochondria restores tumorigenic potential in mitochondrial DNA-deficient cancer cells , 2017, eLife.

[150]  Hua Xu,et al.  Tunneling nanotubes promote intercellular mitochondria transfer followed by increased invasiveness in bladder cancer cells , 2017, Oncotarget.

[151]  D. Hall,et al.  C. elegans Neurons Jettison Protein Aggregates and Mitochondria Under Neurotoxic Stress , 2017, Nature.

[152]  E. Boyden,et al.  Influenza virus exploits tunneling nanotubes for cell-to-cell spread , 2017, Scientific Reports.

[153]  J. Neuzil,et al.  Exosome-derived microRNAs in cancer metabolism: possible implications in cancer diagnostics and therapy , 2017, Experimental & Molecular Medicine.

[154]  Joshua M. Weiss,et al.  Extracellular Vesicles in Cancer: Cell-to-Cell Mediators of Metastasis. , 2016, Cancer cell.

[155]  D. Freund,et al.  Tunneling nanotubes mediate the transfer of stem cell marker CD133 between hematopoietic progenitor cells. , 2016, Experimental hematology.

[156]  H. Tse,et al.  Mitochondrial transfer of mesenchymal stem cells effectively protects corneal epithelial cells from mitochondrial damage , 2016, Cell Death & Disease.

[157]  Erdun Bao,et al.  Mesenchymal Stromal Cells Derived Extracellular Vesicles Ameliorate Acute Renal Ischemia Reperfusion Injury by Inhibition of Mitochondrial Fission through miR-30 , 2016, Stem cells international.

[158]  J. Olivo-Marin,et al.  Tunneling nanotubes spread fibrillar α‐synuclein by intercellular trafficking of lysosomes , 2016, The EMBO journal.

[159]  F. Sánchez‐Madrid,et al.  Mitochondria Know No Boundaries: Mechanisms and Functions of Intercellular Mitochondrial Transfer , 2016, Front. Cell Dev. Biol..

[160]  Matthew G. Vander Heiden,et al.  Altered metabolite levels in cancer: implications for tumour biology and cancer therapy , 2016, Nature Reviews Cancer.

[161]  Larry A. Sklar,et al.  Inhibition of Dihydroorotate Dehydrogenase Overcomes Differentiation Blockade in Acute Myeloid Leukemia , 2016, Cell.

[162]  Jianlin Lei,et al.  The architecture of the mammalian respirasome , 2016, Nature.

[163]  H. Tse,et al.  iPSC-MSCs with High Intrinsic MIRO1 and Sensitivity to TNF-α Yield Efficacious Mitochondrial Transfer to Rescue Anthracycline-Induced Cardiomyopathy , 2016, Stem cell reports.

[164]  M. Berridge,et al.  Mitochondrial Transfer from Astrocytes to Neurons following Ischemic Insult: Guilt by Association? , 2016, Cell metabolism.

[165]  Martin Picard,et al.  The rise of mitochondria in medicine. , 2016, Mitochondrion.

[166]  P. D. del Nido,et al.  Intracoronary Delivery of Mitochondria to the Ischemic Heart for Cardioprotection , 2016, PloS one.

[167]  Xinnan Wang,et al.  Transporting mitochondria in neurons , 2016, F1000Research.

[168]  V. Imbert,et al.  Protective mitochondrial transfer from bone marrow stromal cells to acute myeloid leukemic cells during chemotherapy. , 2016, Blood.

[169]  O. Kretz,et al.  Mitochondrial Dynamics Controls T Cell Fate through Metabolic Programming , 2016, Cell.

[170]  E. Lo,et al.  Transfer of mitochondria from astrocytes to neurons after stroke , 2016, Nature.

[171]  C. Gustafsson,et al.  Maintenance and Expression of Mammalian Mitochondrial DNA. , 2016, Annual review of biochemistry.

[172]  M. N. Islam,et al.  Intercellular mitochondrial transfer: bioenergetic crosstalk between cells. , 2016, Current opinion in genetics & development.

[173]  Andrew J. Roger,et al.  A Eukaryote without a Mitochondrial Organelle , 2016, Current Biology.

[174]  Eleni Fotopoulou,et al.  Mitochondrial genetic diversity, selection and recombination in a canine transmissible cancer , 2016, eLife.

[175]  L. S. Churchman,et al.  Synchronized mitochondrial and cytosolic translation programs , 2016, Nature.

[176]  Ting-Hsiang Wu,et al.  Modifying the Mitochondrial Genome. , 2016, Cell metabolism.

[177]  G. Pigino,et al.  Microtubule doublets are double-track railways for intraflagellar transport trains , 2016, Science.

[178]  Shinn-Zong Lin,et al.  Transferring Xenogenic Mitochondria Provides Neural Protection against Ischemic Stress in Ischemic Rat Brains , 2016, Cell transplantation.

[179]  M. Matthay,et al.  Mitochondrial Transfer via Tunneling Nanotubes is an Important Mechanism by Which Mesenchymal Stem Cells Enhance Macrophage Phagocytosis in the In Vitro and In Vivo Models of ARDS , 2016, Stem cells.

[180]  P. Navas,et al.  The CoQH2/CoQ Ratio Serves as a Sensor of Respiratory Chain Efficiency. , 2016, Cell reports.

[181]  A. Spradling,et al.  Mouse oocytes differentiate through organelle enrichment from sister cyst germ cells , 2016, Science.

[182]  S. Kuo,et al.  Allogeneic/xenogeneic transplantation of peptide-labeled mitochondria in Parkinson's disease: restoration of mitochondria functions and attenuation of 6-hydroxydopamine-induced neurotoxicity. , 2016, Translational research : the journal of laboratory and clinical medicine.

[183]  A. Rustom The missing link: does tunnelling nanotube-based supercellularity provide a new understanding of chronic and lifestyle diseases? , 2016, Open Biology.

[184]  Ruiyan Zhang,et al.  Bone marrow-derived mesenchymal stem cells rescue injured H9c2 cells via transferring intact mitochondria through tunneling nanotubes in an in vitro simulated ischemia/reperfusion model , 2015, Molecular medicine reports.

[185]  B. McEwen,et al.  Mitochondrial functions modulate neuroendocrine, metabolic, inflammatory, and transcriptional responses to acute psychological stress , 2015, Proceedings of the National Academy of Sciences.

[186]  O. Garaschuk,et al.  Brain tumour cells interconnect to a functional and resistant network , 2015, Nature.

[187]  Simon C Watkins,et al.  Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs , 2015, Nature Communications.

[188]  Robert W. Taylor,et al.  Mutations causing mitochondrial disease: What is new and what challenges remain? , 2015, Science.

[189]  P. Ferdinandy,et al.  Connexin 43 is an emerging therapeutic target in ischemia/reperfusion injury, cardioprotection and neuroprotection. , 2015, Pharmacology & therapeutics.

[190]  S. Jakobs,et al.  Cross-strand binding of TFAM to a single mtDNA molecule forms the mitochondrial nucleoid , 2015, Proceedings of the National Academy of Sciences.

[191]  J. Neuzil,et al.  Mitochondrial DNA in Tumor Initiation, Progression, and Metastasis: Role of Horizontal mtDNA Transfer. , 2015, Cancer research.

[192]  M. Peter,et al.  Mitotic redistribution of the mitochondrial network by Miro and Cenp-F , 2015, Nature Communications.

[193]  Leonora Balaj,et al.  Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. , 2015, Bioscience.

[194]  D. Zorov,et al.  Enabling Technologies for Cell-Based Clinical Translation Improving the Post-Stroke Therapeutic Potency of Mesenchymal Multipotent Stromal Cells by Cocultivation With Cortical Neurons : The Role of Crosstalk Between Cells , 2015 .

[195]  D. Sabatini,et al.  An Essential Role of the Mitochondrial Electron Transport Chain in Cell Proliferation Is to Enable Aspartate Synthesis , 2015, Cell.

[196]  Brenda M Ogle,et al.  Apoptosis‐induced cancer cell fusion: a mechanism of breast cancer metastasis , 2015, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[197]  J. Meldolesi,et al.  Ectosomes and exosomes: shedding the confusion between extracellular vesicles. , 2015, Trends in cell biology.

[198]  A. Bunck,et al.  Mosaic Deficiency in Mitochondrial Oxidative Metabolism Promotes Cardiac Arrhythmia during Aging. , 2015, Cell metabolism.

[199]  Andrés Caicedo,et al.  MitoCeption as a new tool to assess the effects of mesenchymal stem/stromal cell mitochondria on cancer cell metabolism and function , 2015, Scientific Reports.

[200]  C. Zurzolo,et al.  Prion aggregates transfer through tunneling nanotubes in endocytic vesicles , 2015, Prion.

[201]  Luca Lambertini,et al.  Stable heteroplasmy at the single-cell level is facilitated by intercellular exchange of mtDNA , 2015, Nucleic acids research.

[202]  E. Murchison,et al.  The cancer which survived: insights from the genome of an 11000 year-old cancer. , 2015, Current opinion in genetics & development.

[203]  William Lee,et al.  Mitochondrial DNA copy number is regulated by DNA methylation and demethylation of POLGA in stem and cancer cells and their differentiated progeny , 2015, Cell Death and Disease.

[204]  H. Gerdes,et al.  Transfer of mitochondria via tunneling nanotubes rescues apoptotic PC12 cells , 2015, Cell Death and Differentiation.

[205]  David A. Eccles,et al.  Mitochondrial genome acquisition restores respiratory function and tumorigenic potential of cancer cells without mitochondrial DNA. , 2015, Cell metabolism.

[206]  L. O’Driscoll,et al.  Biological properties of extracellular vesicles and their physiological functions , 2015, Journal of extracellular vesicles.

[207]  P. Chinnery,et al.  Disturbed mitochondrial dynamics and neurodegenerative disorders , 2015, Nature Reviews Neurology.

[208]  R. Carstens,et al.  Mitochondrial retrograde signaling induces epithelial–mesenchymal transition and generates breast cancer stem cells , 2014, Oncogene.

[209]  Y. Li,et al.  Discovery of the migrasome, an organelle mediating release of cytoplasmic contents during cell migration , 2014, Cell Research.

[210]  H. Ishwaran,et al.  Exosome Transfer from Stromal to Breast Cancer Cells Regulates Therapy Resistance Pathways , 2014, Cell.

[211]  M. Gelb,et al.  Platelets release mitochondria serving as substrate for bactericidal group IIA-secreted phospholipase A2 to promote inflammation. , 2014, Blood.

[212]  Vincent Procaccio,et al.  Progressive increase in mtDNA 3243A>G heteroplasmy causes abrupt transcriptional reprogramming , 2014, Proceedings of the National Academy of Sciences.

[213]  H. Tse,et al.  Mitochondrial transfer of induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial cells attenuates cigarette smoke-induced damage. , 2014, American journal of respiratory cell and molecular biology.

[214]  R. Kalluri,et al.  PGC-1α mediates mitochondrial biogenesis and oxidative phosphorylation to promote metastasis , 2014, Nature Cell Biology.

[215]  S. Pulst,et al.  Loss of Miro1-directed mitochondrial movement results in a novel murine model for neuron disease , 2014, Proceedings of the National Academy of Sciences.

[216]  John M. Asara,et al.  Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function , 2014, Nature.

[217]  Shan Jiang,et al.  Yap1 Activation Enables Bypass of Oncogenic Kras Addiction in Pancreatic Cancer , 2014, Cell.

[218]  Iain G. Johnston,et al.  MtDNA segregation in heteroplasmic tissues is common in vivo and modulated by haplotype differences and developmental stage. , 2014, Cell reports.

[219]  V. Uloza,et al.  Long-Distance Communication between Laryngeal Carcinoma Cells , 2014, PloS one.

[220]  Nathan A. Bihlmeyer,et al.  Transcellular degradation of axonal mitochondria , 2014, Proceedings of the National Academy of Sciences.

[221]  M. Nireekshan Kumar,et al.  Miro1 regulates intercellular mitochondrial transport & enhances mesenchymal stem cell rescue efficacy , 2014, The EMBO journal.

[222]  K. Manova-Todorova,et al.  Tumor exosomes induce tunneling nanotubes in lipid raft-enriched regions of human mesothelioma cells. , 2014, Experimental cell research.

[223]  Huixia Lu,et al.  Mesenchymal stem cells rescue injured endothelial cells in an in vitro ischemia-reperfusion model via tunneling nanotube like structure-mediated mitochondrial transfer. , 2014, Microvascular research.

[224]  Peter J. Campbell,et al.  Transmissible Dog Cancer Genome Reveals the Origin and History of an Ancient Cell Lineage , 2014, Science.

[225]  J. Bhattacharya,et al.  When cells become organelle donors. , 2013, Physiology.

[226]  C. Kukat,et al.  mtDNA makes a U-turn for the mitochondrial nucleoid. , 2013, Trends in cell biology.

[227]  J. Donoghue,et al.  The regulation of mitochondrial DNA copy number in glioblastoma cells , 2013, Cell Death and Differentiation.

[228]  Haifa Qiao,et al.  Motile axonal mitochondria contribute to the variability of presynaptic strength. , 2013, Cell reports.

[229]  M. Wong,et al.  Genetic basis of cell-cell fusion mechanisms. , 2013, Trends in genetics : TIG.

[230]  C. López-Otín,et al.  Supercomplex Assembly Determines Electron Flux in the Mitochondrial Electron Transport Chain , 2013, Science.

[231]  Imre Mäger,et al.  Extracellular vesicles: biology and emerging therapeutic opportunities , 2013, Nature Reviews Drug Discovery.

[232]  S. Rafii,et al.  Preferential transfer of mitochondria from endothelial to cancer cells through tunneling nanotubes modulates chemoresistance , 2013, Journal of Translational Medicine.

[233]  Douglas B. Cowan,et al.  Transplantation of Autologously‐Derived Mitochondria Protects the Heart from Ischemia‐Reperfusion Injury , 2013, American journal of physiology. Heart and circulatory physiology.

[234]  I. Lai,et al.  Isolated Mitochondria Infusion Mitigates Ischemia-Reperfusion Injury of the Liver in Rats , 2013, Shock.

[235]  C. Harding,et al.  Exosomes: Looking back three decades and into the future , 2013, The Journal of cell biology.

[236]  O. Lindvall,et al.  Embryonic Stem Cell‐Derived Neural Stem Cells Fuse with Microglia and Mature Neurons , 2012, Stem cells.

[237]  S. Dimauro,et al.  Human mitochondrial DNA: roles of inherited and somatic mutations , 2012, Nature Reviews Genetics.

[238]  Adrian Carr,et al.  Biochemical Diversification through Foreign Gene Expression in Bdelloid Rotifers , 2012, PLoS genetics.

[239]  D. Wallace Mitochondria and cancer , 2012, Nature Reviews Cancer.

[240]  J. Visvader,et al.  Cancer stem cells: current status and evolving complexities. , 2012, Cell stem cell.

[241]  H. Haller,et al.  Vascular smooth muscle cells initiate proliferation of mesenchymal stem cells by mitochondrial transfer via tunneling nanotubes. , 2012, Stem cells and development.

[242]  F. Sánchez‐Madrid,et al.  Intercellular communication: diverse structures for exchange of genetic information , 2012, Nature Reviews Molecular Cell Biology.

[243]  D. Rowlands,et al.  Mitochondrial transfer from bone-marrow–derived stromal cells to pulmonary alveoli protects against acute lung injury , 2012, Nature Medicine.

[244]  H. Gerdes,et al.  Multi-Level Communication of Human Retinal Pigment Epithelial Cells via Tunneling Nanotubes , 2012, PloS one.

[245]  K. Manova-Todorova,et al.  Tunneling Nanotubes Provide a Unique Conduit for Intercellular Transfer of Cellular Contents in Human Malignant Pleural Mesothelioma , 2012, PloS one.

[246]  F. Fontanesi,et al.  Mitochondrial complex I plays an essential role in human respirasome assembly. , 2012, Cell metabolism.

[247]  Ju Han Kim,et al.  Mesenchymal Stem Cells Transfer Mitochondria to the Cells with Virtually No Mitochondrial Function but Not with Pathogenic mtDNA Mutations , 2012, PloS one.

[248]  Carolyn Tregidgo,et al.  Genome Sequencing and Analysis of the Tasmanian Devil and Its Transmissible Cancer , 2012, Cell.

[249]  H. Gendelman,et al.  Human Immunodeficiency Virus type 1 Endocytic Trafficking Through Macrophage Bridging Conduits Facilitates Spread of Infection , 2011, Journal of Neuroimmune Pharmacology.

[250]  Ming Xu,et al.  Long-distance intercellular connectivity between cardiomyocytes and cardiofibroblasts mediated by membrane nanotubes. , 2011, Cardiovascular research.

[251]  L. Hlatky,et al.  Intercellular Communication by Exchange of Cytoplasmic Material via Tunneling Nano-Tube Like Structures in Primary Human Renal Epithelial Cells , 2011, PloS one.

[252]  H. Gendelman,et al.  Macrophage Bridging Conduit Trafficking of HIV-1 Through the Endoplasmic Reticulum and Golgi Network , 2011, Journal of proteome research.

[253]  P. Lesault,et al.  Human Mesenchymal Stem Cells Reprogram Adult Cardiomyocytes Toward a Progenitor‐Like State Through Partial Cell Fusion and Mitochondria Transfer , 2011, Stem cells.

[254]  Chi V. Dang,et al.  Otto Warburg's contributions to current concepts of cancer metabolism , 2011, Nature Reviews Cancer.

[255]  J. Shuai,et al.  Active generation and propagation of Ca2+ signals within tunneling membrane nanotubes. , 2011, Biophysical journal.

[256]  Y. Wang,et al.  Tunneling-nanotube development in astrocytes depends on p53 activation , 2011, Cell Death and Differentiation.

[257]  M. Berardi,et al.  Survival of the fittest: metabolic adaptations in cancer. , 2011, Current opinion in genetics & development.

[258]  A. Burt,et al.  Mitochondrial Capture by a Transmissible Cancer , 2011, Science.

[259]  Nickolay V. Bukoreshtliev,et al.  Animal cells connected by nanotubes can be electrically coupled through interposed gap-junction channels , 2010, Proceedings of the National Academy of Sciences.

[260]  G. Sukhikh,et al.  Cytoplasm and organelle transfer between mesenchymal multipotent stromal cells and renal tubular cells in co-culture. , 2010, Experimental cell research.

[261]  H. Westerblad,et al.  Myogenic skeletal muscle satellite cells communicate by tunnelling nanotubes , 2010, Journal of cellular physiology.

[262]  M. Goligorsky,et al.  Adriamycin nephropathy: a failure of endothelial progenitor cell-induced repair. , 2010, The American journal of pathology.

[263]  A. MacAskill,et al.  Control of mitochondrial transport and localization in neurons. , 2010, Trends in Cell Biology.

[264]  M. Soares,et al.  Mechanisms of cell protection by heme oxygenase-1. , 2010, Annual review of pharmacology and toxicology.

[265]  W. Junger,et al.  Circulating Mitochondrial DAMPs Cause Inflammatory Responses to Injury , 2009, Nature.

[266]  Manolis Kellis,et al.  The Tasmanian Devil Transcriptome Reveals Schwann Cell Origins of a Clonally Transmissible Cancer , 2009, Science.

[267]  A. Cselenyák,et al.  Mesenchymal stem cells rescue cardiomyoblasts from cell death in an in vitro ischemia model via direct cell-to-cell connections , 2010, BMC Cell Biology.

[268]  S. Kimura,et al.  M-Sec promotes membrane nanotube formation by interacting with Ral and the exocyst complex , 2009, Nature Cell Biology.

[269]  A. Burt,et al.  Origins and Evolution of a Transmissible Cancer , 2009, Evolution; international journal of organic evolution.

[270]  L. Cantley,et al.  Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation , 2009, Science.

[271]  E. Hodneland,et al.  Selective block of tunneling nanotube (TNT) formation inhibits intercellular organelle transfer between PC12 cells , 2009, FEBS letters.

[272]  S. Houser,et al.  Concentration-dependent inhibition of angiogenesis by mesenchymal stem cells. , 2009, Blood.

[273]  K. Gousset,et al.  Tunnelling nanotubes , 2009, Prion.

[274]  Nicolas Chenouard,et al.  Prions hijack tunnelling nanotubes for intercellular spread , 2009, Nature Cell Biology.

[275]  D. Paul,et al.  Gap junctions. , 2009, Cold Spring Harbor perspectives in biology.

[276]  Douglas B. Cowan,et al.  Injection of isolated mitochondria during early reperfusion for cardioprotection. , 2009, American journal of physiology. Heart and circulatory physiology.

[277]  J. Berman,et al.  Tunneling nanotubes (TNT) are induced by HIV-infection of macrophages: a potential mechanism for intercellular HIV trafficking. , 2009, Cellular immunology.

[278]  J. Enríquez,et al.  Respiratory active mitochondrial supercomplexes. , 2008, Molecular cell.

[279]  Gerhard J Schütz,et al.  Different types of cell-to-cell connections mediated by nanotubular structures. , 2008, Biophysical journal.

[280]  H. Gerdes,et al.  Intercellular transfer mediated by tunneling nanotubes. , 2008, Current opinion in cell biology.

[281]  J. Palmer,et al.  Horizontal gene transfer in eukaryotic evolution , 2008, Nature Reviews Genetics.

[282]  M. Meselson,et al.  Massive Horizontal Gene Transfer in Bdelloid Rotifers , 2008, Science.

[283]  J. Hayashi,et al.  ROS-Generating Mitochondrial DNA Mutations Can Regulate Tumor Cell Metastasis , 2008, Science.

[284]  Lina A. Thoren,et al.  Myeloid and lymphoid contribution to non-haematopoietic lineages through irradiation-induced heterotypic cell fusion , 2008, Nature Cell Biology.

[285]  H. Gerdes,et al.  The art of cellular communication: tunneling nanotubes bridge the divide , 2008, Histochemistry and Cell Biology.

[286]  David C Samuels,et al.  What causes mitochondrial DNA deletions in human cells? , 2008, Nature Genetics.

[287]  Q. Sattentau,et al.  Membrane nanotubes physically connect T cells over long distances presenting a novel route for HIV-1 transmission , 2008, Nature Cell Biology.

[288]  G. Sukhikh,et al.  Cell-to-cell cross-talk between mesenchymal stem cells and cardiomyocytes in co-culture , 2007, Journal of cellular and molecular medicine.

[289]  Keshav K. Singh,et al.  Xenogenic transfer of isolated murine mitochondria into human rho0 cells can improve respiratory function. , 2007, Rejuvenation research.

[290]  J. Ho,et al.  Mitochondria transfer can enhance the murine embryo development , 2007, Journal of Assisted Reproduction and Genetics.

[291]  C. Gustafsson,et al.  DNA replication and transcription in mammalian mitochondria. , 2007, Annual review of biochemistry.

[292]  Michael T Ryan,et al.  Mitochondrial-nuclear communications. , 2007, Annual review of biochemistry.

[293]  Walter Neupert,et al.  Why Do We Still Have a Maternally Inherited Mitochondrial DNA ? Insights from Evolutionary Medicine , 2007 .

[294]  H. Gerdes,et al.  Tunneling nanotubes: A new route for the exchange of components between animal cells , 2007, FEBS letters.

[295]  M. Neil,et al.  Structurally Distinct Membrane Nanotubes between Human Macrophages Support Long-Distance Vesicular Traffic or Surfing of Bacteria1 , 2006, The Journal of Immunology.

[296]  R. A. Butow,et al.  Mitochondrial retrograde signaling. , 2006, Annual review of genetics.

[297]  J. Enríquez,et al.  Differences in reactive oxygen species production explain the phenotypes associated with common mouse mitochondrial DNA variants , 2006, Nature Genetics.

[298]  Darwin J. Prockop,et al.  Mitochondrial transfer between cells can rescue aerobic respiration , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[299]  C. Futter,et al.  EGF stimulates annexin 1‐dependent inward vesiculation in a multivesicular endosome subpopulation , 2006, The EMBO journal.

[300]  Simon C Watkins,et al.  Functional connectivity between immune cells mediated by tunneling nanotubules. , 2005, Immunity.

[301]  R. Brandes,et al.  Cell-to-Cell Connection of Endothelial Progenitor Cells With Cardiac Myocytes by Nanotubes: A Novel Mechanism for Cell Fate Changes? , 2005, Circulation research.

[302]  John A. Hall,et al.  mtDNA mutations increase tumorigenicity in prostate cancer. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[303]  Elisabetta Dejana,et al.  Endothelial cell–cell junctions: happy together , 2004, Nature Reviews Molecular Cell Biology.

[304]  Hans-Hermann Gerdes,et al.  Nanotubular Highways for Intercellular Organelle Transport , 2004, Science.

[305]  S. Ohta A multi-functional organelle mitochondrion is involved in cell death, proliferation and disease. , 2003, Current medicinal chemistry.

[306]  Klaus Pfeffer,et al.  Fusion of bone-marrow-derived cells with Purkinje neurons, cardiomyocytes and hepatocytes , 2003, Nature.

[307]  Helen M. Blau,et al.  Stable reprogrammed heterokaryons form spontaneously in Purkinje neurons after bone marrow transplant , 2003, Nature Cell Biology.

[308]  J. Mullikin,et al.  Revisiting the mouse mitochondrial DNA sequence. , 2003, Nucleic acids research.

[309]  Jeffrey D. Palmer,et al.  Widespread horizontal transfer of mitochondrial genes in flowering plants , 2003, Nature.

[310]  Sandra L. Schmid,et al.  Regulated portals of entry into the cell , 2003, Nature.

[311]  Graça Raposo,et al.  The Biogenesis and Functions of Exosomes , 2002, Traffic.

[312]  E. Scott,et al.  Bone marrow cells adopt the phenotype of other cells by spontaneous cell fusion , 2002, Nature.

[313]  C. W. Birky,et al.  The inheritance of genes in mitochondria and chloroplasts: laws, mechanisms, and models. , 2001, Annual review of genetics.

[314]  B F Lang,et al.  Mitochondrial evolution. , 1999, Science.

[315]  M. King,et al.  Human cells lacking mtDNA: repopulation with exogenous mitochondria by complementation. , 1989, Science.