201
|
Chekhonin IV, Chistiakov DA, Grinenko NF, Gurina OI. Glioma Cell and Astrocyte Co-cultures As a Model to Study Tumor-Tissue Interactions: A Review of Methods. Cell Mol Neurobiol 2018; 38:1179-1195. [PMID: 29744691 PMCID: PMC11481938 DOI: 10.1007/s10571-018-0588-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 04/23/2018] [Indexed: 02/08/2023]
Abstract
Astrocytes are a dominant cell type that envelopes the glioma bed. Typically, that is followed by formation of contacts between astrocytes and glioma cells and accompanied by change in astrocyte phenotype, a phenomenon known as a 'reactive astrogliosis.' Generally considered glioma-promoting, astrocytes have many controversial peculiarities in communication with tumor cells, which need thorough examination in vitro. This review is devoted to in vitro co-culture studies of glioma cells and astrocytes. Firstly, we list several fundamental works which allow understanding the modalities of co-culturing. Cell-to-cell interactions between astrocytes and glioma cells, the roles of astrocytes in tumor metabolism, and glioma-related angiogenesis are reviewed. In the review, we also discuss communications between glioma stem cells and astrocytes. Co-cultures of glioma cells and astrocytes are used for studying anti-glioma treatment approaches. We also enumerate surgical, chemotherapeutic, and radiotherapeutic methods assessed in co-culture experiments. In conclusion, we underline collisions in the field and point out the role of the co-cultures for neurobiological studies.
Collapse
Affiliation(s)
- Ivan V Chekhonin
- Department of Fundamental and Applied Neurobiology, V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Kropotkinskiy pereulok 23, Moscow, 119034, Russian Federation.
| | - Dimitry A Chistiakov
- Department of Fundamental and Applied Neurobiology, V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Kropotkinskiy pereulok 23, Moscow, 119034, Russian Federation
| | - Nadezhda F Grinenko
- Department of Fundamental and Applied Neurobiology, V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Kropotkinskiy pereulok 23, Moscow, 119034, Russian Federation
| | - Olga I Gurina
- Department of Fundamental and Applied Neurobiology, V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Kropotkinskiy pereulok 23, Moscow, 119034, Russian Federation
| |
Collapse
|
202
|
The role of metabolism and tunneling nanotube-mediated intercellular mitochondria exchange in cancer drug resistance. Biochem J 2018; 475:2305-2328. [PMID: 30064989 DOI: 10.1042/bcj20170712] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/11/2018] [Accepted: 07/03/2018] [Indexed: 12/14/2022]
Abstract
Intercellular communications play a major role in tissue homeostasis. In pathologies such as cancer, cellular interactions within the tumor microenvironment (TME) contribute to tumor progression and resistance to therapy. Tunneling nanotubes (TNTs) are newly discovered long-range intercellular connections that allow the exchange between cells of various cargos, ranging from ions to whole organelles such as mitochondria. TNT-transferred mitochondria were shown to change the metabolism and functional properties of recipient cells as reported for both normal and cancer cells. Metabolic plasticity is now considered a hallmark of cancer as it notably plays a pivotal role in drug resistance. The acquisition of cancer drug resistance was also associated to TNT-mediated mitochondria transfer, a finding that relates to the role of mitochondria as a hub for many metabolic pathways. In this review, we first give a brief overview of the various mechanisms of drug resistance and of the cellular communication means at play in the TME, with a special focus on the recently discovered TNTs. We further describe recent studies highlighting the role of the TNT-transferred mitochondria in acquired cancer cell drug resistance. We also present how changes in metabolic pathways, including glycolysis, pentose phosphate and lipid metabolism, are linked to cancer cell resistance to therapy. Finally, we provide examples of novel therapeutic strategies targeting mitochondria and cell metabolism as a way to circumvent cancer cell drug resistance.
Collapse
|
203
|
Nguyen Q, Shiva S. Moving mitochondria - Breathing new signaling into asthmatic airways. Redox Biol 2018; 18:244-245. [PMID: 30056272 PMCID: PMC6079482 DOI: 10.1016/j.redox.2018.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 07/13/2018] [Accepted: 07/19/2018] [Indexed: 11/17/2022] Open
Affiliation(s)
- Quyen Nguyen
- Division of Pulmonary Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Sruti Shiva
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| |
Collapse
|
204
|
Understanding the Role of Dysfunctional and Healthy Mitochondria in Stroke Pathology and Its Treatment. Int J Mol Sci 2018; 19:ijms19072127. [PMID: 30037107 PMCID: PMC6073421 DOI: 10.3390/ijms19072127] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/12/2018] [Accepted: 07/19/2018] [Indexed: 12/21/2022] Open
Abstract
Stroke remains a major cause of death and disability in the United States and around the world. Solid safety and efficacy profiles of novel stroke therapeutics have been generated in the laboratory, but most failed in clinical trials. Investigations into the pathology and treatment of the disease remain a key research endeavor in advancing scientific understanding and clinical applications. In particular, cell-based regenerative medicine, specifically stem cell transplantation, may hold promise as a stroke therapy, because grafted cells and their components may recapitulate the growth and function of the neurovascular unit, which arguably represents the alpha and omega of stroke brain pathology and recovery. Recent evidence has implicated mitochondria, organelles with a central role in energy metabolism and stress response, in stroke progression. Recognizing that stem cells offer a source of healthy mitochondria—one that is potentially transferrable into ischemic cells—may provide a new therapeutic tool. To this end, deciphering cellular and molecular processes underlying dysfunctional mitochondria may reveal innovative strategies for stroke therapy. Here, we review recent studies capturing the intimate participation of mitochondrial impairment in stroke pathology, and showcase promising methods of healthy mitochondria transfer into ischemic cells to critically evaluate the potential of mitochondria-based stem cell therapy for stroke patients.
Collapse
|
205
|
Tan DQ, Suda T. Reactive Oxygen Species and Mitochondrial Homeostasis as Regulators of Stem Cell Fate and Function. Antioxid Redox Signal 2018; 29:149-168. [PMID: 28708000 DOI: 10.1089/ars.2017.7273] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
SIGNIFICANCE The precise role and impact of reactive oxygen species (ROS) in stem cells, which are essential for lifelong tissue homeostasis and regeneration, remain of significant interest to the field. The long-term regenerative potential of a stem cell compartment is determined by the delicate balance between quiescence, self-renewal, and differentiation, all of which can be influenced by ROS levels. Recent Advances: The past decade has seen a growing appreciation for the importance of ROS and redox homeostasis in various stem cell compartments, particularly those of hematopoietic, neural, and muscle tissues. In recent years, the importance of proteostasis and mitochondria in relation to stem cell biology and redox homeostasis has garnered considerable interest. CRITICAL ISSUES Here, we explore the reciprocal relationship between ROS and stem cells, with significant emphasis on mitochondria as a core component of redox homeostasis. We discuss how redox signaling, involving cell-fate determining protein kinases and transcription factors, can control stem cell function and fate. We also address the impact of oxidative stress on stem cells, especially oxidative damage of lipids, proteins, and nucleic acids. We further discuss ROS management in stem cells, and present recent evidence supporting the importance of mitochondrial activity and its modulation (via mitochondrial clearance, biogenesis, dynamics, and distribution [i.e., segregation and transfer]) in stem cell redox homeostasis. FUTURE DIRECTIONS Therefore, elucidating the intricate links between mitochondria, cellular metabolism, and redox homeostasis is envisioned to be critical for our understanding of ROS in stem cell biology and its therapeutic relevance in regenerative medicine. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Darren Q Tan
- Cancer Science Institute of Singapore, National University of Singapore , Singapore, Singapore
| | - Toshio Suda
- Cancer Science Institute of Singapore, National University of Singapore , Singapore, Singapore
| |
Collapse
|
206
|
Sarmah D, Kaur H, Saraf J, Vats K, Pravalika K, Wanve M, Kalia K, Borah A, Kumar A, Wang X, Yavagal DR, Dave KR, Bhattacharya P. Mitochondrial Dysfunction in Stroke: Implications of Stem Cell Therapy. Transl Stroke Res 2018; 10:10.1007/s12975-018-0642-y. [PMID: 29926383 DOI: 10.1007/s12975-018-0642-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/21/2018] [Accepted: 06/12/2018] [Indexed: 01/06/2023]
Abstract
Stroke is a debilitating condition which is also the second leading cause of death and disability worldwide. Despite the benefits and promises shown by numerous neuroprotective agents in animal stroke models, their clinical translation has not been a complete success. Hence, search for treatment options have directed researchers towards utilising stem cells. Mitochondria has a major involvement in the pathophysiology of stroke and a number of other conditions. Stem cells have shown the ability to transfer mitochondria to the damaged cells and to help revive cell energetics in the recipient cell. The present review discusses how stem cells could be employed to protect neurons and mitochondria in stroke and also the various mechanisms involved in neuroprotection.
Collapse
Affiliation(s)
- Deepaneeta Sarmah
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Harpreet Kaur
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Jackson Saraf
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Kanchan Vats
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Kanta Pravalika
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Madhuri Wanve
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Kiran Kalia
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| | - Akhilesh Kumar
- Department of Botany, Banaras Hindu University, Varanasi, India
| | - Xin Wang
- Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Dileep R Yavagal
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Kunjan R Dave
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Pallab Bhattacharya
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
207
|
Association of Inflammatory Responses and ECM Disorganization with HMGB1 Upregulation and NLRP3 Inflammasome Activation in the Injured Rotator Cuff Tendon. Sci Rep 2018; 8:8918. [PMID: 29891998 PMCID: PMC5995925 DOI: 10.1038/s41598-018-27250-2] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/31/2018] [Indexed: 12/25/2022] Open
Abstract
Inflammation and extracellular matrix (ECM) disorganization following the rotator cuff tendon injuries (RCTI) delay the repair and healing process and the molecular mechanisms underlying RCTI pathology are largely unknown. Here, we examined the role of HMGB1 and NLRP3 inflammasome pathway in the inflammation and ECM disorganization in RCTI. This hypothesis was tested in a tenotomy-RCTI rat model by transecting the RC tendon from the humerus. H&E and pentachrome staining revealed significant changes in the morphology, architecture and ECM organization in RC tendon tissues following RCTI when compared with contralateral control. Severity of the injury was high in the first two weeks with improvement in 3–4 weeks following RCTI, and this correlated with the healing response. The expression of proteins associated with increased HMGB-1 and upregulation of NLRP3 inflammasome pathway, TLR4, TLR2, TREM-1, RAGE, ASC, Caspase-1, and IL-1β, in the first two weeks following RCTI followed by decline in 3–4 weeks. These results suggest the association of inflammatory responses and ECM disorganization with HMGB1 upregulation and NLRP3 inflammasome activation in the RC tendons and could provide novel target(s) for development of better therapeutic strategies in the management of RCTI.
Collapse
|
208
|
Weiss JN, Levy S. Stem Cell Ophthalmology Treatment Study: bone marrow derived stem cells in the treatment of Retinitis Pigmentosa. Stem Cell Investig 2018; 5:18. [PMID: 30050918 PMCID: PMC6043757 DOI: 10.21037/sci.2018.04.02] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 03/20/2018] [Indexed: 01/11/2023]
Abstract
BACKGROUND Seventeen patients with bilateral visual loss due to Retinitis Pigmentosa (RP) underwent autologous bone marrow derived stem cell (BMSC) treatment within the Stem Cell Ophthalmology Treatment Study (SCOTS and SCOTS 2). Both are National Institutes of Health (NIH) and Office of Human Research Protection (OHRP) compliant Institutional Review Board (IRB) approved clinical studies utilizing using autologous BMSC in the treatment of retinal and optic nerve diseases that meet inclusion criteria. METHODS The average age of the patients treated was 48.8 years. The average duration of disease prior to treatment was 27.6 years and ranged from 4 to approximately 60 years. Affected eyes were treated with either retrobulbar, subtenons and intravenous BMSC or retrobulbar, subtenons, intravitreal and intravenous. Follow up was provided a minimum of 6 months. The primary outcome was visual acuity as measured by Snellen or converted to LogMAR. RESULTS Following therapy in SCOTS or SCOTS 2, 11 patients (64.7%) showed improved binocular vision averaging 10.23 lines of Snellen acuity per eye over pre-treatment acuity; 8 patients (35.3%) remaining stable over the follow up period; no patients experiencing loss of overall acuity. In 33 treated eyes, 15 eyes (45.5%) improved an average of 7.9 lines of Snellen acuity, 15 eyes (45.5%) remained stable, and 3 eyes (9%) worsened by an average of 1.7 lines of Snellen acuity. Improvements ranged from 1 to 27 lines of vision. Using the LogMAR Scale and calculating delta as a ratio to pre-treatment vision in improved eyes, acuity improvement ranged from 23% to 90% with an average of 40.9% visual acuity improvement over baseline vision. Evaluation of all patients and eyes capable of LogMAR vision showed an average of 31% improvement in vision over baseline. Findings were of statistical significance (P=0.016). There were no surgical complications. CONCLUSIONS The BMSC protocols of the SCOTS achieved meaningful visual acuity improvements or stability in RP that were of statistical significance. Duration of disease did not appear to affect the ability of eyes to respond. Safety was confirmed. Possible mechanisms by which improvement occurred may include transdifferentiation of BMSC into Neuronal Nuclei (NeuN) positive cells, BMSC paracrine secretions or neurotrophic factors and hormones, transfer of mitochondria, release of messenger RNA or other compounds via exosomes or microvesicles. Given the successful outcome in this otherwise progressive condition, consideration should be given to providing this treatment option.
Collapse
|
209
|
Pedicini L, Miteva KT, Hawley V, Gaunt HJ, Appleby HL, Cubbon RM, Marszalek K, Kearney MT, Beech DJ, McKeown L. Homotypic endothelial nanotubes induced by wheat germ agglutinin and thrombin. Sci Rep 2018; 8:7569. [PMID: 29765077 PMCID: PMC5953990 DOI: 10.1038/s41598-018-25853-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/27/2018] [Indexed: 12/21/2022] Open
Abstract
Endothelial barrier formation is maintained by intercellular communication through junctional proteins. The mechanisms involved in maintaining endothelial communication subsequent to barrier disruption remain unclear. It is known that low numbers of endothelial cells can be interconnected by homotypic actin-driven tunneling nanotubes (TNTs) which could be important for intercellular transfer of information in vascular physiology. Here we sought insight into the triggers for TNT formation. Wheat germ agglutinin, a C-type lectin and known label for TNTs, unexpectedly caused striking induction of TNTs. A succinylated derivative was by contrast inactive, suggesting mediation by a sialylated protein. Through siRNA-mediated knockdown we identified that this protein was likely to be CD31, an important sialylated membrane protein normally at endothelial cell junctions. We subsequently considered thrombin as a physiological inducer of endothelial TNTs because it reduces junctional contact. Thrombin reduced junctional contact, redistributed CD31 and induced TNTs, but its effect on TNTs was CD31-independent. Thrombin-induced TNTs nevertheless required PKCα, a known mediator of thrombin-dependent junctional remodelling, suggesting a necessity for junctional proteins in TNT formation. Indeed, TNT-inducing effects of wheat germ agglutinin and thrombin were both correlated with cortical actin rearrangement and similarly Ca2+-dependent, suggesting common underlying mechanisms. Once formed, Ca2+ signalling along TNTs was observed.
Collapse
Affiliation(s)
- Lucia Pedicini
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Katarina T Miteva
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Verity Hawley
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Hannah J Gaunt
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Hollie L Appleby
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Katarzyna Marszalek
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Lynn McKeown
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
210
|
Wang J, Li H, Yao Y, Zhao T, Chen YY, Shen YL, Wang LL, Zhu Y. Stem cell-derived mitochondria transplantation: a novel strategy and the challenges for the treatment of tissue injury. Stem Cell Res Ther 2018; 9:106. [PMID: 29653590 PMCID: PMC5899391 DOI: 10.1186/s13287-018-0832-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Damage of mitochondria in the initial period of tissue injury aggravates the severity of injury. Restoration of mitochondria dysfunction and mitochondrial-based therapeutics represent a potentially effective therapeutic strategy. Recently, mitochondrial transfer from stem cells has been demonstrated to play a significant role in rescuing injured tissues. The possible mechanisms of mitochondria released from stem cells, the pathways of mitochondria transfer between the donor stem cells and recipient cells, and the internalization of mitochondria into recipient cells are discussed. Moreover, a novel strategy for tissue injury based on the concept of stem cell-derived mitochondrial transplantation is pointed out, and the advantages and challenges are summarized.
Collapse
Affiliation(s)
- Jingyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Heyangzi Li
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Ying Yao
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Tengfei Zhao
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Ying-Ying Chen
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yue-Liang Shen
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Lin-Lin Wang
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
| | - Yongjian Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
211
|
Mahfouzi SH, Amoabediny G, Doryab A, Safiabadi-Tali SH, Ghanei M. Noninvasive Real-Time Assessment of Cell Viability in a Three-Dimensional Tissue. Tissue Eng Part C Methods 2018; 24:197-204. [DOI: 10.1089/ten.tec.2017.0371] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Seyed Hossein Mahfouzi
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
- The Research Center for New Technologies in Life Science Engineering, University of Tehran, Tehran, Iran
| | - Ghassem Amoabediny
- The Research Center for New Technologies in Life Science Engineering, University of Tehran, Tehran, Iran
- Department of Biochemical and Pharmaceutical Engineering, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Ali Doryab
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
- The Research Center for New Technologies in Life Science Engineering, University of Tehran, Tehran, Iran
| | - Seyed Hamid Safiabadi-Tali
- The Research Center for New Technologies in Life Science Engineering, University of Tehran, Tehran, Iran
- Department of Biochemical and Pharmaceutical Engineering, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mostafa Ghanei
- Chemical Injuries Research Center and Department of Pulmonary Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
212
|
Paliwal S, Chaudhuri R, Agrawal A, Mohanty S. Regenerative abilities of mesenchymal stem cells through mitochondrial transfer. J Biomed Sci 2018; 25:31. [PMID: 29602309 PMCID: PMC5877369 DOI: 10.1186/s12929-018-0429-1] [Citation(s) in RCA: 216] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/14/2018] [Indexed: 12/27/2022] Open
Abstract
The past decade has witnessed an upsurge in studies demonstrating mitochondrial transfer as one of the emerging mechanisms through which mesenchymal stem cells (MSCs) can regenerate and repair damaged cells or tissues. It has been found to play a critical role in healing several diseases related to brain injury, cardiac myopathies, muscle sepsis, lung disorders and acute respiratory disorders. Several studies have shown that various mechanisms are involved in mitochondrial transfer that includes tunnel tube formation, micro vesicle formation, gap junctions, cell fusion and others modes of transfer. Few studies have investigated the mechanisms that contribute to mitochondrial transfer, primarily comprising of signaling pathways involved in tunnel tube formation that facilitates tunnel tube formation for movement of mitochondria from one cell to another. Various stress signals such as release of damaged mitochondria, mtDNA and mitochondrial products along with elevated reactive oxygen species levels trigger the transfer of mitochondria from MSCs to recipient cells. However, extensive cell signaling pathways that lead to mitochondrial transfer from healthy cells are still under investigation and the changes that contribute to restoration of mitochondrial bioenergetics in recipient cells remain largely elusive. In this review, we have discussed the phenomenon of mitochondrial transfer from MSCs to neighboring stressed cells, and how this aids in cellular repair and regeneration of different organs such as lung, heart, eye, brain and kidney. The potential scope of mitochondrial transfer in providing novel therapeutic strategies for treatment of various pathophysiological conditions has also been discussed.
Collapse
Affiliation(s)
- Swati Paliwal
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Rituparna Chaudhuri
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Anurag Agrawal
- Molecular Immunogenetics Laboratory and Centre of Excellence for Translational Research in Asthma & Lung disease, CSIR-Institute of Genomics and Integrative Biology, Delhi, 110007, India.
| | - Sujata Mohanty
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
213
|
Čamernik K, Barlič A, Drobnič M, Marc J, Jeras M, Zupan J. Mesenchymal Stem Cells in the Musculoskeletal System: From Animal Models to Human Tissue Regeneration? Stem Cell Rev Rep 2018; 14:346-369. [DOI: 10.1007/s12015-018-9800-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
214
|
González-Nieto D, Fernández-García L, Pérez-Rigueiro J, Guinea GV, Panetsos F. Hydrogels-Assisted Cell Engraftment for Repairing the Stroke-Damaged Brain: Chimera or Reality. Polymers (Basel) 2018; 10:polym10020184. [PMID: 30966220 PMCID: PMC6415003 DOI: 10.3390/polym10020184] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/06/2018] [Accepted: 02/11/2018] [Indexed: 01/07/2023] Open
Abstract
The use of advanced biomaterials as a structural and functional support for stem cells-based therapeutic implants has boosted the development of tissue engineering applications in multiple clinical fields. In relation to neurological disorders, we are still far from the clinical reality of restoring normal brain function in neurodegenerative diseases and cerebrovascular disorders. Hydrogel polymers show unique mechanical stiffness properties in the range of living soft tissues such as nervous tissue. Furthermore, the use of these polymers drastically enhances the engraftment of stem cells as well as their capacity to produce and deliver neuroprotective and neuroregenerative factors in the host tissue. Along this article, we review past and current trends in experimental and translational research to understand the opportunities, benefits, and types of tentative hydrogel-based applications for the treatment of cerebral disorders. Although the use of hydrogels for brain disorders has been restricted to the experimental area, the current level of knowledge anticipates an intense development of this field to reach clinics in forthcoming years.
Collapse
Affiliation(s)
- Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain.
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain.
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain.
| | - Laura Fernández-García
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain.
| | - José Pérez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain.
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain.
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid 28040 Madrid, Spain.
| | - Gustavo V Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain.
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain.
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid 28040 Madrid, Spain.
| | - Fivos Panetsos
- Neurocomputing and Neurorobotics Research Group: Faculty of Biology and Faculty of Optics, Universidad Complutense de Madrid, 28040 Madrid, Spain.
- Instituto de Investigación Sanitaria, Hospital Clínico San Carlos Madrid, IdISSC, 28040 Madrid, Spain.
| |
Collapse
|
215
|
Neuroprotective Effects of Bioactive Compounds and MAPK Pathway Modulation in "Ischemia"-Stressed PC12 Pheochromocytoma Cells. Brain Sci 2018; 8:brainsci8020032. [PMID: 29419806 PMCID: PMC5836051 DOI: 10.3390/brainsci8020032] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/24/2018] [Accepted: 02/02/2018] [Indexed: 02/08/2023] Open
Abstract
This review surveys the efforts taken to investigate in vitro neuroprotective features of synthetic compounds and cell-released growth factors on PC12 clonal cell line temporarily deprived of oxygen and glucose followed by reoxygenation (OGD/R). These cells have been used previously to mimic some of the properties of in vivo brain ischemia-reperfusion-injury (IRI) and have been instrumental in identifying common mechanisms such as calcium overload, redox potential, lipid peroxidation and MAPKs modulation. In addition, they were useful for establishing the role of certain membrane penetrable cocktails of antioxidants as well as potential growth factors which may act in neuroprotection. Pharmacological mechanisms of neuroprotection addressing modulation of the MAPK cascade and increased redox potential by natural products, drugs and growth factors secreted by stem cells, in either undifferentiated or nerve growth factor-differentiated PC12 cells exposed to ischemic conditions are discussed for future prospects in neuroprotection studies.
Collapse
|
216
|
Mitochondria are transported along microtubules in membrane nanotubes to rescue distressed cardiomyocytes from apoptosis. Cell Death Dis 2018; 9:81. [PMID: 29362447 PMCID: PMC5833423 DOI: 10.1038/s41419-017-0145-x] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/11/2017] [Accepted: 11/13/2017] [Indexed: 12/20/2022]
Abstract
Membrane nanotubes (MNTs) act as “highways” between cells to facilitate the transfer of multiple signals and play an important role in many diseases. Our previous work reported on the transfer of mitochondria via MNTs between cardiomyocytes (CMs) and cardiac myofibroblasts (MFs); however, the elucidation of the underlying mechanism and pathophysiological significance of this transfer requires additional study. In this study, we determined that the mean movement velocity of mitochondria in MNTs between CMs and MFs was approximately 17.5 ± 2.1 nm/s. Meanwhile, treatment with microtubule polymerisation inhibitors nocodazole or colcemid in cell culture decreased mitochondrial velocity, and knockdown of the microtubule motor protein kinesin family member 5B (KIF5B) led to a similar effect, indicating that mitochondrial movement was dependent on microtubules and the motor protein KIF5B. Furthermore, we showed that hypoxia/reoxygenation-induced CM apoptosis was attenuated by coculture with intact or hypoxia/reoxygenation-treated MFs, which transferred mitochondria to CMs. This rescue was prevented either by separating the cells using Transwell culture or by impairing mitochondrial transfer with nocodazole or colcemid treatment. In conclusion, as a novel means of intercellular communication, MNTs rescue distressed CMs from apoptosis by transporting mitochondria along microtubules via KIF5B.
Collapse
|
217
|
Wang J, Liu X, Qiu Y, Shi Y, Cai J, Wang B, Wei X, Ke Q, Sui X, Wang Y, Huang Y, Li H, Wang T, Lin R, Liu Q, Xiang AP. Cell adhesion-mediated mitochondria transfer contributes to mesenchymal stem cell-induced chemoresistance on T cell acute lymphoblastic leukemia cells. J Hematol Oncol 2018; 11:11. [PMID: 29357914 PMCID: PMC5778754 DOI: 10.1186/s13045-018-0554-z] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 01/12/2018] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Despite the high cure rate of T cell acute lymphoblastic leukemia (T-ALL), drug resistance to chemotherapy remains a significant clinical problem. Bone marrow mesenchymal stem cells (MSCs) protect leukemic cells from chemotherapy, but the underlying mechanisms are poorly understood. In this study, we aimed to uncover the mechanism of MSC-induced chemoresistance in T-ALL cells, thus providing a promising clinical therapy target. METHODS Cell viability was determined using the viability assay kit CCK-8. The mitochondrial ROS levels were detected using the fluorescent probe MitoSOX™ Red, and fluorescence intensity was measured by flow cytometry. In vitro, MSCs and Jurkat cells were cocultured. MSCs were labeled with green fluorescent protein (GFP), and Jurkat cells were labeled with the mitochondria-specific dye MitoTracker Red. Bidirectional mitochondrial transfer was detected by flow cytometry and confocal microscopy. The mechanism of mitochondria transfer was analyzed by inhibitor assays. Transcripts related to Jurkat cell/MSC adhesion in the coculture system were assessed by qRT-PCR. After treatment with a neutralizing antibody against a key adhesion molecule, mitochondria transfer from Jurkat cells to MSCs was again detected by flow cytometry and confocal microscopy. Finally, we verified our findings using human primary T-ALL cells cocultured with MSCs. RESULTS Chemotherapeutic drugs caused intracellular oxidative stress in Jurkat cells. Jurkat cells transfer mitochondria to MSCs but receive few mitochondria from MSCs, resulting in chemoresistance. This process of mitochondria transfer is mediated by tunneling nanotubes, which are protrusions that extend from the cell membrane . Moreover, we found that most Jurkat cells adhered to MSCs in the coculture system, which was mediated by the adhesion molecule ICAM-1. Treatment with a neutralizing antibody against ICAM-1 led to a decreased number of adhering Jurkat cells, decreased mitochondria transfer, and increased chemotherapy-induced cell death. CONCLUSIONS We show evidence that mitochondria transfer from Jurkat cells to MSCs, which is mediated by cell adhesion, may be a potential therapeutic target for T-ALL treatment.
Collapse
Affiliation(s)
- Jiancheng Wang
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 74# Zhongshan 2nd Road, Guangzhou, Guangdong, China.,Biotherapy Center, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xin Liu
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 74# Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Yuan Qiu
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 74# Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Yue Shi
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 74# Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Jianye Cai
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 74# Zhongshan 2nd Road, Guangzhou, Guangdong, China.,Biotherapy Center, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Boyan Wang
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 74# Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Xiaoyue Wei
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 74# Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Qiong Ke
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 74# Zhongshan 2nd Road, Guangzhou, Guangdong, China.,Biotherapy Center, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xin Sui
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 74# Zhongshan 2nd Road, Guangzhou, Guangdong, China.,The First Affiliated Hospital of Xi'an Jiaotong University Medical College, Xi'an, Shaanxi, 710061, China
| | - Yi Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 74# Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Yinong Huang
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 74# Zhongshan 2nd Road, Guangzhou, Guangdong, China.,Biotherapy Center, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Hongyu Li
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 74# Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Tao Wang
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 74# Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Ren Lin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Andy Peng Xiang
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China. .,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 74# Zhongshan 2nd Road, Guangzhou, Guangdong, China. .,Biotherapy Center, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China. .,Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 511436, China. .,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
218
|
Morrison TJ, Jackson MV, Cunningham EK, Kissenpfennig A, McAuley DF, O'Kane CM, Krasnodembskaya AD. Mesenchymal Stromal Cells Modulate Macrophages in Clinically Relevant Lung Injury Models by Extracellular Vesicle Mitochondrial Transfer. Am J Respir Crit Care Med 2017; 196:1275-1286. [PMID: 28598224 DOI: 10.1164/rccm.201701-0170oc] [Citation(s) in RCA: 554] [Impact Index Per Article: 69.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
RATIONALE Acute respiratory distress syndrome (ARDS) remains a major cause of respiratory failure in critically ill patients. Mesenchymal stromal cells (MSCs) are a promising candidate for a cell-based therapy. However, the mechanisms of MSCs' effects in ARDS are not well understood. In this study, we focused on the paracrine effect of MSCs on macrophage polarization and the role of extracellular vesicle (EV)-mediated mitochondrial transfer. OBJECTIVES To determine the effects of human MSCs on macrophage function in the ARDS environment and to elucidate the mechanisms of these effects. METHODS Human monocyte-derived macrophages (MDMs) were studied in noncontact coculture with human MSCs when stimulated with LPS or bronchoalveolar lavage fluid (BALF) from patients with ARDS. Murine alveolar macrophages (AMs) were cultured ex vivo with/without human MSC-derived EVs before adoptive transfer to LPS-injured mice. MEASUREMENTS AND MAIN RESULTS MSCs suppressed cytokine production, increased M2 macrophage marker expression, and augmented phagocytic capacity of human MDMs stimulated with LPS or ARDS BALF. These effects were partially mediated by CD44-expressing EVs. Adoptive transfer of AMs pretreated with MSC-derived EVs reduced inflammation and lung injury in LPS-injured mice. Inhibition of oxidative phosphorylation in MDMs prevented the modulatory effects of MSCs. Generating dysfunctional mitochondria in MSCs using rhodamine 6G pretreatment also abrogated these effects. CONCLUSIONS In the ARDS environment, MSCs promote an antiinflammatory and highly phagocytic macrophage phenotype through EV-mediated mitochondrial transfer. MSC-induced changes in macrophage phenotype critically depend on enhancement of macrophage oxidative phosphorylation. AMs treated with MSC-derived EVs ameliorate lung injury in vivo.
Collapse
Affiliation(s)
- Thomas J Morrison
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Megan V Jackson
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Erin K Cunningham
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Adrien Kissenpfennig
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Daniel F McAuley
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Cecilia M O'Kane
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Anna D Krasnodembskaya
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
219
|
Singh B, Modica-Napolitano JS, Singh KK. Defining the momiome: Promiscuous information transfer by mobile mitochondria and the mitochondrial genome. Semin Cancer Biol 2017; 47:1-17. [PMID: 28502611 PMCID: PMC5681893 DOI: 10.1016/j.semcancer.2017.05.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/20/2017] [Accepted: 05/07/2017] [Indexed: 12/30/2022]
Abstract
Mitochondria are complex intracellular organelles that have long been identified as the powerhouses of eukaryotic cells because of the central role they play in oxidative metabolism. A resurgence of interest in the study of mitochondria during the past decade has revealed that mitochondria also play key roles in cell signaling, proliferation, cell metabolism and cell death, and that genetic and/or metabolic alterations in mitochondria contribute to a number of diseases, including cancer. Mitochondria have been identified as signaling organelles, capable of mediating bidirectional intracellular information transfer: anterograde (from nucleus to mitochondria) and retrograde (from mitochondria to nucleus). More recently, evidence is now building that the role of mitochondria extends to intercellular communication as well, and that the mitochondrial genome (mtDNA) and even whole mitochondria are indeed mobile and can mediate information transfer between cells. We define this promiscuous information transfer function of mitochondria and mtDNA as "momiome" to include all mobile functions of mitochondria and the mitochondrial genome. Herein, we review the "momiome" and explore its role in cancer development, progression, and treatment.
Collapse
Affiliation(s)
- Bhupendra Singh
- Department of Genetics, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Keshav K Singh
- Department of Genetics, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Environmental Health, Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Aging, University of Alabama at Birmingham, Birmingham, AL, USA; UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA; Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA.
| |
Collapse
|
220
|
Berridge MV, Herst PM, Rowe MR, Schneider R, McConnell MJ. Mitochondrial transfer between cells: Methodological constraints in cell culture and animal models. Anal Biochem 2017; 552:75-80. [PMID: 29158129 DOI: 10.1016/j.ab.2017.11.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 01/18/2023]
Abstract
Interest in the recently discovered phenomenon of mitochondrial transfer between mammalian cells has gained momentum since it was first described in cell culture systems more than a decade ago. Mitochondria-targeting fluorescent dyes have been repurposed and are now widely used in these studies and in acute disease models, sometimes without due consideration of their limitations, while vectors containing mitochondrially-imported fluorescent proteins have complemented the use of mitochondria-targeting dyes. Genetic approaches that use mitochondrial DNA polymorphisms have also been used in some in vitro studies and in tumor models and are particularly useful where mtDNA is damaged or deleted. These approaches can also be used to study the long-term consequences of mitochondrial transfer such as in bone marrow and organ transplantation and in tumour biology where inherent mitochondrial damage is often a key feature. As research on intercellular mitochondrial transfer moves from cell culture into animal models and human diseases it will be important to understand the limitations of the various techniques in order to apply appropriate methodologies to address physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- M V Berridge
- Cancer Cell Biology, Malaghan Institute of Medical Research, PO Box 7060, Wellington 6242, New Zealand.
| | - P M Herst
- Cancer Cell Biology, Malaghan Institute of Medical Research, PO Box 7060, Wellington 6242, New Zealand; Department of Radiation Therapy, University of Otago, PO Box 7343, Wellington 6242, New Zealand
| | - M R Rowe
- Department of Radiation Therapy, University of Otago, PO Box 7343, Wellington 6242, New Zealand; School of Biological Sciences, Victoria University, PO Box 600, Wellington 6140, New Zealand
| | - R Schneider
- Department of Radiation Therapy, University of Otago, PO Box 7343, Wellington 6242, New Zealand; School of Biological Sciences, Victoria University, PO Box 600, Wellington 6140, New Zealand
| | - M J McConnell
- School of Biological Sciences, Victoria University, PO Box 600, Wellington 6140, New Zealand
| |
Collapse
|
221
|
Griessinger E, Moschoi R, Biondani G, Peyron JF. Mitochondrial Transfer in the Leukemia Microenvironment. Trends Cancer 2017; 3:828-839. [PMID: 29198439 DOI: 10.1016/j.trecan.2017.10.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 10/06/2017] [Accepted: 10/10/2017] [Indexed: 12/18/2022]
Abstract
The bone marrow microenvironment (BMME) is a complex ecosystem that instructs and protects hematopoietic stem cells (HSCs) and their malignant counterparts, the leukemia-initiating cells (LICs). Within the physical and functional crosstalk that takes place between HSCs, LICs, and the BMME, the transfer of organelles and of mitochondria in particular is an important new intercellular communication mode in addition to adhesion molecules, tunneling nanotubes (TNTs), and the paracrine secretion of cytokines, (onco)metabolites, and extracellular vesicles (EVs). In this review we discuss the functional roles of mitochondrial transfer between BMME and leukemic cells, and give insights into this new mechanism of drug resistance whose understanding will open the way to innovative anticancer adjuvant treatments.
Collapse
Affiliation(s)
- Emmanuel Griessinger
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1065, Hôpital de l'Archet 2, 06204 Nice CEDEX, France.
| | - Ruxanda Moschoi
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1065, Hôpital de l'Archet 2, 06204 Nice CEDEX, France
| | - Giulia Biondani
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1065, Hôpital de l'Archet 2, 06204 Nice CEDEX, France
| | - Jean-François Peyron
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1065, Hôpital de l'Archet 2, 06204 Nice CEDEX, France.
| |
Collapse
|
222
|
Keller KE, Bradley JM, Sun YY, Yang YF, Acott TS. Tunneling Nanotubes are Novel Cellular Structures That Communicate Signals Between Trabecular Meshwork Cells. Invest Ophthalmol Vis Sci 2017; 58:5298-5307. [PMID: 29049733 PMCID: PMC5656416 DOI: 10.1167/iovs.17-22732] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Purpose The actin cytoskeleton of trabecular meshwork (TM) cells plays a role in regulating aqueous humor outflow. Many studies have investigated stress fibers, but F-actin also assembles into other supramolecular structures including filopodia. Recently, specialized filopodia called tunneling nanotubes (TNTs) have been described, which communicate molecular signals and organelles directly between cells. Here, we investigate TNT formation by TM cells. Methods Human TM cells were labeled separately with the fluorescent dyes, DiO and DiD, or with mitochondrial dye. Fixed or live TM cells were imaged using confocal microscopy. Image analysis software was used to track fluorescent vesicles and count the number and length of filopodia. The number of fluorescently labeled vesicles transferred between cells was counted in response to specific inhibitors of the actin cytoskeleton. Human TM tissue was stained with phalloidin. Results Live-cell confocal imaging of cultured TM cells showed transfer of fluorescently labeled vesicles and mitochondria via TNTs. In TM tissue, a long (160 μm) actin-rich cell process bridged an intertrabecular space and did not adhere to the substratum. Treatment of TM cells with CK-666, an Arp2/3 inhibitor, significantly decreased the number and length of filopodia, decreased transfer of fluorescently labeled vesicles and induced thick stress fibers compared to vehicle control. Conversely, inhibiting stress fibers using Y27632 increased transfer of vesicles and induced long cell processes. Conclusions Identification of TNTs provides a means by which TM cells can directly communicate with each other over long distances. This may be particularly important to overcome limitations of diffusion-based signaling in the aqueous humor fluid environment.
Collapse
Affiliation(s)
- Kate E Keller
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - John M Bradley
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Ying Ying Sun
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Yong-Feng Yang
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Ted S Acott
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
223
|
Rocca CJ, Goodman SM, Dulin JN, Haquang JH, Gertsman I, Blondelle J, Smith JLM, Heyser CJ, Cherqui S. 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. [PMID: 29070698 PMCID: PMC5735830 DOI: 10.1126/scitranslmed.aaj2347] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/31/2017] [Accepted: 06/13/2017] [Indexed: 12/14/2022]
Abstract
Friedreich's ataxia (FRDA) is an incurable autosomal recessive neurodegenerative disease caused by reduced expression of the mitochondrial protein frataxin due to an intronic GAA-repeat expansion in the FXN gene. We report the therapeutic efficacy of transplanting wild-type mouse hematopoietic stem and progenitor cells (HSPCs) into the YG8R mouse model of FRDA. In the HSPC-transplanted YG8R mice, development of muscle weakness and locomotor deficits was abrogated as was degeneration of large sensory neurons in the dorsal root ganglia (DRGs) and mitochondrial capacity was improved in brain, skeletal muscle, and heart. Transplanted HSPCs engrafted and then differentiated into microglia in the brain and spinal cord and into macrophages in the DRGs, heart, and muscle of YG8R FRDA mice. We observed the transfer of wild-type frataxin and Cox8 mitochondrial proteins from HSPC-derived microglia/macrophages to FRDA mouse neurons and muscle myocytes in vivo. Our results show the HSPC-mediated phenotypic rescue of FRDA in YG8R mice and suggest that this approach should be investigated further as a strategy for treating FRDA.
Collapse
Affiliation(s)
- Celine J Rocca
- Division of Genetics, Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Spencer M Goodman
- Division of Genetics, Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jennifer N Dulin
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Joseph H Haquang
- Division of Genetics, Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ilya Gertsman
- Division of Genetics, Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jordan Blondelle
- Division of Cardiovascular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Janell L M Smith
- Division of Genetics, Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Charles J Heyser
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stephanie Cherqui
- Division of Genetics, Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
224
|
Antunes MA, Lapa E Silva JR, Rocco PR. Mesenchymal stromal cell therapy in COPD: from bench to bedside. Int J Chron Obstruct Pulmon Dis 2017; 12:3017-3027. [PMID: 29081655 PMCID: PMC5652911 DOI: 10.2147/copd.s146671] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
COPD is the most frequent chronic respiratory disease and a leading cause of morbidity and mortality. The major risk factor for COPD development is cigarette smoke, and the most efficient treatment for COPD is smoking cessation. However, even after smoking cessation, inflammation, apoptosis, and oxidative stress may persist and continue contributing to disease progression. Although current therapies for COPD (primarily based on anti-inflammatory agents) contribute to the reduction of airway obstruction and minimize COPD exacerbations, none can avoid disease progression or reduce mortality. Within this context, recent advances in mesenchymal stromal cell (MSC) therapy have made this approach a strong candidate for clinical use in the treatment of several pulmonary diseases. MSCs can be readily harvested from diverse tissues and expanded with high efficiency, and have strong immunosuppressive properties. Preclinical studies have demonstrated encouraging outcomes of MSCs therapy for lung disorders, including emphysema. These findings instigated research groups to assess the impact of MSCs in human COPD/emphysema, but clinical results have fallen short of expectations. However, MSCs have demonstrated a good adjuvant role in the clinical scenario. Trials that used MSCs combined with another, primary treatment (eg, endobronchial valves) found that patients derived greater benefit in pulmonary function tests and/or quality of life reports, as well as reductions in systemic markers of inflammation. The present review summarizes and describes the more recent preclinical studies that have been published about MSC therapy for COPD/emphysema and discusses what has already been applied about MSCs treatment in COPD patients in the clinical setting.
Collapse
Affiliation(s)
- Mariana A Antunes
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro (UFRJ), RJ, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - José Roberto Lapa E Silva
- Institute of Thoracic Medicine, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Patricia Rm Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro (UFRJ), RJ, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
225
|
Wagenaar N, Nijboer CH, van Bel F. Repair of neonatal brain injury: bringing stem cell-based therapy into clinical practice. Dev Med Child Neurol 2017; 59:997-1003. [PMID: 28786482 DOI: 10.1111/dmcn.13528] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/07/2017] [Indexed: 12/15/2022]
Abstract
Hypoxic-ischaemic brain injury is one of most important causes of neonatal mortality and long-term neurological morbidity in infants born at term. At present, only hypothermia in infants with perinatal hypoxic-ischaemic encephalopathy has shown benefit as a neuroprotective strategy. Otherwise, current treatment options for neonatal brain injury mainly focus on controlling (associated) symptoms. Regeneration of the injured neonatal brain with stem cell-based therapies is emerging and experimental results are promising. At present, increasing efforts are made to bring stem cell-based therapies to the clinic. Among all progenitor cell types, mesenchymal stromal (stem) cells seem to be most promising for human use given their neuroregenerative properties and favourable safety profile. This review summarizes the actual state, potential hurdles and possibilities of stem cell-based therapy for neonatal brain injury in the clinical setting. An early version of this paper was presented at the Groningen Early Intervention Meeting which was held in April 2016.
Collapse
Affiliation(s)
- Nienke Wagenaar
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Cora H Nijboer
- Laboratory of NeuroImmunology and Developmental Origins of Disease, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Frank van Bel
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| |
Collapse
|
226
|
Mesenchymal Stem Cells in Sepsis and Associated Organ Dysfunction: A Promising Future or Blind Alley? Stem Cells Int 2017; 2017:7304121. [PMID: 29098010 PMCID: PMC5618761 DOI: 10.1155/2017/7304121] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 08/06/2017] [Indexed: 12/17/2022] Open
Abstract
Sepsis, newly defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection, is the most common cause of death in ICUs and one of the principal causes of death worldwide. Although substantial progress has been made in the understanding of fundamental mechanisms of sepsis, translation of these advances into clinically effective therapies has been disappointing. Given the extreme complexity of sepsis pathogenesis, the paradigm “one disease, one drug” is obviously flawed and combinations of multiple targets that involve early immunomodulation and cellular protection are needed. In this context, the immune-reprogramming properties of cell-based therapy using mesenchymal stem cells (MSC) represent an emerging therapeutic strategy in sepsis and associated organ dysfunction. This article provides an update of the current knowledge regarding MSC in preclinical models of sepsis and sepsis-induced acute kidney injury. Recommendations for further translational research in this field are discussed.
Collapse
|
227
|
Chang JC, Hoel F, Liu KH, Wei YH, Cheng FC, Kuo SJ, Tronstad KJ, Liu CS. Peptide-mediated delivery of donor mitochondria improves mitochondrial function and cell viability in human cybrid cells with the MELAS A3243G mutation. Sci Rep 2017; 7:10710. [PMID: 28878349 PMCID: PMC5587702 DOI: 10.1038/s41598-017-10870-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 08/16/2017] [Indexed: 02/07/2023] Open
Abstract
The cell penetrating peptide, Pep-1, has been shown to facilitate cellular uptake of foreign mitochondria but further research is required to evaluate the use of Pep-1-mediated mitochondrial delivery (PMD) in treating mitochondrial defects. Presently, we sought to determine whether mitochondrial transplantation rescue mitochondrial function in a cybrid cell model of mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) disease. Following PMD, recipient cells had internalized donor mitochondria after 1 h, and expressed higher levels of normal mitochondrial DNA, particularly at the end of the treatment and 11 days later. After 4 days, mitochondrial respiratory function had recovered and biogenesis was evident in the Pep-1 and PMD groups, compared to the untreated MELAS group. However, only PMD was able to reverse the fusion-to-fission ratio of mitochondrial morphology, and mitochondria shaping proteins resembled the normal pattern seen in the control group. Cell survival following hydrogen peroxide-induced oxidative stress was also improved in the PMD group. Finally, we observed that PMD partially normalized cytokine expression, including that of interleukin (IL)-7, granulocyte macrophage–colony-stimulating factor (GM-CSF), and vascular endothelial growth factor (VEGF), in the MELAS cells. Presently, our data further confirm the protective effects of PMD as well in MELAS disease.
Collapse
Affiliation(s)
- Jui-Chih Chang
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Fredrik Hoel
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Ko-Hung Liu
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Yau-Huei Wei
- Department of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan.,Department of Medicine, Mackay Medical College, Taipei, Taiwan
| | - Fu-Chou Cheng
- Stem Cell Center, Department of Medical Research, Taichung Veterans General Hospital, Changhua, Taiwan
| | - Shou-Jen Kuo
- Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | | | - Chin-San Liu
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan. .,Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan.
| |
Collapse
|
228
|
Tunneling nanotubes promote intercellular mitochondria transfer followed by increased invasiveness in bladder cancer cells. Oncotarget 2017; 8:15539-15552. [PMID: 28107184 PMCID: PMC5362504 DOI: 10.18632/oncotarget.14695] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 12/27/2016] [Indexed: 01/15/2023] Open
Abstract
Intercellular transfer of organelles via tunneling nanotubes (TNTs) is a novel means of cell-to-cell communication. Here we demonstrate the existence of TNTs between co-cultured RT4 and T24 bladder cancer cells using light microscopy, fluorescence imaging, and scanning electron microscopy (SEM). Spontaneous unidirectional transfer of mitochondria from T24 to RT4 cells was detected using fluorescence imaging and flow cytometry. The distribution of mitochondria migrated from T24 cells was in good agreement with the original mitochondria in RT4 cells, which may imply mitochondrial fusion. We detected cytoskeleton reconstruction in RT4-Mito-T24 cells by observing F-actin redistribution. Akt, mTOR, and their downstream mediators were activated and increased. The resultant increase in the invasiveness of bladder cancer cells was detected in vitro and in vivo. These data indicate that TNTs promote intercellular mitochondrial transfer between heterogeneous cells, followed by an increase in the invasiveness of bladder cancer cells.
Collapse
|
229
|
Raik S, Kumar A, Bhattacharyya S. Insights into cell-free therapeutic approach: Role of stem cell "soup-ernatant". Biotechnol Appl Biochem 2017; 65:104-118. [PMID: 28321921 DOI: 10.1002/bab.1561] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/02/2017] [Indexed: 12/16/2022]
Abstract
Current advances in medicine have revolutionized the field of regenerative medicine dramatically with newly evolved therapies for repair or replacement of degenerating or injured tissues. Stem cells (SCs) can be harvested from different sources for clinical therapeutics, which include fetal tissues, umbilical cord blood, embryos, and adult tissues. SCs can be isolated and differentiated into desired lineages for tissue regeneration and cell replacement therapy. However, several loopholes need to be addressed properly before this can be extended for large-scale therapeutic application. These include a careful approach for patient safety during SC treatments and tolerance of recipients. SC treatments are associated with a number of risk factors and require successful integration and survival of transplanted cells in the desired microenvironment with concurrent tissue regeneration. Recent studies have focused on developing alternatives that can replace the cell-based therapy using paracrine factors. The development of stem "cell free" therapies can be devoted mainly to the use of soluble factors (secretome), extracellular vesicles, and mitochondrial transfer. The present review emphasizes on the paradigms related to the use of SC-based therapeutics and the potential applications of a cell-free approach as an alternative to cell-based therapy in the area of regenerative medicine.
Collapse
Affiliation(s)
- Shalini Raik
- Department of Biophysics, PGIMER, Chandigarh, India
| | - Ajay Kumar
- Department of Biophysics, PGIMER, Chandigarh, India
| | | |
Collapse
|
230
|
Nawaz M, Fatima F. Extracellular Vesicles, Tunneling Nanotubes, and Cellular Interplay: Synergies and Missing Links. Front Mol Biosci 2017; 4:50. [PMID: 28770210 PMCID: PMC5513920 DOI: 10.3389/fmolb.2017.00050] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 07/03/2017] [Indexed: 12/15/2022] Open
Abstract
The process of intercellular communication seems to have been a highly conserved evolutionary process. Higher eukaryotes use several means of intercellular communication to address both the changing physiological demands of the body and to fight against diseases. In recent years, there has been an increasing interest in understanding how cell-derived nanovesicles, known as extracellular vesicles (EVs), can function as normal paracrine mediators of intercellular communication, but can also elicit disease progression and may be used for innovative therapies. Over the last decade, a large body of evidence has accumulated to show that cells use cytoplasmic extensions comprising open-ended channels called tunneling nanotubes (TNTs) to connect cells at a long distance and facilitate the exchange of cytoplasmic material. TNTs are a different means of communication to classical gap junctions or cell fusions; since they are characterized by long distance bridging that transfers cytoplasmic organelles and intracellular vesicles between cells and represent the process of heteroplasmy. The role of EVs in cell communication is relatively well-understood, but how TNTs fit into this process is just emerging. The aim of this review is to describe the relationship between TNTs and EVs, and to discuss the synergies between these two crucial processes in the context of normal cellular cross-talk, physiological roles, modulation of immune responses, development of diseases, and their combinatory effects in tissue repair. At the present time this review appears to be the first summary of the implications of the overlapping roles of TNTs and EVs. We believe that a better appreciation of these parallel processes will improve our understanding on how these nanoscale conduits can be utilized as novel tools for targeted therapies.
Collapse
Affiliation(s)
- Muhammad Nawaz
- Department of Pathology and Forensic Medicine, Ribeirao Preto Medical School, University of São PauloSão Paulo, Brazil.,Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Farah Fatima
- Department of Pathology and Forensic Medicine, Ribeirao Preto Medical School, University of São PauloSão Paulo, Brazil
| |
Collapse
|
231
|
Leaw B, Nair S, Lim R, Thornton C, Mallard C, Hagberg H. Mitochondria, Bioenergetics and Excitotoxicity: New Therapeutic Targets in Perinatal Brain Injury. Front Cell Neurosci 2017; 11:199. [PMID: 28747873 PMCID: PMC5506196 DOI: 10.3389/fncel.2017.00199] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 06/26/2017] [Indexed: 12/30/2022] Open
Abstract
Injury to the fragile immature brain is implicated in the manifestation of long-term neurological disorders, including childhood disability such as cerebral palsy, learning disability and behavioral disorders. Advancements in perinatal practice and improved care mean the majority of infants suffering from perinatal brain injury will survive, with many subtle clinical symptoms going undiagnosed until later in life. Hypoxic-ischemia is the dominant cause of perinatal brain injury, and constitutes a significant socioeconomic burden to both developed and developing countries. Therapeutic hypothermia is the sole validated clinical intervention to perinatal asphyxia; however it is not always neuroprotective and its utility is limited to developed countries. There is an urgent need to better understand the molecular pathways underlying hypoxic-ischemic injury to identify new therapeutic targets in such a small but critical therapeutic window. Mitochondria are highly implicated following ischemic injury due to their roles as the powerhouse and main energy generators of the cell, as well as cell death processes. While the link between impaired mitochondrial bioenergetics and secondary energy failure following loss of high-energy phosphates is well established after hypoxia-ischemia (HI), there is emerging evidence that the roles of mitochondria in disease extend far beyond this. Indeed, mitochondrial turnover, including processes such as mitochondrial biogenesis, fusion, fission and mitophagy, affect recovery of neurons after injury and mitochondria are involved in the regulation of the innate immune response to inflammation. This review article will explore these mitochondrial pathways, and finally will summarize past and current efforts in targeting these pathways after hypoxic-ischemic injury, as a means of identifying new avenues for clinical intervention.
Collapse
Affiliation(s)
- Bryan Leaw
- The Ritchie Centre, Hudson Institute of Medical ResearchClayton, VIC, Australia
| | - Syam Nair
- Perinatal Center, Institute of Physiology and Neuroscience, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Rebecca Lim
- The Ritchie Centre, Hudson Institute of Medical ResearchClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University ClaytonClayton, VIC, Australia
| | - Claire Thornton
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' HospitalLondon, United Kingdom
| | - Carina Mallard
- Perinatal Center, Institute of Physiology and Neuroscience, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Henrik Hagberg
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' HospitalLondon, United Kingdom.,Perinatal Center, Department of Clinical Sciences, Sahlgrenska Academy, Gothenburg UniversityGothenburg, Sweden
| |
Collapse
|
232
|
Modulation of oxidative phosphorylation and redox homeostasis in mitochondrial NDUFS4 deficiency via mesenchymal stem cells. Stem Cell Res Ther 2017. [PMID: 28646906 PMCID: PMC5482938 DOI: 10.1186/s13287-017-0601-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Disorders of the oxidative phosphorylation (OXPHOS) system represent a large group among the inborn errors of metabolism. The most frequently observed biochemical defect is isolated deficiency of mitochondrial complex I (CI). No effective treatment strategies for CI deficiency are so far available. The purpose of this study was to investigate whether and how mesenchymal stem cells (MSCs) are able to modulate metabolic function in fibroblast cell models of CI deficiency. Methods We used human and murine fibroblasts with a defect in the nuclear DNA encoded NDUFS4 subunit of CI. Fibroblasts were co-cultured with MSCs under different stress conditions and intercellular mitochondrial transfer was assessed by flow cytometry and fluorescence microscopy. Reactive oxygen species (ROS) levels were measured using MitoSOX-Red. Protein levels of CI were analysed by blue native polyacrylamide gel electrophoresis (BN-PAGE). Results Direct cellular interactions and mitochondrial transfer between MSCs and human as well as mouse fibroblast cell lines were demonstrated. Mitochondrial transfer was visible in 13.2% and 6% of fibroblasts (e.g. fibroblasts containing MSC mitochondria) for human and mouse cell lines, respectively. The transfer rate could be further stimulated via treatment of cells with TNF-α. MSCs effectively lowered cellular ROS production in NDUFS4-deficient fibroblast cell lines (either directly via co-culture or indirectly via incubation of cell lines with cell-free MSC supernatant). However, CI protein expression and activity were not rescued by MSC treatment. Conclusion This study demonstrates the interplay between MSCs and fibroblast cell models of isolated CI deficiency including transfer of mitochondria as well as modulation of cellular ROS levels. Further exploration of these cellular interactions might help to develop MSC-based treatment strategies for human CI deficiency. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0601-7) contains supplementary material, which is available to authorized users.
Collapse
|
233
|
Tadokoro R, Takahashi Y. Intercellular transfer of organelles during body pigmentation. Curr Opin Genet Dev 2017; 45:132-138. [PMID: 28605672 DOI: 10.1016/j.gde.2017.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 05/29/2017] [Accepted: 05/29/2017] [Indexed: 01/16/2023]
Abstract
The intercellular transfer of the melanin-producing organelle, called melanosome, from melanocytes to adjacent keratinocytes, is largely responsible for the coat colors and skin pigmentation of amniotes (birds, reptiles, and mammals). Although several hypotheses of melanin-transfer were proposed mainly by in vitro studies and electron microscopies, how the melanosome transfer takes place in the actual skin remained unclear. With advances in technologies of gene manipulations and high-resolution microscopy that allow direct visualization of plasma membrane, we are beginning to understand the amazing behaviors and dynamics of melanocytes. Studies in melanosome transfer further provide a clue to understand a general principle of intercellular organelle transport, including the intercellular translocations of mitochondria.
Collapse
Affiliation(s)
- Ryosuke Tadokoro
- Department of Zoology, Graduate School of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto 606-8502, Japan
| | - Yoshiko Takahashi
- Department of Zoology, Graduate School of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto 606-8502, Japan; AMED Core Research for Evolutional Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development (AMED), Chiyoda-ku, Tokyo 100-0004, Japan.
| |
Collapse
|
234
|
Mao F, Tu Q, Wang L, Chu F, Li X, Li HS, Xu W. Mesenchymal stem cells and their therapeutic applications in inflammatory bowel disease. Oncotarget 2017; 8:38008-38021. [PMID: 28402942 PMCID: PMC5514968 DOI: 10.18632/oncotarget.16682] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/06/2017] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem or stromal cells (MSCs) are non-hematopoietic stem cells that facilitate tissue regeneration through mechanisms involving self-renewal and differentiation, supporting angiogenesis and tissue cell survival, and limiting inflammation. MSCs were originally identified and expanded in long-term cultures of cells from bone marrow and other organs; and their native identity was recently confined into pericytes and adventitial cells in vascularized tissue. The multipotency, as well as the trophic and immunosuppressive effects, of MSCs have prompted the rapid development of clinical applications for many diseases involving tissue inflammation and immune disorders, including inflammatory bowel disease. Although standard criteria have been established to define MSCs, their therapeutic efficacy has varied significantly among studies due to their natural heterogenicity. Thus, understanding the biological and immunological features of MSCs is critical to standardize and optimize MSCs-based therapy. In this review, we highlight the cellular and molecular mechanisms involved in MSCs-mediated tissue repair and immunosuppression. We also provide an update on the current development of MSCs-based clinical trials, with a detailed discussion of MSC-based cell therapy in inflammatory bowel disease.
Collapse
Affiliation(s)
- Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Qiang Tu
- Jiangning Hospital of Nanjing, Nanjing, Jiangsu, P.R. China
| | - Li Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Fuliang Chu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xia Li
- Department of Gastroenterology, Binzhou Medical University Yantai Affiliated Hospital, Yantai, Shandong, P.R. China
| | - Haiyan S. Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wenrong Xu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| |
Collapse
|
235
|
Cell Connections by Tunneling Nanotubes: Effects of Mitochondrial Trafficking on Target Cell Metabolism, Homeostasis, and Response to Therapy. Stem Cells Int 2017; 2017:6917941. [PMID: 28659978 PMCID: PMC5474251 DOI: 10.1155/2017/6917941] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/21/2017] [Accepted: 03/30/2017] [Indexed: 01/16/2023] Open
Abstract
Intercellular communications play a major role in tissue homeostasis and responses to external cues. Novel structures for this communication have recently been described. These tunneling nanotubes (TNTs) consist of thin-extended membrane protrusions that connect cells together. TNTs allow the cell-to-cell transfer of various cellular components, including proteins, RNAs, viruses, and organelles, such as mitochondria. Mesenchymal stem cells (MSCs) are both naturally present and recruited to many different tissues where their interaction with resident cells via secreted factors has been largely documented. Their immunosuppressive and repairing capacities constitute the basis for many current clinical trials. MSCs recruited to the tumor microenvironment also play an important role in tumor progression and resistance to therapy. MSCs are now the focus of intense scrutiny due to their capacity to form TNTs and transfer mitochondria to target cells, either in normal physiological or in pathological conditions, leading to changes in cell energy metabolism and functions, as described in this review.
Collapse
|
236
|
Mahrouf-Yorgov M, Augeul L, Da Silva CC, Jourdan M, Rigolet M, Manin S, Ferrera R, Ovize M, Henry A, Guguin A, Meningaud JP, Dubois-Randé JL, Motterlini R, Foresti R, Rodriguez AM. Mesenchymal stem cells sense mitochondria released from damaged cells as danger signals to activate their rescue properties. Cell Death Differ 2017; 24:1224-1238. [PMID: 28524859 PMCID: PMC5520168 DOI: 10.1038/cdd.2017.51] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 03/05/2017] [Accepted: 03/07/2017] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) protect tissues against cell death induced by ischemia/reperfusion insults. This therapeutic effect seems to be controlled by physiological cues released by the local microenvironment following injury. Recent lines of evidence indicate that MSC can communicate with their microenvironment through bidirectional exchanges of mitochondria. In particular, in vitro and in vivo studies report that MSCs rescue injured cells through delivery of their own mitochondria. However, the role of mitochondria conveyed from somatic cells to MSC remains unknown. By using a co-culture system consisting of MSC and distressed somatic cells such as cardiomyocytes or endothelial cells, we showed that mitochondria from suffering cells acted as danger-signaling organelles that triggered the anti-apoptotic function of MSC. We demonstrated that foreign somatic-derived mitochondria were engulfed and degraded by MSC, leading to induction of the cytoprotective enzyme heme oxygenase-1 (HO-1) and stimulation of mitochondrial biogenesis. As a result, the capacity of MSC to donate their mitochondria to injured cells to combat oxidative stress injury was enhanced. We found that similar mechanisms - activation of autophagy, HO-1 and mitochondrial biogenesis - occurred after exposure of MSC to exogenous mitochondria isolated from somatic cells, strengthening the idea that somatic mitochondria alert MSC of a danger situation and subsequently promote an adaptive reparative response. In addition, the cascade of events triggered by the transfer of somatic mitochondria into MSC was recapitulated in a model of myocardial infarction in vivo. Specifically, MSC engrafted into infarcted hearts of mice reduced damage, upregulated HO-1 and increased mitochondrial biogenesis, while inhibition of mitophagy or HO-1 failed to protect against cardiac apoptosis. In conclusion, our study reveals a new facet about the role of mitochondria released from dying cells as a key environmental cue that controls the cytoprotective function of MSC and opens novel avenues to improve the effectiveness of MSC-based therapies.
Collapse
Affiliation(s)
- Meriem Mahrouf-Yorgov
- Université Paris-Est, UMR-S955, UPEC, Créteil, Paris, France.,INSERM, Unité 955 Team 12, Créteil, Paris, France
| | - Lionel Augeul
- INSERM UMR-1060, Laboratoire CarMeN, Université Lyon 1, Faculté de Médecine, Rockefeller, Lyon, France
| | - Claire Crola Da Silva
- INSERM UMR-1060, Laboratoire CarMeN, Université Lyon 1, Faculté de Médecine, Rockefeller, Lyon, France
| | - Maud Jourdan
- Université Paris-Est, UMR-S955, UPEC, Créteil, Paris, France.,INSERM, Unité 955 Team 12, Créteil, Paris, France
| | - Muriel Rigolet
- Université Paris-Est, UMR-S955, UPEC, Créteil, Paris, France.,INSERM U955 Team 10, Créteil, Paris, France
| | - Sylvie Manin
- Université Paris-Est, UMR-S955, UPEC, Créteil, Paris, France.,INSERM, Unité 955 Team 12, Créteil, Paris, France
| | - René Ferrera
- INSERM UMR-1060, Laboratoire CarMeN, Université Lyon 1, Faculté de Médecine, Rockefeller, Lyon, France
| | - Michel Ovize
- INSERM UMR-1060, Laboratoire CarMeN, Université Lyon 1, Faculté de Médecine, Rockefeller, Lyon, France.,Hospices Civils de Lyon, Hôpital Louis Pradel, Service d'Explorations Fonctionnelles, Cardiovasculaires and Centre d'Investigation Clinique, Lyon, France
| | - Adeline Henry
- Université Paris-Est, UMR-S955, UPEC, Créteil, Paris, France.,INSERM U955, Plateforme de Cytométrie en flux, Créteil, Paris, France
| | - Aurélie Guguin
- Université Paris-Est, UMR-S955, UPEC, Créteil, Paris, France.,INSERM U955, Plateforme de Cytométrie en flux, Créteil, Paris, France
| | - Jean-Paul Meningaud
- Service de Chirurgie Plastique et Maxillo-Faciale, AP-HP, Hôpital Henri Mondor-A. Chenevier, Créteil, Paris, France
| | - Jean-Luc Dubois-Randé
- Université Paris-Est, UMR-S955, UPEC, Créteil, Paris, France.,Fédération de Cardiologie, AP-HP, Hôpital Henri Mondor-A. Chenevier, Créteil, Paris, France
| | - Roberto Motterlini
- Université Paris-Est, UMR-S955, UPEC, Créteil, Paris, France.,INSERM, Unité 955 Team 12, Créteil, Paris, France
| | - Roberta Foresti
- Université Paris-Est, UMR-S955, UPEC, Créteil, Paris, France.,INSERM, Unité 955 Team 12, Créteil, Paris, France
| | - Anne-Marie Rodriguez
- Université Paris-Est, UMR-S955, UPEC, Créteil, Paris, France.,INSERM, Unité 955 Team 12, Créteil, Paris, France
| |
Collapse
|
237
|
Nzigou Mombo B, Gerbal-Chaloin S, Bokus A, Daujat-Chavanieu M, Jorgensen C, Hugnot JP, Vignais ML. MitoCeption: Transferring Isolated Human MSC Mitochondria to Glioblastoma Stem Cells. J Vis Exp 2017. [PMID: 28287607 DOI: 10.3791/55245] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mitochondria play a central role for cell metabolism, energy production and control of apoptosis. Inadequate mitochondrial function has been found responsible for very diverse diseases, ranging from neurological pathologies to cancer. Interestingly, mitochondria have recently been shown to display the capacity to be transferred between cell types, notably from human mesenchymal stem cells (MSC) to cancer cells in coculture conditions, with metabolic and functional consequences for the mitochondria recipient cells, further enhancing the current interest for the biological properties of these organelles. Evaluating the effects of the transferred MSC mitochondria in the target cells is of primary importance to understand the biological outcome of such cell-cell interactions. The MitoCeption protocol described here allows the transfer of the mitochondria isolated beforehand from the donor cells to the target cells, using MSC mitochondria and glioblastoma stem cells (GSC) as a model system. This protocol has previously been used to transfer mitochondria, isolated from MSCs, to adherent MDA-MB-231 cancer cells. This mitochondria transfer protocol is adapted here for GSCs that present the specific particularity of growing as neurospheres in vitro. The transfer of the isolated mitochondria can be followed by fluorescence-activated cell sorting (FACS) and confocal imaging using mitochondria vital dyes. The use of mitochondria donor and target cells with distinct haplotypes (SNPs) also allows detection of the transferred mitochondria based on the concentration of their circular mitochondrial DNA (mtDNA) in the target cells. Once the protocol has been validated with these criteria, the cells harboring the transferred mitochondria can be further analyzed to determine the effects of the exogenous mitochondria on biological properties such as cell metabolism, plasticity, proliferation and response to therapy.
Collapse
Affiliation(s)
- Brice Nzigou Mombo
- Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Montpellier University
| | - Sabine Gerbal-Chaloin
- Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Montpellier University
| | - Aleksandra Bokus
- Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Montpellier University
| | | | - Christian Jorgensen
- Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Montpellier University
| | - Jean-Philippe Hugnot
- Institute of Neurosciences of Montpellier (INM), INSERM U1051, Montpellier University
| | - Marie-Luce Vignais
- Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Montpellier University;
| |
Collapse
|
238
|
Herst PM, Rowe MR, Carson GM, Berridge MV. Functional Mitochondria in Health and Disease. Front Endocrinol (Lausanne) 2017; 8:296. [PMID: 29163365 PMCID: PMC5675848 DOI: 10.3389/fendo.2017.00296] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 10/16/2017] [Indexed: 01/10/2023] Open
Abstract
The ability to rapidly adapt cellular bioenergetic capabilities to meet rapidly changing environmental conditions is mandatory for normal cellular function and for cancer progression. Any loss of this adaptive response has the potential to compromise cellular function and render the cell more susceptible to external stressors such as oxidative stress, radiation, chemotherapeutic drugs, and hypoxia. Mitochondria play a vital role in bioenergetic and biosynthetic pathways and can rapidly adjust to meet the metabolic needs of the cell. Increased demand is met by mitochondrial biogenesis and fusion of individual mitochondria into dynamic networks, whereas a decrease in demand results in the removal of superfluous mitochondria through fission and mitophagy. Effective communication between nucleus and mitochondria (mito-nuclear cross talk), involving the generation of different mitochondrial stress signals as well as the nuclear stress response pathways to deal with these stressors, maintains bioenergetic homeostasis under most conditions. However, when mitochondrial DNA (mtDNA) mutations accumulate and mito-nuclear cross talk falters, mitochondria fail to deliver critical functional outputs. Mutations in mtDNA have been implicated in neuromuscular and neurodegenerative mitochondriopathies and complex diseases such as diabetes, cardiovascular diseases, gastrointestinal disorders, skin disorders, aging, and cancer. In some cases, drastic measures such as acquisition of new mitochondria from donor cells occurs to ensure cell survival. This review starts with a brief discussion of the evolutionary origin of mitochondria and summarizes how mutations in mtDNA lead to mitochondriopathies and other degenerative diseases. Mito-nuclear cross talk, including various stress signals generated by mitochondria and corresponding stress response pathways activated by the nucleus are summarized. We also introduce and discuss a small family of recently discovered hormone-like mitopeptides that modulate body metabolism. Under conditions of severe mitochondrial stress, mitochondria have been shown to traffic between cells, replacing mitochondria in cells with damaged and malfunctional mtDNA. Understanding the processes involved in cellular bioenergetics and metabolic adaptation has the potential to generate new knowledge that will lead to improved treatment of many of the metabolic, degenerative, and age-related inflammatory diseases that characterize modern societies.
Collapse
Affiliation(s)
- Patries M. Herst
- Cancer Cell Biology, Malaghan Institute of Medical Research, Wellington, New Zealand
- Department of Radiation Therapy, University of Otago, Wellington, New Zealand
- *Correspondence: Patries M. Herst, ; Michael V. Berridge,
| | - Matthew R. Rowe
- School of Biological Sciences, Victoria University, Wellington, New Zealand
| | - Georgia M. Carson
- Cancer Cell Biology, Malaghan Institute of Medical Research, Wellington, New Zealand
- School of Biological Sciences, Victoria University, Wellington, New Zealand
| | - Michael V. Berridge
- Cancer Cell Biology, Malaghan Institute of Medical Research, Wellington, New Zealand
- *Correspondence: Patries M. Herst, ; Michael V. Berridge,
| |
Collapse
|
239
|
Jackson MV, Krasnodembskaya AD. Analysis of Mitochondrial Transfer in Direct Co-cultures of Human Monocyte-derived Macrophages (MDM) and Mesenchymal Stem Cells (MSC). Bio Protoc 2017; 7:e2255. [PMID: 28534038 DOI: 10.21769/bioprotoc.2255] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSC) are adult stem cells which have been shown to improve survival, enhance bacterial clearance and alleviate inflammation in pre-clinical models of acute respiratory distress syndrome (ARDS) and sepsis. These diseases are characterised by uncontrolled inflammation often underpinned by bacterial infection. The mechanisms of MSC immunomodulatory effects are not fully understood yet. We sought to investigate MSC cell contact-dependent communication with alveolar macrophages (AM), professional phagocytes which play an important role in the lung inflammatory responses and anti-bacterial defence. With the use of a basic direct co-culture system, confocal microscopy and flow cytometry we visualised and effectively quantified MSC mitochondrial transfer to AM through tunnelling nanotubes (TNT). To model the human AM, primary monocytes were isolated from human donor blood and differentiated into macrophages (monocyte derived macrophages, MDM) in the presence of granulocyte macrophage colony-stimulating factor (GM-CSF), thus allowing adaptation of an AM-like phenotype (de Almeida et al., 2000; Guilliams et al., 2013). Human bone-marrow derived MSC, were labelled with mitochondria-specific fluorescent stain, washed extensively, seeded into the tissue culture plate with MDMs at the ratio of 1:20 (MSC/MDM) and co-cultured for 24 h. TNT formation and mitochondrial transfer were visualised by confocal microscopy and semi-quantified by flow cytometry. By using the method we described here we established that MSC use TNTs as the means to transfer mitochondria to macrophages. Further studies demonstrated that mitochondrial transfer enhances macrophage oxidative phosphorylation and phagocytosis. When TNT formation was blocked by cytochalasin B, MSC effect on macrophage phagocytosis was completely abrogated. This is the first study to demonstrate TNT-mediated mitochondrial transfer from MSC to innate immune cells.
Collapse
Affiliation(s)
- Megan V Jackson
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland
| | - Anna D Krasnodembskaya
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland
| |
Collapse
|
240
|
Cencioni C, Atlante S, Savoia M, Martelli F, Farsetti A, Capogrossi MC, Zeiher AM, Gaetano C, Spallotta F. The double life of cardiac mesenchymal cells: Epimetabolic sensors and therapeutic assets for heart regeneration. Pharmacol Ther 2016; 171:43-55. [PMID: 27742569 DOI: 10.1016/j.pharmthera.2016.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Organ-specific mesenchymal cells naturally reside in the stroma, where they are exposed to some environmental variables affecting their biology and functions. Risk factors such as diabetes or aging influence their adaptive response. In these cases, permanent epigenetic modifications may be introduced in the cells with important consequences on their local homeostatic activity and therapeutic potential. Numerous results suggest that mesenchymal cells, virtually present in every organ, may contribute to tissue regeneration mostly by paracrine mechanisms. Intriguingly, the heart is emerging as a source of different cells, including pericytes, cardiac progenitors, and cardiac fibroblasts. According to phenotypic, functional, and molecular criteria, these should be classified as mesenchymal cells. Not surprisingly, in recent years, the attention on these cells as therapeutic tools has grown exponentially, although only very preliminary data have been obtained in clinical trials to date. In this review, we summarized the state of the art about the phenotypic features, functions, regenerative properties, and clinical applicability of mesenchymal cells, with a particular focus on those of cardiac origin.
Collapse
Affiliation(s)
- Chiara Cencioni
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Sandra Atlante
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Matteo Savoia
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Universitá Cattolica, Institute of Medical Pathology, 00138 Rome, Italy; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, San Donato Milanese, Milan 20097, Italy.
| | - Antonella Farsetti
- Consiglio Nazionale delle Ricerche, Istituto di Biologia Cellulare e Neurobiologia, Roma, Italy; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Maurizio C Capogrossi
- Laboratorio di Patologia Vascolare, Istituto Dermopatico dell'Immacolata, Roma, Italy.
| | - Andreas M Zeiher
- Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Carlo Gaetano
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Francesco Spallotta
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| |
Collapse
|
241
|
Torralba D, Baixauli F, Sánchez-Madrid F. Mitochondria Know No Boundaries: Mechanisms and Functions of Intercellular Mitochondrial Transfer. Front Cell Dev Biol 2016; 4:107. [PMID: 27734015 PMCID: PMC5039171 DOI: 10.3389/fcell.2016.00107] [Citation(s) in RCA: 291] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/14/2016] [Indexed: 12/25/2022] Open
Abstract
Mitochondria regulate multiple cell processes, including calcium signaling, apoptosis and cell metabolism. Mitochondria contain their own circular genome encoding selected subunits of the oxidative phosphorylation complexes. Recent findings reveal that, in addition to being maternally inherited, mitochondria can traverse cell boundaries and thus be horizontally transferred between cells. Although, the physiological relevance of this phenomenon is still under debate, mitochondria uptake rescues mitochondrial respiration defects in recipient cells and regulates signaling, proliferation or chemotherapy resistance in vitro and in vivo. In this review, we outline the pathophysiological consequences of horizontal mitochondrial transfer and offer a perspective on the cellular and molecular mechanisms mediating their intercellular transmission, including tunneling nanotubes, extracellular vesicles, cellular fusion, and GAP junctions. The physiological relevance of mitochondrial transfer and the potential therapeutic application of this exchange for treating mitochondrial-related diseases are discussed.
Collapse
Affiliation(s)
- Daniel Torralba
- Signaling and Inflammation Program, Centro Nacional Investigaciones CardiovascularesMadrid, Spain; Servicio de Inmunología, Instituto Investigación Sanitaria Princesa, Universidad Autonoma de MadridMadrid, Spain
| | - Francesc Baixauli
- Signaling and Inflammation Program, Centro Nacional Investigaciones CardiovascularesMadrid, Spain; Servicio de Inmunología, Instituto Investigación Sanitaria Princesa, Universidad Autonoma de MadridMadrid, Spain
| | - Francisco Sánchez-Madrid
- Signaling and Inflammation Program, Centro Nacional Investigaciones CardiovascularesMadrid, Spain; Servicio de Inmunología, Instituto Investigación Sanitaria Princesa, Universidad Autonoma de MadridMadrid, Spain
| |
Collapse
|
242
|
Mesenchymal stem cell-conditioned media ameliorate diabetic endothelial dysfunction by improving mitochondrial bioenergetics via the Sirt1/AMPK/PGC-1α pathway. Clin Sci (Lond) 2016; 130:2181-2198. [PMID: 27613156 DOI: 10.1042/cs20160235] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 09/05/2016] [Indexed: 02/05/2023]
Abstract
Vasculopathy is a major complication of diabetes. Impaired mitochondrial bioenergetics and biogenesis due to oxidative stress are a critical causal factor for diabetic endothelial dysfunction. Sirt1, an NAD+-dependent enzyme, is known to play an important protective role through deacetylation of many substrates involved in oxidative phosphorylation and reactive oxygen species generation. Mesenchymal stem cell-conditioned medium (MSC-CM) has emerged as a promising cell-free therapy due to the trophic actions of mesenchymal stem cell (MSC)-secreted molecules. In the present study, we investigated the therapeutic potential of MSC-CMs in diabetic endothelial dysfunction, focusing on the Sirt1 signalling pathway and the relevance to mitochondrial function. We found that high glucose-stimulated MSC-CM attenuated several glucotoxicity-induced processes, oxidative stress and apoptosis of endothelial cells of the human umbilical vein. MSC-CM perfusion in diabetic rats ameliorated compromised aortic vasodilatation and alleviated oxidative stress in aortas. We further demonstrated that these effects were dependent on improved mitochondrial function and up-regulation of Sirt1 expression. MSC-CMs activated the phosphorylation of phosphoinositide 3-kinase (PI3K) and protein kinase B (Akt), leading to direct interaction between Akt and Sirt1, and subsequently enhanced Sirt1 expression. In addition, both MSC-CM and Sirt1 activation could increase the expression of peroxisome proliferator-activated receptor γ co-activator-1α (PGC-1α), as well as increase the mRNA expression of its downstream, mitochondrial, biogenesis-related genes. This indirect regulation was mediated by activation of AMP-activated protein kinase (AMPK). Overall our findings indicated that MSC-CM had protective effects on endothelial cells, with respect to glucotoxicity, by ameliorating mitochondrial dysfunction via the PI3K/Akt/Sirt1 pathway, and Sirt1 potentiated mitochondrial biogenesis, through the Sirt1/AMPK/PGC-1α pathway.
Collapse
|
243
|
Doynova M, Berretta A, Jones M, Jasoni C, Vickers M, O'Sullivan J. Interactions between mitochondrial and nuclear DNA in mammalian cells are non-random. Mitochondrion 2016; 30:187-96. [DOI: 10.1016/j.mito.2016.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 06/29/2016] [Accepted: 08/04/2016] [Indexed: 12/18/2022]
|
244
|
Abstract
The past decade has seen an explosion of research directed toward better understanding of the mechanisms of mesenchymal stem/stromal cell (MSC) function during rescue and repair of injured organs and tissues. In addition to delineating cell–cell signaling and molecular controls for MSC differentiation, the field has made particular progress in defining several other mechanisms through which administered MSCs can promote tissue rescue/repair. These include: 1) paracrine activity that involves secretion of proteins/peptides and hormones; 2) transfer of mitochondria by way of tunneling nanotubes or microvesicles; and 3) transfer of exosomes or microvesicles containing RNA and other molecules. Improved understanding of MSC function holds great promise for the application of cell therapy and also for the development of powerful cell-derived therapeutics for regenerative medicine. Focusing on these three mechanisms, we discuss MSC-mediated effects on immune cell responses, cell survival, and fibrosis and review recent progress with MSC-based or MSC-derived therapeutics.
Collapse
Affiliation(s)
- Jeffrey L Spees
- University of Vermont, Burlington, VT, USA. .,Department of Medicine, Stem Cell Core, University of Vermont, 208 South Park Drive, Ste 2, Colchester, VT, 05446, USA.
| | - Ryang Hwa Lee
- Institute for Regenerative Medicine, Texas A & M University College of Medicine, 206 Olsen Blvd., Room 228, MS1114, College Station, TX, 77845, USA
| | - Carl A Gregory
- Institute for Regenerative Medicine, Texas A & M University College of Medicine, 206 Olsen Blvd., Room 228, MS1114, College Station, TX, 77845, USA.
| |
Collapse
|
245
|
Li G, Yu F, Lei T, Gao H, Li P, Sun Y, Huang H, Mu Q. Bone marrow mesenchymal stem cell therapy in ischemic stroke: mechanisms of action and treatment optimization strategies. Neural Regen Res 2016; 11:1015-24. [PMID: 27482235 PMCID: PMC4962565 DOI: 10.4103/1673-5374.184506] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Animal and clinical studies have confirmed the therapeutic effect of bone marrow mesenchymal stem cells on cerebral ischemia, but their mechanisms of action remain poorly understood. Here, we summarize the transplantation approaches, directional migration, differentiation, replacement, neural circuit reconstruction, angiogenesis, neurotrophic factor secretion, apoptosis, immunomodulation, multiple mechanisms of action, and optimization strategies for bone marrow mesenchymal stem cells in the treatment of ischemic stroke. We also explore the safety of bone marrow mesenchymal stem cell transplantation and conclude that bone marrow mesenchymal stem cell transplantation is an important direction for future treatment of cerebral ischemia. Determining the optimal timing and dose for the transplantation are important directions for future research.
Collapse
Affiliation(s)
- Guihong Li
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, Jilin Province, China; Department of Neurosurgery, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Fengbo Yu
- School of Pharmacy, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Ting Lei
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Haijun Gao
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Peiwen Li
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yuxue Sun
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Haiyan Huang
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qingchun Mu
- Department of Neurosurgery, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| |
Collapse
|
246
|
Scholkmann F. Long range physical cell-to-cell signalling via mitochondria inside membrane nanotubes: a hypothesis. Theor Biol Med Model 2016; 13:16. [PMID: 27267202 PMCID: PMC4896004 DOI: 10.1186/s12976-016-0042-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 05/27/2016] [Indexed: 02/07/2023] Open
Abstract
Coordinated interaction of single cells by cell-to-cell communication (signalling) enables complex behaviour necessary for the functioning of multicellular organisms. A quite newly discovered cell-to-cell signalling mechanism relies on nanotubular cell-co-cell connections, termed "membrane nanotubes" (MNTs). The present paper presents the hypothesis that mitochondria inside MNTs can form a connected structure (mitochondrial network) which enables the exchange of energy and signals between cells. It is proposed that two modes of energy and signal transmission may occur: electrical/electrochemical and electromagnetic (optical). Experimental work supporting the hypothesis is reviewed, and suggestions for future research regarding the discussed topic are given.
Collapse
Affiliation(s)
- Felix Scholkmann
- Biomedical Optics Research Laboratory, Department of Neonatology, University Hospital Zurich, University of Zurich, Frauenklinikstr. 10, 8091, Zurich, Switzerland.
- Research Office for Complex Physical and Biological Systems (ROCoS), Mutschellenstr. 179, 8038, Zurich, Switzerland.
| |
Collapse
|
247
|
Rustom A. The missing link: does tunnelling nanotube-based supercellularity provide a new understanding of chronic and lifestyle diseases? Open Biol 2016; 6:160057. [PMID: 27278648 PMCID: PMC4929939 DOI: 10.1098/rsob.160057] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 05/10/2016] [Indexed: 12/21/2022] Open
Abstract
Tunnelling nanotubes (TNTs) are increasingly recognized as central players in a multitude of cellular mechanisms and diseases. Although their existence and functions in animal organisms are still elusive, emerging evidence suggests that they are involved in developmental processes, tissue regeneration, viral infections or pathogen transfer, stem cell differentiation, immune responses as well as initiation and progression of neurodegenerative disorders and cancer (see Sisakhtnezhad & Khosravi 2015 Eur. J. Cell Biol. 94, 429-443. (doi:10.1016/j.ejcb.2015.06.010)). A broader field of vision, including their striking functional and structural resemblance with nanotube-mediated phenomena found throughout the phylogenetic tree, from plants down to bacteria, points to a universal, conserved and tightly regulated mechanism of cellular assemblies. Based on our initial definition of TNTs as open-ended channels mediating membrane continuity between connected cells (Rustom et al. 2004 Science 303, 1007-1010. (doi:10.1126/science.1093133)), it is suggested that animal tissues represent supercellular assemblies that-besides opening discrete communication pathways-balance diverse stress factors caused by pathological changes or fluctuating physiological and environmental conditions, such as oxidative stress or nutrient shortage. By combining current knowledge about nanotube formation, intercellular transfer and communication phenomena as well as associated molecular pathways, a model evolves, predicting that the linkage between reactive oxygen species, TNT-based supercellularity and the intercellular shuttling of materials will have significant impact on diverse body functions, such as cell survival, redox/metabolic homeostasis and mitochondrial heteroplasmy. It implies that TNTs are intimately linked to the physiological and pathological state of animal cells and represent a central joint element of diverse diseases, such as neurodegenerative disorders, diabetes or cancer.
Collapse
Affiliation(s)
- Amin Rustom
- Interdisciplinary Center for Neurosciences (IZN), Institute of Neurobiology, University of Heidelberg, INF 364, 69120 Heidelberg, Germany
| |
Collapse
|
248
|
Sinha P, Islam MN, Bhattacharya S, Bhattacharya J. Intercellular mitochondrial transfer: bioenergetic crosstalk between cells. Curr Opin Genet Dev 2016; 38:97-101. [PMID: 27235808 DOI: 10.1016/j.gde.2016.05.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 05/02/2016] [Accepted: 05/09/2016] [Indexed: 12/18/2022]
Abstract
Mitochondrial transfer from donor cells to cells of injured tissues is a promising cell-based therapy for effectively bringing about recovery of tissue bioenergetics. Here, we review recent studies on intercellular mitochondrial transfer in organs and cells. We also review studies that shed light on our current understanding of the known mechanisms and conditions that lead to intercellular mitochondrial transfer.
Collapse
Affiliation(s)
- Pratik Sinha
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Mohammad N Islam
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Sunita Bhattacharya
- Department of Medicine, Columbia University, New York, NY 10032, USA; Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Jahar Bhattacharya
- Department of Medicine, Columbia University, New York, NY 10032, USA; Department of Physiology & Cellular Biophysics, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
249
|
Berridge MV, McConnell MJ, Grasso C, Bajzikova M, Kovarova J, Neuzil J. Horizontal transfer of mitochondria between mammalian cells: beyond co-culture approaches. Curr Opin Genet Dev 2016; 38:75-82. [PMID: 27219870 DOI: 10.1016/j.gde.2016.04.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 03/23/2016] [Accepted: 04/05/2016] [Indexed: 12/21/2022]
Abstract
Current dogma holds that genes are the property of individual mammalian cells and partition between daughter cells during cell division. However, and rather unexpectedly, recent research has demonstrated horizontal cell-to-cell transfer of mitochondria and mitochondrial DNA in several mammalian cell culture systems. Furthermore, unequivocal evidence that mitochondrial DNA transfer occurs in vivo has now been published. While these studies show horizontal transfer of mitochondrial DNA in pathological settings, it is also possible that intercellular mitochondrial transfer is a fundamental physiological process with a role in development and tissue homeostasis.
Collapse
Affiliation(s)
- Michael V Berridge
- Cancer Cell and Molecular Biology Group, Malaghan Institute of Medical Research, Wellington 6012, New Zealand.
| | - Melanie J McConnell
- Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Carole Grasso
- Cancer Cell and Molecular Biology Group, Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Martina Bajzikova
- Molecular Therapy Group, Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic
| | - Jaromira Kovarova
- Molecular Therapy Group, Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic
| | - Jiri Neuzil
- Molecular Therapy Group, Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic; Mitochondria, Apoptosis and Cancer Research Group, School of Medical Science and Griffith Health Institute, Griffith University, Southport, QLD 4222, Australia.
| |
Collapse
|
250
|
Wu TH, Sagullo E, Case D, Zheng X, Li Y, Hong JS, TeSlaa T, Patananan AN, McCaffery JM, Niazi K, Braas D, Koehler CM, Graeber TG, Chiou PY, Teitell MA. Mitochondrial Transfer by Photothermal Nanoblade Restores Metabolite Profile in Mammalian Cells. Cell Metab 2016; 23:921-9. [PMID: 27166949 PMCID: PMC5062745 DOI: 10.1016/j.cmet.2016.04.007] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 02/10/2016] [Accepted: 04/12/2016] [Indexed: 12/17/2022]
Abstract
mtDNA sequence alterations are challenging to generate but desirable for basic studies and potential correction of mtDNA diseases. Here, we report a new method for transferring isolated mitochondria into somatic mammalian cells using a photothermal nanoblade, which bypasses endocytosis and cell fusion. The nanoblade rescued the pyrimidine auxotroph phenotype and respiration of ρ0 cells that lack mtDNA. Three stable isogenic nanoblade-rescued clones grown in uridine-free medium showed distinct bioenergetics profiles. Rescue lines 1 and 3 reestablished nucleus-encoded anapleurotic and catapleurotic enzyme gene expression patterns and had metabolite profiles similar to the parent cells from which the ρ0 recipient cells were derived. By contrast, rescue line 2 retained a ρ0 cell metabolic phenotype despite growth in uridine-free selection. The known influence of metabolite levels on cellular processes, including epigenome modifications and gene expression, suggests metabolite profiling can help assess the quality and function of mtDNA-modified cells.
Collapse
Affiliation(s)
- Ting-Hsiang Wu
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Enrico Sagullo
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Dana Case
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xin Zheng
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yanjing Li
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jason S Hong
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tara TeSlaa
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alexander N Patananan
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - J Michael McCaffery
- Integrated Imaging Center, Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kayvan Niazi
- NantWorks, LLC, Culver City, CA 90232, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Daniel Braas
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA Metabolomics Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Carla M Koehler
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA Metabolomics Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Pei-Yu Chiou
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Michael A Teitell
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|