101
|
Chen G, Kroemer G, Kepp O. Mitophagy: An Emerging Role in Aging and Age-Associated Diseases. Front Cell Dev Biol 2020; 8:200. [PMID: 32274386 PMCID: PMC7113588 DOI: 10.3389/fcell.2020.00200] [Citation(s) in RCA: 228] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial dysfunction constitutes one of the hallmarks of aging and is characterized by irregular mitochondrial morphology, insufficient ATP production, accumulation of mitochondrial DNA (mtDNA) mutations, increased production of mitochondrial reactive oxygen species (ROS) and the consequent oxidative damage to nucleic acids, proteins and lipids. Mitophagy, a mitochondrial quality control mechanism enabling the degradation of damaged and superfluous mitochondria, prevents such detrimental effects and reinstates cellular homeostasis in response to stress. To date, there is increasing evidence that mitophagy is significantly impaired in several human pathologies including aging and age-related diseases such as neurodegenerative disorders, cardiovascular pathologies and cancer. Therapeutic interventions aiming at the induction of mitophagy may have the potency to ameliorate these dysfunctions. In this review, we summarize recent findings on mechanisms controlling mitophagy and its role in aging and the development of human pathologies.
Collapse
Affiliation(s)
- Guo Chen
- The State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Guido Kroemer
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, UMR 1138, Centre de Recherche des Cordeliers, Paris, France
- Equipe 11 Labellisée par la Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Sorbonne Université, Paris, France
- Université Paris-Saclay, Faculté de Médecine, Kremlin-Bicêtre, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou, China
- Karolinska Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Oliver Kepp
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, UMR 1138, Centre de Recherche des Cordeliers, Paris, France
- Equipe 11 Labellisée par la Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Sorbonne Université, Paris, France
- Université Paris-Saclay, Faculté de Médecine, Kremlin-Bicêtre, France
| |
Collapse
|
102
|
Mitochondrial peptide BRAWNIN is essential for vertebrate respiratory complex III assembly. Nat Commun 2020; 11:1312. [PMID: 32161263 PMCID: PMC7066179 DOI: 10.1038/s41467-020-14999-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 02/14/2020] [Indexed: 11/08/2022] Open
Abstract
The emergence of small open reading frame (sORF)-encoded peptides (SEPs) is rapidly expanding the known proteome at the lower end of the size distribution. Here, we show that the mitochondrial proteome, particularly the respiratory chain, is enriched for small proteins. Using a prediction and validation pipeline for SEPs, we report the discovery of 16 endogenous nuclear encoded, mitochondrial-localized SEPs (mito-SEPs). Through functional prediction, proteomics, metabolomics and metabolic flux modeling, we demonstrate that BRAWNIN, a 71 a.a. peptide encoded by C12orf73, is essential for respiratory chain complex III (CIII) assembly. In human cells, BRAWNIN is induced by the energy-sensing AMPK pathway, and its depletion impairs mitochondrial ATP production. In zebrafish, Brawnin deletion causes complete CIII loss, resulting in severe growth retardation, lactic acidosis and early death. Our findings demonstrate that BRAWNIN is essential for vertebrate oxidative phosphorylation. We propose that mito-SEPs are an untapped resource for essential regulators of oxidative metabolism.
Collapse
|
103
|
Hinge A, He J, Bartram J, Javier J, Xu J, Fjellman E, Sesaki H, Li T, Yu J, Wunderlich M, Mulloy J, Kofron M, Salomonis N, Grimes HL, Filippi MD. Asymmetrically Segregated Mitochondria Provide Cellular Memory of Hematopoietic Stem Cell Replicative History and Drive HSC Attrition. Cell Stem Cell 2020; 26:420-430.e6. [PMID: 32059807 DOI: 10.1016/j.stem.2020.01.016] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/29/2019] [Accepted: 01/23/2020] [Indexed: 01/08/2023]
Abstract
The metabolic requirements of hematopoietic stem cells (HSCs) change with their cell cycle activity. However, the underlying role of mitochondria remains ill-defined. Here we found that, after mitochondrial activation with replication, HSCs irreversibly remodel the mitochondrial network and that this network is not repaired after HSC re-entry into quiescence, contrary to hematopoietic progenitors. HSCs keep and accumulate dysfunctional mitochondria through asymmetric segregation during active division. Mechanistically, mitochondria aggregate and depolarize after stress because of loss of activity of the mitochondrial fission regulator Drp1 onto mitochondria. Genetic and pharmacological studies indicate that inactivation of Drp1 causes loss of HSC regenerative potential while maintaining HSC quiescence. Molecularly, HSCs carrying dysfunctional mitochondria can re-enter quiescence but fail to synchronize the transcriptional control of core cell cycle and metabolic components in subsequent division. Thus, loss of fidelity of mitochondrial morphology and segregation is one type of HSC divisional memory and drives HSC attrition.
Collapse
Affiliation(s)
- Ashwini Hinge
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jingyi He
- Pediatric Research Institute, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R China
| | - James Bartram
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jose Javier
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Juying Xu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ellen Fjellman
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hiromi Sesaki
- Department of Cell Biology, John Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tingyu Li
- Child Nutrition Research Center in the Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, P.R China
| | - Jie Yu
- Department of Hematology and Oncology, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R China
| | - Mark Wunderlich
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - James Mulloy
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Matthew Kofron
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, USA
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, USA
| | - H Leighton Grimes
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, USA
| | - Marie-Dominique Filippi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
104
|
Moruno-Manchon JF, Lejault P, Wang Y, McCauley B, Honarpisheh P, Morales Scheihing DA, Singh S, Dang W, Kim N, Urayama A, Zhu L, Monchaud D, McCullough LD, Tsvetkov AS. Small-molecule G-quadruplex stabilizers reveal a novel pathway of autophagy regulation in neurons. eLife 2020; 9:e52283. [PMID: 32043463 PMCID: PMC7012600 DOI: 10.7554/elife.52283] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 01/07/2020] [Indexed: 12/22/2022] Open
Abstract
Guanine-rich DNA sequences can fold into four-stranded G-quadruplex (G4-DNA) structures. G4-DNA regulates replication and transcription, at least in cancer cells. Here, we demonstrate that, in neurons, pharmacologically stabilizing G4-DNA with G4 ligands strongly downregulates the Atg7 gene. Atg7 is a critical gene for the initiation of autophagy that exhibits decreased transcription with aging. Using an in vitro assay, we show that a putative G-quadruplex-forming sequence (PQFS) in the first intron of the Atg7 gene folds into a G4. An antibody specific to G4-DNA and the G4-DNA-binding protein PC4 bind to the Atg7 PQFS. Mice treated with a G4 stabilizer develop memory deficits. Brain samples from aged mice contain G4-DNA structures that are absent in brain samples from young mice. Overexpressing the G4-DNA helicase Pif1 in neurons exposed to the G4 stabilizer improves phenotypes associated with G4-DNA stabilization. Our findings indicate that G4-DNA is a novel pathway for regulating autophagy in neurons.
Collapse
Affiliation(s)
- Jose F Moruno-Manchon
- Department of Neurobiology and AnatomyThe University of Texas McGovern Medical School at HoustonHoustonUnited States
| | - Pauline Lejault
- Institut de Chimie Moléculaire (ICMUB), UBFC Dijon, CNRS UMR6302DijonFrance
| | - Yaoxuan Wang
- Department of Neurobiology and AnatomyThe University of Texas McGovern Medical School at HoustonHoustonUnited States
| | - Brenna McCauley
- Huffington Center on AgingBaylor College of MedicineHoustonUnited States
| | - Pedram Honarpisheh
- Department of NeurologyThe University of Texas McGovern Medical School at HoustonHoustonUnited States
- The University of Texas Graduate School of Biomedical SciencesHoustonUnited States
| | - Diego A Morales Scheihing
- Department of NeurologyThe University of Texas McGovern Medical School at HoustonHoustonUnited States
| | - Shivani Singh
- Department of Microbiology and Molecular GeneticsThe University of Texas McGovern Medical School at HoustonHoustonUnited States
| | - Weiwei Dang
- Huffington Center on AgingBaylor College of MedicineHoustonUnited States
| | - Nayun Kim
- Department of Microbiology and Molecular GeneticsThe University of Texas McGovern Medical School at HoustonHoustonUnited States
| | - Akihiko Urayama
- Department of NeurologyThe University of Texas McGovern Medical School at HoustonHoustonUnited States
- The University of Texas Graduate School of Biomedical SciencesHoustonUnited States
| | - Liang Zhu
- Biostatistics and Epidemiology Research Design Core Center for Clinical and Translational SciencesThe University of Texas McGovern Medical School at HoustonHoustonUnited States
- Department of Internal MedicineThe University of Texas McGovern Medical School at HoustonHoustonUnited States
| | - David Monchaud
- Institut de Chimie Moléculaire (ICMUB), UBFC Dijon, CNRS UMR6302DijonFrance
| | - Louise D McCullough
- Department of NeurologyThe University of Texas McGovern Medical School at HoustonHoustonUnited States
- The University of Texas Graduate School of Biomedical SciencesHoustonUnited States
| | - Andrey S Tsvetkov
- Department of Neurobiology and AnatomyThe University of Texas McGovern Medical School at HoustonHoustonUnited States
- The University of Texas Graduate School of Biomedical SciencesHoustonUnited States
- UTHealth Consortium on AgingThe University of Texas McGovern Medical School at HoustonHoustonUnited States
| |
Collapse
|
105
|
Gökerküçük EB, Tramier M, Bertolin G. Imaging Mitochondrial Functions: from Fluorescent Dyes to Genetically-Encoded Sensors. Genes (Basel) 2020; 11:E125. [PMID: 31979408 PMCID: PMC7073610 DOI: 10.3390/genes11020125] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/18/2022] Open
Abstract
Mitochondria are multifunctional organelles that are crucial to cell homeostasis. They constitute the major site of energy production for the cell, they are key players in signalling pathways using secondary messengers such as calcium, and they are involved in cell death and redox balance paradigms. Mitochondria quickly adapt their dynamics and biogenesis rates to meet the varying energy demands of the cells, both in normal and in pathological conditions. Therefore, understanding simultaneous changes in mitochondrial functions is crucial in developing mitochondria-based therapy options for complex pathological conditions such as cancer, neurological disorders, and metabolic syndromes. To this end, fluorescence microscopy coupled to live imaging represents a promising strategy to track these changes in real time. In this review, we will first describe the commonly available tools to follow three key mitochondrial functions using fluorescence microscopy: Calcium signalling, mitochondrial dynamics, and mitophagy. Then, we will focus on how the development of genetically-encoded fluorescent sensors became a milestone for the understanding of these mitochondrial functions. In particular, we will show how these tools allowed researchers to address several biochemical activities in living cells, and with high spatiotemporal resolution. With the ultimate goal of tracking multiple mitochondrial functions simultaneously, we will conclude by presenting future perspectives for the development of novel genetically-encoded fluorescent biosensors.
Collapse
Affiliation(s)
| | | | - Giulia Bertolin
- Univ Rennes, CNRS, IGDR [Institut de génétique et développement de Rennes] UMR 6290, F-35000 Rennes, France
| |
Collapse
|
106
|
Intestinal microbes influence development of thymic lymphocytes in early life. Proc Natl Acad Sci U S A 2020; 117:2570-2578. [PMID: 31964813 DOI: 10.1073/pnas.1915047117] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The thymus generates cells of the T cell lineage that seed the lymphatic and blood systems. Transcription factor regulatory networks control the lineage programming and maturation of thymic precursor cells. Whether extrathymic antigenic events, such as the microbial colonization of the mucosal tract also shape the thymic T cell repertoire is unclear. We show here that intestinal microbes influence the thymic homeostasis of PLZF-expressing cells in early life. Impaired thymic development of PLZF+ innate lymphocytes in germ-free (GF) neonatal mice is restored by colonization with a human commensal, Bacteroides fragilis, but not with a polysaccharide A (PSA) deficient isogenic strain. Plasmacytoid dendritic cells influenced by microbes migrate from the colon to the thymus in early life to regulate PLZF+ cell homeostasis. Importantly, perturbations in thymic PLZF+ cells brought about by alterations in early gut microbiota persist into adulthood and are associated with increased susceptibility to experimental colitis. Our studies identify a pathway of communication between intestinal microbes and thymic lymphocytes in the neonatal period that can modulate host susceptibility to immune-mediated diseases later in life.
Collapse
|
107
|
Varuzhanyan G, Rojansky R, Sweredoski MJ, Graham RL, Hess S, Ladinsky MS, Chan DC. Mitochondrial fusion is required for spermatogonial differentiation and meiosis. eLife 2019; 8:51601. [PMID: 31596236 PMCID: PMC6805159 DOI: 10.7554/elife.51601] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 09/27/2019] [Indexed: 01/22/2023] Open
Abstract
Differentiating cells tailor their metabolism to fulfill their specialized functions. We examined whether mitochondrial fusion is important for metabolic tailoring during spermatogenesis. Acutely after depletion of mitofusins Mfn1 and Mfn2, spermatogenesis arrests due to failure to accomplish a metabolic shift during meiosis. This metabolic shift includes increased mitochondrial content, mitochondrial elongation, and upregulation of oxidative phosphorylation (OXPHOS). With long-term mitofusin loss, all differentiating germ cell types are depleted, but proliferation of stem-like undifferentiated spermatogonia remains unaffected. Thus, compared with undifferentiated spermatogonia, differentiating spermatogonia and meiotic spermatocytes have cell physiologies that require high levels of mitochondrial fusion. Proteomics in fibroblasts reveals that mitofusin-null cells downregulate respiratory chain complexes and mitochondrial ribosomal subunits. Similarly, mitofusin depletion in immortalized spermatocytes or germ cells in vivo results in reduced OXPHOS subunits and activity. We reveal that by promoting OXPHOS, mitofusins enable spermatogonial differentiation and a metabolic shift during meiosis.
Collapse
Affiliation(s)
- Grigor Varuzhanyan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Rebecca Rojansky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Michael J Sweredoski
- Proteome Exploration Laboratory of the Beckman Institute, California Institute of Technology, Pasadena, United States
| | - Robert Lj Graham
- Proteome Exploration Laboratory of the Beckman Institute, California Institute of Technology, Pasadena, United States
| | - Sonja Hess
- Proteome Exploration Laboratory of the Beckman Institute, California Institute of Technology, Pasadena, United States
| | - Mark S Ladinsky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| |
Collapse
|
108
|
Mohiuddin M, Lee NH, Moon JY, Han WM, Anderson SE, Choi JJ, Shin E, Nakhai SA, Tran T, Aliya B, Kim DY, Gerold A, Hansen LM, Taylor WR, Jang YC. Critical Limb Ischemia Induces Remodeling of Skeletal Muscle Motor Unit, Myonuclear-, and Mitochondrial-Domains. Sci Rep 2019; 9:9551. [PMID: 31266969 PMCID: PMC6606576 DOI: 10.1038/s41598-019-45923-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 06/20/2019] [Indexed: 11/09/2022] Open
Abstract
Critical limb ischemia, the most severe form of peripheral artery disease, leads to extensive damage and alterations to skeletal muscle homeostasis. Although recent research has investigated the tissue-specific responses to ischemia, the role of the muscle stem cell in the regeneration of its niche components within skeletal muscle has been limited. To elucidate the regenerative mechanism of the muscle stem cell in response to ischemic insults, we explored cellular interactions between the vasculature, neural network, and muscle fiber within the muscle stem cell niche. Using a surgical murine hindlimb ischemia model, we first discovered a significant increase in subsynaptic nuclei and remodeling of the neuromuscular junction following ischemia-induced denervation. In addition, ischemic injury causes significant alterations to the myofiber through a muscle stem cell-mediated accumulation of total myonuclei and a concomitant decrease in myonuclear domain size, possibly to enhance the transcriptional and translation output and restore muscle mass. Results also revealed an accumulation of total mitochondrial content per myonucleus in ischemic myofibers to compensate for impaired mitochondrial function and high turnover rate. Taken together, the findings from this study suggest that the muscle stem cell plays a role in motor neuron reinnervation, myonuclear accretion, and mitochondrial biogenesis for skeletal muscle regeneration following ischemic injury.
Collapse
Affiliation(s)
- Mahir Mohiuddin
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Nan Hee Lee
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - June Young Moon
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Woojin M Han
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Shannon E Anderson
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jeongmoon J Choi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Eunjung Shin
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Shadi A Nakhai
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Thu Tran
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Berna Aliya
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Do Young Kim
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Aimee Gerold
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Laura M Hansen
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - W Robert Taylor
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Young C Jang
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
109
|
Tehrani KF, Latchoumane CV, Southern WM, Pendleton EG, Maslesa A, Karumbaiah L, Call JA, Mortensen LJ. Five-dimensional two-photon volumetric microscopy of in-vivo dynamic activities using liquid lens remote focusing. BIOMEDICAL OPTICS EXPRESS 2019; 10:3591-3604. [PMID: 31360606 PMCID: PMC6640832 DOI: 10.1364/boe.10.003591] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/29/2019] [Accepted: 06/04/2019] [Indexed: 05/08/2023]
Abstract
Multi-photon scanning microscopy provides a robust tool for optical sectioning, which can be used to capture fast biological events such as blood flow, mitochondrial activity, and neuronal action potentials. For many studies, it is important to visualize several different focal planes at a rate akin to the biological event frequency. Typically, a microscope is equipped with mechanical elements to move either the sample or the objective lens to capture volumetric information, but these strategies are limited due to their slow speeds or inertial artifacts. To overcome this problem, remote focusing methods have been developed to shift the focal plane axially without physical movement of the sample or the microscope. Among these methods is liquid lens technology, which adjusts the focus of the lens by changing the wettability of the liquid and hence its curvature. Liquid lenses are inexpensive active optical elements that have the potential for fast multi-photon volumetric imaging, hence a promising and accessible approach for the study of biological systems with complex dynamics. Although remote focusing using liquid lens technology can be used for volumetric point scanning multi-photon microscopy, optical aberrations and the effects of high energy laser pulses have been concerns in its implementation. In this paper, we characterize a liquid lens and validate its use in relevant biological applications. We measured optical aberrations that are caused by the liquid lens, and calculated its response time, defocus hysteresis, and thermal response to a pulsed laser. We applied this method of remote focusing for imaging and measurement of multiple in-vivo specimens, including mesenchymal stem cell dynamics, mouse tibialis anterior muscle mitochondrial electrical potential fluctuations, and mouse brain neural activity. Our system produces 5 dimensional (x,y,z,λ,t) data sets at the speed of 4.2 volumes per second over volumes as large as 160 x 160 x 35 µm3.
Collapse
Affiliation(s)
- Kayvan Forouhesh Tehrani
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - Charles V. Latchoumane
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - W. Michael Southern
- Department of Kinesiology, University of Georgia, Athens, GA 30602, USA
- Currently with: Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Emily G. Pendleton
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - Ana Maslesa
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - Lohitash Karumbaiah
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - Jarrod A. Call
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
- Department of Kinesiology, University of Georgia, Athens, GA 30602, USA
| | - Luke J. Mortensen
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
110
|
Luchsinger LL, Strikoudis A, Danzl NM, Bush EC, Finlayson MO, Satwani P, Sykes M, Yazawa M, Snoeck HW. Harnessing Hematopoietic Stem Cell Low Intracellular Calcium Improves Their Maintenance In Vitro. Cell Stem Cell 2019; 25:225-240.e7. [PMID: 31178255 DOI: 10.1016/j.stem.2019.05.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 03/20/2019] [Accepted: 04/26/2019] [Indexed: 12/17/2022]
Abstract
The specific cellular physiology of hematopoietic stem cells (HSCs) is underexplored, and their maintenance in vitro remains challenging. We discovered that culture of HSCs in low calcium increased their maintenance as determined by phenotype, function, and single-cell expression signature. HSCs are endowed with low intracellular calcium conveyed by elevated activity of glycolysis-fueled plasma membrane calcium efflux pumps and a low-bone-marrow interstitial fluid calcium concentration. Low-calcium conditions inhibited calpain proteases, which target ten-eleven translocated (TET) enzymes, of which TET2 was required for the effect of low calcium conditions on HSC maintenance in vitro. These observations reveal a physiological feature of HSCs that can be harnessed to improve their maintenance in vitro.
Collapse
Affiliation(s)
- Larry L Luchsinger
- Columbia Center of Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; New York Blood Center, Lindsley F. Kimball Research Institute, New York, NY 10065, USA
| | - Alexandros Strikoudis
- Columbia Center of Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Nichole M Danzl
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Erin C Bush
- JP Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Michael O Finlayson
- JP Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Prakash Satwani
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Megan Sykes
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Masayuki Yazawa
- Department of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hans-Willem Snoeck
- Columbia Center of Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
111
|
Kim Y, Yang DS, Katti P, Glancy B. Protein composition of the muscle mitochondrial reticulum during postnatal development. J Physiol 2019; 597:2707-2727. [PMID: 30919448 PMCID: PMC6826232 DOI: 10.1113/jp277579] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/11/2019] [Indexed: 01/24/2023] Open
Abstract
KEY POINTS Muscle mitochondrial networks changed from a longitudinal, fibre parallel orientation to a perpendicular configuration during postnatal development. Mitochondrial dynamics, mitophagy and calcium uptake proteins were abundant during early postnatal development. Mitochondrial biogenesis and oxidative phosphorylation proteins were upregulated throughout muscle development. Postnatal muscle mitochondrial network formation is accompanied by a change in protein expression profile from mitochondria designed for co-ordinated cellular assembly to mitochondria highly specialized for cellular energy metabolism. ABSTRACT Striated muscle mitochondria form connected networks capable of rapid cellular energy distribution. However, the mitochondrial reticulum is not formed at birth and the mechanisms driving network development remain unclear. In the present study, we aimed to establish the network formation timecourse and protein expression profile during postnatal development of the murine muscle mitochondrial reticulum. Two-photon microscopy was used to observe mitochondrial network orientation in tibialis anterior (TA) muscles of live mice at postnatal days (P) 1, 7, 14, 21 and 42, respectively. All muscle fibres maintained a longitudinal, fibre parallel mitochondrial network orientation early in development (P1-7). Mixed networks were most common at P14 but, by P21, almost all fibres had developed the perpendicular mitochondrial orientation observed in mature, glycolytic fibres. Tandem mass tag proteomics were then applied to examine changes in 6869 protein abundances in developing TA muscles. Mitochondrial proteins increased by 32% from P1 to P42. In addition, both nuclear- and mitochondrial-DNA encoded oxidative phosphorylation (OxPhos) components were increased during development, whereas OxPhos assembly factors decreased. Although mitochondrial dynamics and mitophagy were induced at P1-7, mitochondrial biogenesis was enhanced after P14. Moreover, calcium signalling proteins and the mitochondrial calcium uniporter had the highest expression early in postnatal development. In conclusion, mitochondrial networks transform from a fibre parallel to perpendicular orientation during the second and third weeks after birth in murine glycolytic skeletal muscle. This structural transition is accompanied by a change in protein expression profile from mitochondria designed for co-ordinated cellular assembly to mitochondria highly specialized for cellular energy metabolism.
Collapse
Affiliation(s)
- Yuho Kim
- National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaMDUSA
| | - Daniel S. Yang
- National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaMDUSA
| | - Prasanna Katti
- National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaMDUSA
| | - Brian Glancy
- National Heart, Lung, and Blood Institute National Institutes of HealthBethesdaMDUSA
- National Institute of Arthritis and Musculoskeletal and Skin DiseasesNational Institutes of HealthBethesdaMDUSA
| |
Collapse
|
112
|
Zheng Y, Zhang X, Wu X, Jiang L, Ahsan A, Ma S, Xiao Z, Han F, Qin ZH, Hu W, Chen Z. Somatic autophagy of axonal mitochondria in ischemic neurons. J Cell Biol 2019; 218:1891-1907. [PMID: 30979799 PMCID: PMC6548140 DOI: 10.1083/jcb.201804101] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 01/01/2019] [Accepted: 03/15/2019] [Indexed: 12/12/2022] Open
Abstract
How mitochondria damaged in distal axons are cleared is not understood. Zheng et al. find that axonal mitochondria return to neuronal soma for mitophagy after ischemic insult. These spatial features of neuronal mitophagy provide insight into how neurons control mitochondrial quality under pathological conditions. Mitophagy protects against ischemic neuronal injury by eliminating damaged mitochondria, but it is unclear how mitochondria in distal axons are cleared. We find that oxygen and glucose deprivation-reperfusion reduces mitochondrial content in both cell bodies and axons. Axonal mitochondria elimination was not abolished in Atg7fl/fl;nes-Cre neurons, suggesting the absence of direct mitophagy in axons. Instead, axonal mitochondria were enwrapped by autophagosomes in soma and axon-derived mitochondria prioritized for elimination by autophagy. Intriguingly, axonal mitochondria showed prompt loss of anterograde motility but increased retrograde movement upon reperfusion. Anchoring of axonal mitochondria by syntaphilin blocked neuronal mitophagy and aggravated injury. Conversely, induced binding of mitochondria to dynein reinforced retrograde transport and enhanced mitophagy to prevent mitochondrial dysfunction and attenuate neuronal injury. Therefore, we reveal somatic autophagy of axonal mitochondria in ischemic neurons and establish a direct link of retrograde mitochondrial movement with mitophagy. Our findings may provide a new concept for reducing ischemic neuronal injury by correcting mitochondrial motility.
Collapse
Affiliation(s)
- Yanrong Zheng
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Xiangnan Zhang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Xiaoli Wu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Lei Jiang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Anil Ahsan
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Shijia Ma
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Ziyu Xiao
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Feng Han
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Soochow University School of Pharmaceutical Sciences, Suzhou, China
| | - Weiwei Hu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Zhong Chen
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| |
Collapse
|
113
|
Yang C, Hashimoto M, Lin QXX, Tan DQ, Suda T. Sphingosine-1-phosphate signaling modulates terminal erythroid differentiation through the regulation of mitophagy. Exp Hematol 2019; 72:47-59.e1. [DOI: 10.1016/j.exphem.2019.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/10/2019] [Accepted: 01/11/2019] [Indexed: 01/17/2023]
|
114
|
Southern WM, Nichenko AS, Tehrani KF, McGranahan MJ, Krishnan L, Qualls AE, Jenkins NT, Mortensen LJ, Yin H, Yin A, Guldberg RE, Greising SM, Call JA. PGC-1α overexpression partially rescues impaired oxidative and contractile pathophysiology following volumetric muscle loss injury. Sci Rep 2019; 9:4079. [PMID: 30858541 PMCID: PMC6411870 DOI: 10.1038/s41598-019-40606-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/20/2019] [Indexed: 12/26/2022] Open
Abstract
Volumetric muscle loss (VML) injury is characterized by a non-recoverable loss of muscle fibers due to ablative surgery or severe orthopaedic trauma, that results in chronic functional impairments of the soft tissue. Currently, the effects of VML on the oxidative capacity and adaptability of the remaining injured muscle are unclear. A better understanding of this pathophysiology could significantly shape how VML-injured patients and clinicians approach regenerative medicine and rehabilitation following injury. Herein, the data indicated that VML-injured muscle has diminished mitochondrial content and function (i.e., oxidative capacity), loss of mitochondrial network organization, and attenuated oxidative adaptations to exercise. However, forced PGC-1α over-expression rescued the deficits in oxidative capacity and muscle strength. This implicates physiological activation of PGC1-α as a limiting factor in VML-injured muscle's adaptive capacity to exercise and provides a mechanistic target for regenerative rehabilitation approaches to address the skeletal muscle dysfunction.
Collapse
Affiliation(s)
- William M Southern
- Department of Kinesiology, University of Georgia, Athens, GA, 30602, USA.,Regenerative Bioscience Center, University of Georgia, Athens, GA, 30602, USA
| | - Anna S Nichenko
- Department of Kinesiology, University of Georgia, Athens, GA, 30602, USA.,Regenerative Bioscience Center, University of Georgia, Athens, GA, 30602, USA
| | - Kayvan F Tehrani
- Regenerative Bioscience Center, University of Georgia, Athens, GA, 30602, USA
| | | | - Laxminarayanan Krishnan
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Anita E Qualls
- Department of Kinesiology, University of Georgia, Athens, GA, 30602, USA.,Regenerative Bioscience Center, University of Georgia, Athens, GA, 30602, USA
| | - Nathan T Jenkins
- Department of Kinesiology, University of Georgia, Athens, GA, 30602, USA
| | - Luke J Mortensen
- Regenerative Bioscience Center, University of Georgia, Athens, GA, 30602, USA
| | - Hang Yin
- Center for Molecular Medicine, University of Georgia, Athens, GA, 30602, USA.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, 30602, USA
| | - Amelia Yin
- Center for Molecular Medicine, University of Georgia, Athens, GA, 30602, USA.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, 30602, USA
| | - Robert E Guldberg
- Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, 97403, USA
| | - Sarah M Greising
- School of Kinesiology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Jarrod A Call
- Department of Kinesiology, University of Georgia, Athens, GA, 30602, USA. .,Regenerative Bioscience Center, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
115
|
Abstract
Cell-type-specific gene targeting with the Cre/loxP system has become an indispensable technique in experimental neuroscience, particularly for the study of late-born glial cells that make myelin. A plethora of conditional mutants and Cre-expressing mouse lines is now available to the research community that allows laboratories to readily engage in in vivo analyses of oligodendrocytes and their precursor cells. This chapter summarizes concepts and strategies in targeting myelinating glial cells in mice for mutagenesis or imaging, and provides an overview of the most important Cre driver lines successfully used in this rapidly growing field.
Collapse
Affiliation(s)
- Sandra Goebbels
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.
| |
Collapse
|
116
|
Adaniya SM, O-Uchi J, Cypress MW, Kusakari Y, Jhun BS. Posttranslational modifications of mitochondrial fission and fusion proteins in cardiac physiology and pathophysiology. Am J Physiol Cell Physiol 2019; 316:C583-C604. [PMID: 30758993 DOI: 10.1152/ajpcell.00523.2018] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mitochondrial fragmentation frequently occurs in chronic pathological conditions as seen in various human diseases. In fact, abnormal mitochondrial morphology and mitochondrial dysfunction are hallmarks of heart failure (HF) in both human patients and HF animal models. A link between mitochondrial fragmentation and cardiac pathologies has been widely proposed, but the physiological relevance of mitochondrial fission and fusion in the heart is still unclear. Recent studies have increasingly shown that posttranslational modifications (PTMs) of fission and fusion proteins are capable of directly modulating the stability, localization, and/or activity of these proteins. These PTMs include phosphorylation, acetylation, ubiquitination, conjugation of small ubiquitin-like modifier proteins, O-linked-N-acetyl-glucosamine glycosylation, and proteolysis. Thus, understanding the PTMs of fission and fusion proteins may allow us to understand the complexities that determine the balance of mitochondrial fission and fusion as well as mitochondrial function in various cell types and organs including cardiomyocytes and the heart. In this review, we summarize present knowledge regarding the function and regulation of mitochondrial fission and fusion in cardiomyocytes, specifically focusing on the PTMs of each mitochondrial fission/fusion protein. We also discuss the molecular mechanisms underlying abnormal mitochondrial morphology in HF and their contributions to the development of cardiac diseases, highlighting the crucial roles of PTMs of mitochondrial fission and fusion proteins. Finally, we discuss the future potential of manipulating PTMs of fission and fusion proteins as a therapeutic strategy for preventing and/or treating HF.
Collapse
Affiliation(s)
- Stephanie M Adaniya
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota , Minneapolis, Minnesota.,Cardiovascular Research Center, Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University , Providence, Rhode Island
| | - Jin O-Uchi
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota , Minneapolis, Minnesota
| | - Michael W Cypress
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota , Minneapolis, Minnesota
| | - Yoichiro Kusakari
- Department of Cell Physiology, The Jikei University School of Medicine , Tokyo , Japan
| | - Bong Sook Jhun
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota , Minneapolis, Minnesota
| |
Collapse
|
117
|
Pickett SB, Thomas ED, Sebe JY, Linbo T, Esterberg R, Hailey DW, Raible DW. Cumulative mitochondrial activity correlates with ototoxin susceptibility in zebrafish mechanosensory hair cells. eLife 2018; 7:38062. [PMID: 30596476 PMCID: PMC6345563 DOI: 10.7554/elife.38062] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 12/31/2018] [Indexed: 12/11/2022] Open
Abstract
Mitochondria play a prominent role in mechanosensory hair cell damage and death. Although hair cells are thought to be energetically demanding cells, how mitochondria respond to these demands and how this might relate to cell death is largely unexplored. Using genetically encoded indicators, we found that mitochondrial calcium flux and oxidation are regulated by mechanotransduction and demonstrate that hair cell activity has both acute and long-term consequences on mitochondrial function. We tested whether variation in mitochondrial activity reflected differences in the vulnerability of hair cells to the toxic drug neomycin. We observed that susceptibility did not correspond to the acute level of mitochondrial activity but rather to the cumulative history of that activity.
Collapse
Affiliation(s)
- Sarah B Pickett
- Department of Biological Structure, University of Washington, Seattle, United States.,Graduate Program in Neuroscience, University of Washington, Seattle, United States
| | - Eric D Thomas
- Department of Biological Structure, University of Washington, Seattle, United States.,Graduate Program in Neuroscience, University of Washington, Seattle, United States
| | - Joy Y Sebe
- Department of Biological Structure, University of Washington, Seattle, United States
| | - Tor Linbo
- Department of Biological Structure, University of Washington, Seattle, United States
| | - Robert Esterberg
- Department of Biological Structure, University of Washington, Seattle, United States.,Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, United States
| | - Dale W Hailey
- Department of Biological Structure, University of Washington, Seattle, United States.,Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, United States
| | - David W Raible
- Department of Biological Structure, University of Washington, Seattle, United States.,Graduate Program in Neuroscience, University of Washington, Seattle, United States.,Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, United States
| |
Collapse
|
118
|
Jhun BS, O-Uchi J, Adaniya SM, Cypress MW, Yoon Y. Adrenergic Regulation of Drp1-Driven Mitochondrial Fission in Cardiac Physio-Pathology. Antioxidants (Basel) 2018; 7:antiox7120195. [PMID: 30567380 PMCID: PMC6316402 DOI: 10.3390/antiox7120195] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/28/2022] Open
Abstract
Abnormal mitochondrial morphology, especially fragmented mitochondria, and mitochondrial dysfunction are hallmarks of a variety of human diseases including heart failure (HF). Although emerging evidence suggests a link between mitochondrial fragmentation and cardiac dysfunction, it is still not well described which cardiac signaling pathway regulates mitochondrial morphology and function under pathophysiological conditions such as HF. Mitochondria change their shape and location via the activity of mitochondrial fission and fusion proteins. This mechanism is suggested as an important modulator for mitochondrial and cellular functions including bioenergetics, reactive oxygen species (ROS) generation, spatiotemporal dynamics of Ca2+ signaling, cell growth, and death in the mammalian cell- and tissue-specific manners. Recent reports show that a mitochondrial fission protein, dynamin-like/related protein 1 (DLP1/Drp1), is post-translationally modified via cell signaling pathways, which control its subcellular localization, stability, and activity in cardiomyocytes/heart. In this review, we summarize the possible molecular mechanisms for causing post-translational modifications (PTMs) of DLP1/Drp1 in cardiomyocytes, and further discuss how these PTMs of DLP1/Drp1 mediate abnormal mitochondrial morphology and mitochondrial dysfunction under adrenergic signaling activation that contributes to the development and progression of HF.
Collapse
Affiliation(s)
- Bong Sook Jhun
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Jin O-Uchi
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Stephanie M Adaniya
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI 02903, USA.
- Department of Medicine, Division of Cardiology, the Alpert Medical School of Brown University, Providence, RI 02903, USA.
| | - Michael W Cypress
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Yisang Yoon
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
119
|
Bahat A, Goldman A, Zaltsman Y, Khan DH, Halperin C, Amzallag E, Krupalnik V, Mullokandov M, Silberman A, Erez A, Schimmer AD, Hanna JH, Gross A. MTCH2-mediated mitochondrial fusion drives exit from naïve pluripotency in embryonic stem cells. Nat Commun 2018; 9:5132. [PMID: 30510213 PMCID: PMC6277412 DOI: 10.1038/s41467-018-07519-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 11/07/2018] [Indexed: 01/16/2023] Open
Abstract
The role of mitochondria dynamics and its molecular regulators remains largely unknown during naïve-to-primed pluripotent cell interconversion. Here we report that mitochondrial MTCH2 is a regulator of mitochondrial fusion, essential for the naïve-to-primed interconversion of murine embryonic stem cells (ESCs). During this interconversion, wild-type ESCs elongate their mitochondria and slightly alter their glutamine utilization. In contrast, MTCH2-/- ESCs fail to elongate their mitochondria and to alter their metabolism, maintaining high levels of histone acetylation and expression of naïve pluripotency markers. Importantly, enforced mitochondria elongation by the pro-fusion protein Mitofusin (MFN) 2 or by a dominant negative form of the pro-fission protein dynamin-related protein (DRP) 1 is sufficient to drive the exit from naïve pluripotency of both MTCH2-/- and wild-type ESCs. Taken together, our data indicate that mitochondria elongation, governed by MTCH2, plays a critical role and constitutes an early driving force in the naïve-to-primed pluripotency interconversion of murine ESCs.
Collapse
Affiliation(s)
- Amir Bahat
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Andres Goldman
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Yehudit Zaltsman
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Dilshad H Khan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Coral Halperin
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Emmanuel Amzallag
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Vladislav Krupalnik
- Department of Molecular Genetics, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Michael Mullokandov
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Alon Silberman
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jacob H Hanna
- Department of Molecular Genetics, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Atan Gross
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel.
| |
Collapse
|
120
|
Shahen VA, Cantrill LC, Sangani NB, Christodoulou J, Gold WA. A simple and efficient toolset for analysing mitochondrial trafficking in neuronal cells. Acta Histochem 2018; 120:797-805. [PMID: 30224246 DOI: 10.1016/j.acthis.2018.09.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/30/2018] [Accepted: 09/03/2018] [Indexed: 11/25/2022]
Abstract
Mitochondria are crucial for cells, supplying up to 90% of the energy requirements for neurons. Their correct localisation is crucial and ensured by a transport system. Mitochondrial trafficking in neurons is particularly critical, because mitochondria must leave the soma and travel along the axon and dendritic network to facilitate neuronal function. Abnormal mitochondrial trafficking has been reported in several neurological disorders, therefore the ability to quantify and analyse mitochondrial trafficking is vital to improving our understanding of their pathogenesis. Commercial software currently lacks an automated approach for performing such quantitation. Here we demonstrate the development of the Mitochondrial Trafficking and Distribution (MiTrakD) analysis toolset, which consists of simple and free-to-use instructions for mitochondrial trafficking analysis using time-lapse microscopy. MiTrakD utilises existing Fiji (ImageJ) tools for semi-automated, fast and efficient analysis of mitochondrial trafficking and distribution, including velocity, abundance, localisation and distance travelled in neurons. We document MiTrakD's efficiency and accuracy by analysing mitochondrial trafficking using two-dimensional fluorescence images of cortical neurons of wild type mice after 6 days (DIV6), 10 days (DIV10) and 14 days (DIV14) of in vitro incubation. Using MiTrakD we have demonstrated that neurons at all developmental stages exhibited the same percentage of mobile mitochondria, all of which travel in equidistance. Interestingly, the mitochondria in neurons at DIV10 were in greater abundance and were faster than those at DIV6 and DIV14. We can also conclude that MiTrakD is more efficient than manual analysis and is an accurate and reliable tool for performing mitochondrial trafficking analysis in neuronal cells.
Collapse
|
121
|
Guo Y, Jardin BD, Zhou P, Sethi I, Akerberg BN, Toepfer CN, Ai Y, Li Y, Ma Q, Guatimosim S, Hu Y, Varuzhanyan G, VanDusen NJ, Zhang D, Chan DC, Yuan GC, Seidman CE, Seidman JG, Pu WT. Hierarchical and stage-specific regulation of murine cardiomyocyte maturation by serum response factor. Nat Commun 2018; 9:3837. [PMID: 30242271 PMCID: PMC6155060 DOI: 10.1038/s41467-018-06347-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 08/30/2018] [Indexed: 02/06/2023] Open
Abstract
After birth, cardiomyocytes (CM) acquire numerous adaptations in order to efficiently pump blood throughout an animal's lifespan. How this maturation process is regulated and coordinated is poorly understood. Here, we perform a CRISPR/Cas9 screen in mice and identify serum response factor (SRF) as a key regulator of CM maturation. Mosaic SRF depletion in neonatal CMs disrupts many aspects of their maturation, including sarcomere expansion, mitochondrial biogenesis, transverse-tubule formation, and cellular hypertrophy. Maintenance of maturity in adult CMs is less dependent on SRF. This stage-specific activity is associated with developmentally regulated SRF chromatin occupancy and transcriptional regulation. SRF directly activates genes that regulate sarcomere assembly and mitochondrial dynamics. Perturbation of sarcomere assembly but not mitochondrial dynamics recapitulates SRF knockout phenotypes. SRF overexpression also perturbs CM maturation. Together, these data indicate that carefully balanced SRF activity is essential to promote CM maturation through a hierarchy of cellular processes orchestrated by sarcomere assembly.
Collapse
Affiliation(s)
- Yuxuan Guo
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Blake D Jardin
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA, 02215, USA
| | - Pingzhu Zhou
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Isha Sethi
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Brynn N Akerberg
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Christopher N Toepfer
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
- Radcliffe Department of Medicine and Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Yulan Ai
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Yifei Li
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Qing Ma
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Silvia Guatimosim
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG, CEP: 31270-901, Brazil
| | - Yongwu Hu
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Wenzhou Medical University, School of Life Sciences, Wenzhou, China
| | - Grigor Varuzhanyan
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Boulevard, MC 114-96, Pasadena, CA, 91125, USA
| | - Nathan J VanDusen
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Donghui Zhang
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Hubei Collaborative Innovation Center for Green Transformation of Bio-resources, Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, 430062, Wuhan, China
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Boulevard, MC 114-96, Pasadena, CA, 91125, USA
| | - Guo-Cheng Yuan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, MD, 20815, USA
| | - Jonathan G Seidman
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA.
- Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA, 02138, USA.
| |
Collapse
|
122
|
Barrasso AP, Wang S, Tong X, Christiansen AE, Larina IV, Poché RA. Live imaging of developing mouse retinal slices. Neural Dev 2018. [PMID: 30219109 DOI: 10.1186/s13064-018-0120-y.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ex vivo, whole-mount explant culture of the rodent retina has proved to be a valuable approach for studying retinal development. In a limited number of recent studies, this method has been coupled to live fluorescent microscopy with the goal of directly observing dynamic cellular events. However, retinal tissue thickness imposes significant technical limitations. To obtain 3-dimensional images with high quality axial resolution, investigators are restricted to specific areas of the retina and require microscopes, such as 2-photon, with a higher level of depth penetrance. Here, we report a retinal live imaging method that is more amenable to a wider array of imaging systems and does not compromise resolution of retinal cross-sectional area. RESULTS Mouse retinal slice cultures were prepared and standard, inverted confocal microscopy was used to generate movies with high quality resolution of retinal cross-sections. To illustrate the ability of this method to capture discrete, physiologically relevant events during retinal development, we imaged the dynamics of the Fucci cell cycle reporter in both wild type and Cyclin D1 mutant retinal progenitor cells (RPCs) undergoing interkinetic nuclear migration (INM). Like previously reported for the zebrafish, mouse RPCs in G1 phase migrated stochastically and exhibited overall basal drift during development. In contrast, mouse RPCs in G2 phase displayed directed, apical migration toward the ventricular zone prior to mitosis. We also determined that Cyclin D1 knockout RPCs in G2 exhibited a slower apical velocity as compared to wild type. These data are consistent with previous IdU/BrdU window labeling experiments on Cyclin D1 knockout RPCs indicating an elongated cell cycle. Finally, to illustrate the ability to monitor retinal neuron differentiation, we imaged early postnatal horizontal cells (HCs). Time lapse movies uncovered specific HC neurite dynamics consistent with previously published data showing an instructive role for transient vertical neurites in HC mosaic formation. CONCLUSIONS We have detailed a straightforward method to image mouse retinal slice culture preparations that, due to its relative ease, extends live retinal imaging capabilities to a more diverse group of scientists. We have also shown that, by using a slice technique, we can achieve excellent lateral resolution, which is advantageous for capturing intracellular dynamics and overall cell movements during retinal development and differentiation.
Collapse
Affiliation(s)
- Anthony P Barrasso
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.,Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Shang Wang
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xuefei Tong
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Audrey E Christiansen
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Irina V Larina
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.,Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ross A Poché
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA. .,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
123
|
Barrasso AP, Wang S, Tong X, Christiansen AE, Larina IV, Poché RA. Live imaging of developing mouse retinal slices. Neural Dev 2018; 13:23. [PMID: 30219109 PMCID: PMC6139133 DOI: 10.1186/s13064-018-0120-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 09/04/2018] [Indexed: 11/20/2022] Open
Abstract
Background Ex vivo, whole-mount explant culture of the rodent retina has proved to be a valuable approach for studying retinal development. In a limited number of recent studies, this method has been coupled to live fluorescent microscopy with the goal of directly observing dynamic cellular events. However, retinal tissue thickness imposes significant technical limitations. To obtain 3-dimensional images with high quality axial resolution, investigators are restricted to specific areas of the retina and require microscopes, such as 2-photon, with a higher level of depth penetrance. Here, we report a retinal live imaging method that is more amenable to a wider array of imaging systems and does not compromise resolution of retinal cross-sectional area. Results Mouse retinal slice cultures were prepared and standard, inverted confocal microscopy was used to generate movies with high quality resolution of retinal cross-sections. To illustrate the ability of this method to capture discrete, physiologically relevant events during retinal development, we imaged the dynamics of the Fucci cell cycle reporter in both wild type and Cyclin D1 mutant retinal progenitor cells (RPCs) undergoing interkinetic nuclear migration (INM). Like previously reported for the zebrafish, mouse RPCs in G1 phase migrated stochastically and exhibited overall basal drift during development. In contrast, mouse RPCs in G2 phase displayed directed, apical migration toward the ventricular zone prior to mitosis. We also determined that Cyclin D1 knockout RPCs in G2 exhibited a slower apical velocity as compared to wild type. These data are consistent with previous IdU/BrdU window labeling experiments on Cyclin D1 knockout RPCs indicating an elongated cell cycle. Finally, to illustrate the ability to monitor retinal neuron differentiation, we imaged early postnatal horizontal cells (HCs). Time lapse movies uncovered specific HC neurite dynamics consistent with previously published data showing an instructive role for transient vertical neurites in HC mosaic formation. Conclusions We have detailed a straightforward method to image mouse retinal slice culture preparations that, due to its relative ease, extends live retinal imaging capabilities to a more diverse group of scientists. We have also shown that, by using a slice technique, we can achieve excellent lateral resolution, which is advantageous for capturing intracellular dynamics and overall cell movements during retinal development and differentiation. Electronic supplementary material The online version of this article (10.1186/s13064-018-0120-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anthony P Barrasso
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.,Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Shang Wang
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xuefei Tong
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Audrey E Christiansen
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Irina V Larina
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.,Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ross A Poché
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA. .,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
124
|
Kiryu-Seo S, Kiyama H. Mitochondrial behavior during axon regeneration/degeneration in vivo. Neurosci Res 2018; 139:42-47. [PMID: 30179641 DOI: 10.1016/j.neures.2018.08.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/31/2018] [Accepted: 08/22/2018] [Indexed: 01/25/2023]
Abstract
Over the last decade, mitochondrial dynamics beyond function during axon regeneration/degeneration have received attention. Axons have an effective delivery system of mitochondria shuttling between soma and axonal terminals, due to their polarized structure. The proper axonal transport of mitochondria, coordinated with mitochondrial fission/fusion and clearance, is vital for supplying high power energy in injured axons. Many researchers have studied mitochondrial dynamics using in vitro cultured cells with significant progress reported. However, the in vitro culture system is missing a physiological environment including glial cells, immune cells, and endothelial cells, whose communications are indispensable to nerve regeneration/degeneration. In line with this, the understanding of mitochondrial behavior in injured axon in vivo is necessary for promoting the physiological understanding of damaged axons and the development of a therapeutic strategy. In this review, we focus on recent insights into in vivo mitochondrial dynamics during axonal regeneration/degeneration, and introduce the advances of mouse strains to visualize mitochondria in a neuron-specific or an injury-specific manner, which are extremely useful for nerve regeneration/degeneration studies.
Collapse
Affiliation(s)
- Sumiko Kiryu-Seo
- Department of Functional Anatomy and Neuroscience, Graduate School of Medicine, Nagoya University. 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.
| | - Hiroshi Kiyama
- Department of Functional Anatomy and Neuroscience, Graduate School of Medicine, Nagoya University. 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
125
|
Porter C, Ennamorati M, Jain N. In Vivo Photolabeling of Cells in the Colon to Assess Migratory Potential of Hematopoietic Cells in Neonatal Mice. J Vis Exp 2018. [PMID: 30148489 DOI: 10.3791/57929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Enteric bacterial communities are established early in life and influence immune cell development and function. The neonatal microbiota is susceptible to numerous external influences including antibiotics use and diet, which impacts susceptibility to autoimmune and inflammatory diseases. Disorders such as Inflammatory Bowel Disease (IBD) are characterized by a massive influx of immune cells to the intestines. However, immune cells conditioned by the microbiota may additionally emigrate out of the intestines to influence immune responses at extra-intestinal sites. Thus, there is a need to identify and characterize cells that may carry microbial messages from the intestines to distal sites. Here, we describe a method to label cells in the colon of newborn mice in vivo that enables their identification at extra-intestinal sites after migration.
Collapse
Affiliation(s)
- Caryn Porter
- Center for Computational and Integrative Biology, Massachusetts General Hospital;
| | - Maria Ennamorati
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital
| | - Nitya Jain
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital;
| |
Collapse
|
126
|
Lynn H, Howell LM, Diefenbach RJ, Newsome TP. Phototracking Vaccinia Virus Transport Reveals Dynamics of Cytoplasmic Dispersal and a Requirement for A36R and F12L for Exit from the Site of Wrapping. Viruses 2018; 10:v10080390. [PMID: 30042325 PMCID: PMC6115981 DOI: 10.3390/v10080390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 11/16/2022] Open
Abstract
The microtubule cytoskeleton is a primary organizer of viral infections for delivering virus particles to their sites of replication, establishing and maintaining subcellular compartments where distinct steps of viral morphogenesis take place, and ultimately dispersing viral progeny. One of the best characterized examples of virus motility is the anterograde transport of the wrapped virus form of vaccinia virus (VACV) from the trans-Golgi network (TGN) to the cell periphery by kinesin-1. Yet many aspects of this transport event are elusive due to the speed of motility and the challenges of imaging this stage at high resolution over extended time periods. We have established a novel imaging technology to track virus transport that uses photoconvertible fluorescent recombinant viruses to track subsets of virus particles from their site of origin and determine their destination. Here we image virus exit from the TGN and their rate of egress to the cell periphery. We demonstrate a role for kinesin-1 engagement in regulating virus exit from the TGN by removing A36 and F12 function, critical viral mediators of kinesin-1 recruitment to virus particles. Phototracking viral particles and components during infection is a powerful new imaging approach to elucidate mechanisms of virus replication.
Collapse
Affiliation(s)
- Helena Lynn
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Liam M Howell
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Russell J Diefenbach
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia.
| | - Timothy P Newsome
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
127
|
Jarick KJ, Mokhtari Z, Scheller L, Hartweg J, Thusek S, Le DD, Ranecky M, Shaikh H, Qureischi M, Heinze KG, Beilhack A. Photoconversion of Alloreactive T Cells in Murine Peyer's Patches During Acute Graft-Versus-Host Disease: Tracking the Homing Route of Highly Proliferative Cells In Vivo. Front Immunol 2018; 9:1468. [PMID: 30013554 PMCID: PMC6036264 DOI: 10.3389/fimmu.2018.01468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 06/13/2018] [Indexed: 11/13/2022] Open
Abstract
The regulation of immune cell migration throughout the body is essential to warrant immunosurveillance and to maintain immune homeostasis. Marking and tracking of these cells has proven important to study mechanisms of immune cell trafficking and cell interaction in vivo. Photoconversion is a well-suited technique for intravital application because it enables contactless time- and location-specific marking of cells in the tissue without surgically manipulating the microenvironment of the cells in question. However, in dividing cells the converted fluorescent protein may decline quickly. Here, we provide a detailed description of the photoconversion technique and its applicability to tracking highly proliferating T cells from the priming site of T cell activation to peripheral target organs of effector function in a preclinical model. Dendra2+ T cells were photoconverted in the Peyer's patches during the initiation phase of acute graft-versus-host disease (GvHD) and tracked through the mesenteric lymph nodes and the peripheral blood to the small intestine with flow cytometry and intravital two-photon microscopy. Photoconverted alloreactive T cells preserved the full proliferative capacity, homing, and migration of alloreactive T cells in the intestinal lamina propria. We conclusively proved that photoconversion of highly proliferative alloreactive T cells in the Peyer's patches is an effective tool to study trafficking of alloreactive T cells under physiologic conditions and to GvHD target tissues. This technique can also be applied to the study of immune cell tracking under inflammatory and non-inflammatory conditions.
Collapse
Affiliation(s)
- Katja J Jarick
- Interdisciplinary Center for Clinical Research (IZKF) Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital, Würzburg, Germany.,Graduate School of Life Sciences, University of Würzburg, Würzburg, Germany
| | - Zeinab Mokhtari
- Interdisciplinary Center for Clinical Research (IZKF) Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital, Würzburg, Germany
| | - Lukas Scheller
- Interdisciplinary Center for Clinical Research (IZKF) Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital, Würzburg, Germany.,Graduate School of Life Sciences, University of Würzburg, Würzburg, Germany
| | - Julia Hartweg
- Interdisciplinary Center for Clinical Research (IZKF) Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital, Würzburg, Germany.,Graduate School of Life Sciences, University of Würzburg, Würzburg, Germany
| | - Sina Thusek
- Interdisciplinary Center for Clinical Research (IZKF) Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital, Würzburg, Germany
| | - Duc-Dung Le
- Interdisciplinary Center for Clinical Research (IZKF) Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital, Würzburg, Germany
| | - Maria Ranecky
- Interdisciplinary Center for Clinical Research (IZKF) Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital, Würzburg, Germany.,Graduate School of Life Sciences, University of Würzburg, Würzburg, Germany
| | - Haroon Shaikh
- Interdisciplinary Center for Clinical Research (IZKF) Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital, Würzburg, Germany.,Graduate School of Life Sciences, University of Würzburg, Würzburg, Germany
| | - Musga Qureischi
- Interdisciplinary Center for Clinical Research (IZKF) Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital, Würzburg, Germany.,Graduate School of Life Sciences, University of Würzburg, Würzburg, Germany
| | - Katrin G Heinze
- Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Andreas Beilhack
- Interdisciplinary Center for Clinical Research (IZKF) Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital, Würzburg, Germany.,Graduate School of Life Sciences, University of Würzburg, Würzburg, Germany.,Department of Pediatrics, University Hospital, Würzburg, Germany
| |
Collapse
|
128
|
Weil R, Laplantine E, Curic S, Génin P. Role of Optineurin in the Mitochondrial Dysfunction: Potential Implications in Neurodegenerative Diseases and Cancer. Front Immunol 2018; 9:1243. [PMID: 29971063 PMCID: PMC6018216 DOI: 10.3389/fimmu.2018.01243] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 05/17/2018] [Indexed: 12/11/2022] Open
Abstract
Optineurin (Optn) is a 577 aa protein encoded by the Optn gene. Mutations of Optn are associated with normal tension glaucoma and amyotrophic lateral sclerosis, and its gene has also been linked to the development of Paget’s disease of bone and Crohn’s disease. Optn is involved in diverse cellular functions, including NF-κB regulation, membrane trafficking, exocytosis, vesicle transport, reorganization of actin and microtubules, cell cycle control, and autophagy. Besides its role in xenophagy and autophagy of aggregates, Optn has been identified as a primary autophagy receptor, among the five adaptors that translocate to mitochondria during mitophagy. Mitophagy is a selective macroautophagy process during which irreparable mitochondria are degraded, preventing accumulation of defective mitochondria and limiting the release of reactive oxygen species and proapoptotic factors. Mitochondrial quality control via mitophagy is central to the health of cells. One of the important surveillance pathways of mitochondrial health is the recently defined signal transduction pathway involving the mitochondrial PTEN-induced putative kinase 1 (PINK1) protein and the cytosolic RING-between-RING ubiquitin ligase Parkin. Both of these proteins, when mutated, have been identified in certain forms of Parkinson’s disease. By targeting ubiquitinated mitochondria to autophagosomes through its association with autophagy related proteins, Optn is responsible for a critical step in mitophagy. This review reports recent discoveries on the role of Optn in mitophagy and provides insight into its link with neurodegenerative diseases. We will also discuss the involvement of Optn in other pathologies in which mitophagy dysfunctions are involved including cancer.
Collapse
Affiliation(s)
- Robert Weil
- Laboratory of Signaling and Pathogenesis, Institut Pasteur, CNRS UMR3691, Paris, France
| | - Emmanuel Laplantine
- Laboratory of Signaling and Pathogenesis, Institut Pasteur, CNRS UMR3691, Paris, France
| | - Shannel Curic
- Laboratory of Signaling and Pathogenesis, Institut Pasteur, CNRS UMR3691, Paris, France
| | - Pierre Génin
- Laboratory of Signaling and Pathogenesis, Institut Pasteur, CNRS UMR3691, Paris, France
| |
Collapse
|
129
|
Kraus K, Kleene R, Braren I, Loers G, Lutz D, Schachner M. A fragment of adhesion molecule L1 is imported into mitochondria, and regulates mitochondrial metabolism and trafficking. J Cell Sci 2018; 131:jcs.210500. [PMID: 29632241 DOI: 10.1242/jcs.210500] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 03/13/2018] [Indexed: 02/05/2023] Open
Abstract
The cell adhesion molecule L1 (also known as L1CAM) plays important roles in the mammalian nervous system under physiological and pathological conditions. We have previously reported that proteolytic cleavage of L1 by myelin basic protein leads to the generation of a 70 kDa transmembrane L1 fragment (L1-70) that promotes neuronal migration and neuritogenesis. Here, we provide evidence that L1-70 is imported from the cytoplasm into mitochondria. Genetic ablation of L1, inhibition of mitochondrial import of L1-70 or prevention of myelin basic protein-mediated generation of L1-70 all lead to reduced mitochondrial complex I activity, and impaired mitochondrial membrane potential, fusion, fission and motility, as well as increased retrograde transport. We identified NADH dehydrogenase ubiquinone flavoprotein 2 as a binding partner for L1, suggesting that L1-70 interacts with this complex I subunit to regulate complex I activity. The results of our study provide insights into novel functions of L1 in mitochondrial metabolism and cellular dynamics. These functions are likely to ameliorate the consequences of acute nervous system injuries and chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Kristina Kraus
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Ingke Braren
- Vector Core Unit, Institut für Experimentelle Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - David Lutz
- Institut für Strukturelle Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China
| |
Collapse
|
130
|
Moruno-Manchon JF, Uzor NE, Ambati CR, Shetty V, Putluri N, Jagannath C, McCullough LD, Tsvetkov AS. Sphingosine kinase 1-associated autophagy differs between neurons and astrocytes. Cell Death Dis 2018; 9:521. [PMID: 29743513 PMCID: PMC5943283 DOI: 10.1038/s41419-018-0599-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 04/05/2018] [Accepted: 04/12/2018] [Indexed: 02/07/2023]
Abstract
Autophagy is a degradative pathway for removing aggregated proteins, damaged organelles, and parasites. Evidence indicates that autophagic pathways differ between cell types. In neurons, autophagy plays a homeostatic role, compared to a survival mechanism employed by starving non-neuronal cells. We investigated if sphingosine kinase 1 (SK1)-associated autophagy differs between two symbiotic brain cell types-neurons and astrocytes. SK1 synthesizes sphingosine-1-phosphate, which regulates autophagy in non-neuronal cells and in neurons. We found that benzoxazine autophagy inducers upregulate SK1 and neuroprotective autophagy in neurons, but not in astrocytes. Starvation enhances SK1-associated autophagy in astrocytes, but not in neurons. In astrocytes, SK1 is cytoprotective and promotes the degradation of an autophagy substrate, mutant huntingtin, the protein that causes Huntington's disease. Overexpressed SK1 is unexpectedly toxic to neurons, and its toxicity localizes to the neuronal soma, demonstrating an intricate relationship between the localization of SK1's activity and neurotoxicity. Our results underscore the importance of cell type-specific autophagic differences in any efforts to target autophagy therapeutically.
Collapse
Affiliation(s)
- Jose F Moruno-Manchon
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Ndidi-Ese Uzor
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Chandrashekar R Ambati
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Vivekananda Shetty
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Laboratory Medicine, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Louise D McCullough
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
- Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, 77030, USA
| | - Andrey S Tsvetkov
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, 77030, USA.
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
- UT Health Consortium on Aging, The University of Texas McGovern Medical School, Houston, TX, 77030, USA.
| |
Collapse
|
131
|
Harwig MC, Viana MP, Egner JM, Harwig JJ, Widlansky ME, Rafelski SM, Hill RB. Methods for imaging mammalian mitochondrial morphology: A prospective on MitoGraph. Anal Biochem 2018; 552:81-99. [PMID: 29505779 DOI: 10.1016/j.ab.2018.02.022] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/06/2018] [Accepted: 02/22/2018] [Indexed: 12/22/2022]
Abstract
Mitochondria are found in a variety of shapes, from small round punctate structures to a highly interconnected web. This morphological diversity is important for function, but complicates quantification. Consequently, early quantification efforts relied on various qualitative descriptors that understandably reduce the complexity of the network leading to challenges in consistency across the field. Recent application of state-of-the-art computational tools have resulted in more quantitative approaches. This prospective highlights the implementation of MitoGraph, an open-source image analysis platform for measuring mitochondrial morphology initially optimized for use with Saccharomyces cerevisiae. Here Mitograph was assessed on five different mammalian cells types, all of which were accurately segmented by MitoGraph analysis. MitoGraph also successfully differentiated between distinct mitochondrial morphologies that ranged from entirely fragmented to hyper-elongated. General recommendations are also provided for confocal imaging of labeled mitochondria (using mito-YFP, MitoTracker dyes and immunostaining parameters). Widespread adoption of MitoGraph will help achieve a long-sought goal of consistent and reproducible quantification of mitochondrial morphology.
Collapse
Affiliation(s)
- Megan C Harwig
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, United States
| | - Matheus P Viana
- Visual Analytics and Comprehension Group, IBM Research, Brazil; Department of Developmental and Cell Biology and Center for Complex Biological Systems, University of California Irvine, Irvine, CA, 92697, United States
| | - John M Egner
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, United States
| | | | - Michael E Widlansky
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, United States
| | - Susanne M Rafelski
- Department of Developmental and Cell Biology and Center for Complex Biological Systems, University of California Irvine, Irvine, CA, 92697, United States; Assay Development, Allen Institute for Cell Science, Seattle, WA, 98109, United States
| | - R Blake Hill
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, United States.
| |
Collapse
|
132
|
Neutrophils provide cellular communication between ileum and mesenteric lymph nodes at graft-versus-host disease onset. Blood 2018; 131:1858-1869. [PMID: 29463561 DOI: 10.1182/blood-2017-10-812891] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/09/2018] [Indexed: 12/13/2022] Open
Abstract
Conditioning-induced damage of the intestinal tract plays a critical role during the onset of acute graft-versus-host disease (GVHD). Therapeutic interference with these early events of GVHD is difficult, and currently used immunosuppressive drugs mainly target donor T cells. However, not donor T cells but neutrophils reach the sites of tissue injury first, and therefore could be a potential target for GVHD prevention. A detailed analysis of neutrophil fate during acute GVHD and the effect on T cells is difficult because of the short lifespan of this cell type. By using a novel photoconverter reporter system, we show that neutrophils that had been photoconverted in the ileum postconditioning later migrated to mesenteric lymph nodes (mLN). This neutrophil migration was dependent on the intestinal microflora. In the mLN, neutrophils colocalized with T cells and presented antigen on major histocompatibility complex (MHC)-II, thereby affecting T cell expansion. Pharmacological JAK1/JAK2 inhibition reduced neutrophil influx into the mLN and MHC-II expression, thereby interfering with an early event in acute GVHD pathogenesis. In agreement with this finding, neutrophil depletion reduced acute GVHD. We conclude that neutrophils are attracted to the ileum, where the intestinal barrier is disrupted, and then migrate to the mLN, where they participate in alloantigen presentation. JAK1/JAK2-inhibition can interfere with this process, which provides a potential therapeutic strategy to prevent early events of tissue damage-related innate immune cell activation and, ultimately, GVHD.
Collapse
|
133
|
Barrasso AP, Tong X, Poché RA. The mito::mKate2 mouse: A far-red fluorescent reporter mouse line for tracking mitochondrial dynamics in vivo. Genesis 2018; 56:10.1002/dvg.23087. [PMID: 29243279 PMCID: PMC5818295 DOI: 10.1002/dvg.23087] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/30/2017] [Accepted: 12/12/2017] [Indexed: 12/31/2022]
Abstract
Mitochondria are incredibly dynamic organelles that undergo continuous fission and fusion events to control morphology, which profoundly impacts cell physiology including cell cycle progression. This is highlighted by the fact that most major human neurodegenerative diseases are due to specific disruptions in mitochondrial fission or fusion machinery and null alleles of these genes result in embryonic lethality. To gain a better understanding of the pathophysiology of such disorders, tools for the in vivo assessment of mitochondrial dynamics are required. It would be particularly advantageous to simultaneously image mitochondrial fission-fusion coincident with cell cycle progression. To that end, we have generated a new transgenic reporter mouse, called mito::mKate2 that ubiquitously expresses a mitochondria localized far-red mKate2 fluorescent protein. Here we show that mito::mKate2 mice are viable and fertile and that mKate2 fluorescence can be spectrally separated from the previously developed Fucci cell cycle reporters. By crossing mito::mKate2 mice to the ROSA26R-mTmG dual fluorescent Cre reporter line, we also demonstrate the potential utility of mito::mKate2 for genetic mosaic analysis of mitochondrial phenotypes.
Collapse
Affiliation(s)
- Anthony P. Barrasso
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
- Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xuefei Tong
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ross A. Poché
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
134
|
Wilson RJ, Drake JC, Cui D, Zhang M, Perry HM, Kashatus JA, Kusminski CM, Scherer PE, Kashatus DF, Okusa MD, Yan Z. Conditional MitoTimer reporter mice for assessment of mitochondrial structure, oxidative stress, and mitophagy. Mitochondrion 2017; 44:20-26. [PMID: 29274400 PMCID: PMC6387589 DOI: 10.1016/j.mito.2017.12.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/24/2017] [Accepted: 12/15/2017] [Indexed: 11/18/2022]
Abstract
Assessment of structural and functional changes of mitochondria is vital for biomedical research as mitochondria are the power plants essential for biological processes and tissue/organ functions. Others and we have developed a novel reporter gene, pMitoTimer, which codes for a redox sensitive mitochondrial targeted protein that switches from green fluorescence protein (GFP) to red fluorescent protein (DsRed) when oxidized. It has been shown in transfected cells, transgenic C. elegans and Drosophila m., as well as somatically transfected adult skeletal muscle that this reporter gene allows quantifiable assessment of mitochondrial structure, oxidative stress, and lysosomal targeting of mitochondria-containing autophagosomes. Here, we generated CAG-CAT-MitoTimer transgenic mice using a transgene containing MitoTimer downstream of LoxP-flanked bacterial chloramphenicol acetyltransferase (CAT) gene with stop codon under the control of the cytomegalovirus (CMV) enhancer fused to the chicken β-actin promoter (CAG). When CAG-CAT-MitoTimer mice were crossbred with various tissue-specific (muscle, adipose tissue, kidney, and pancreatic tumor) or global Cre transgenic mice, the double transgenic offspring showed MitoTimer expression in tissue-specific or global manner. Lastly, we show that hindlimb ischemia-reperfusion caused early, transient increases of mitochondrial oxidative stress, mitochondrial fragmentation and lysosomal targeting of autophagosomes containing mitochondria as well as a later reduction of mitochondrial content in skeletal muscle along with mitochondrial oxidative stress in sciatic nerve. Thus, we have generated conditional MitoTimer mice and provided proof of principle evidence of their utility to simultaneously assess mitochondrial structure, oxidative stress, and mitophagy in vivo in a tissue-specific, controllable fashion.
Collapse
Affiliation(s)
- Rebecca J Wilson
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, United States; Center for Skeletal Muscle Research at Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Joshua C Drake
- Center for Skeletal Muscle Research at Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Di Cui
- Center for Skeletal Muscle Research at Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Mei Zhang
- Department of Medicine-Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States; Center for Skeletal Muscle Research at Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Heather M Perry
- Department of Medicine-Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Jennifer A Kashatus
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | | | - Philipp E Scherer
- Department of Internal Medicine, UT Southwestern, Dallas, TX, United States
| | - David F Kashatus
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Mark D Okusa
- Department of Medicine-Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Zhen Yan
- Department of Medicine-Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States; Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States; Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, United States; Center for Skeletal Muscle Research at Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States.
| |
Collapse
|
135
|
de Almeida MJ, Luchsinger LL, Corrigan DJ, Williams LJ, Snoeck HW. Dye-Independent Methods Reveal Elevated Mitochondrial Mass in Hematopoietic Stem Cells. Cell Stem Cell 2017; 21:725-729.e4. [PMID: 29198942 DOI: 10.1016/j.stem.2017.11.002] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/25/2017] [Accepted: 11/01/2017] [Indexed: 02/03/2023]
Abstract
Hematopoietic stem cells (HSCs) produce most cellular energy through glycolysis rather than through mitochondrial respiration. Consistent with this notion, mitochondrial mass has been reported to be low in HSCs. However, we found that staining with MitoTracker Green, a commonly used dye to measure mitochondrial content, leads to artefactually low fluorescence specifically in HSCs because of dye efflux. Using mtDNA quantification, enumeration of mitochondrial nucleoids, and fluorescence intensity of a genetically encoded mitochondrial reporter, we unequivocally show here that HSCs and multipotential progenitors (MPPs) have higher mitochondrial mass than lineage-committed progenitors and mature cells. Despite similar mitochondrial mass, respiratory capacity of MPPs exceeds that of HSCs. Furthermore, although elevated mitophagy has been invoked to explain low mitochondrial mass in HSCs, we observed that mitochondrial turnover capacity is comparatively low in HSCs. We propose that the role of mitochondria in HSC biology may have to be revisited in light of these findings.
Collapse
Affiliation(s)
- Mariana Justino de Almeida
- Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Larry L Luchsinger
- Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - David J Corrigan
- Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Linda J Williams
- Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Hans-Willem Snoeck
- Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
136
|
Schultz J, Lee SJ, Cole T, Hoang HD, Vibbert J, Cottee PA, Miller MA, Han SM. The secreted MSP domain of C. elegans VAPB homolog VPR-1 patterns the adult striated muscle mitochondrial reticulum via SMN-1. Development 2017. [PMID: 28634272 PMCID: PMC5482996 DOI: 10.1242/dev.152025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The major sperm protein domain (MSPd) has an extracellular signaling function implicated in amyotrophic lateral sclerosis. Secreted MSPds derived from the C. elegans VAPB homolog VPR-1 promote mitochondrial localization to actin-rich I-bands in body wall muscle. Here we show that the nervous system and germ line are key MSPd secretion tissues. MSPd signals are transduced through the CLR-1 Lar-like tyrosine phosphatase receptor. We show that CLR-1 is expressed throughout the muscle plasma membrane, where it is accessible to MSPd within the pseudocoelomic fluid. MSPd signaling is sufficient to remodel the muscle mitochondrial reticulum during adulthood. An RNAi suppressor screen identified survival of motor neuron 1 (SMN-1) as a downstream effector. SMN-1 acts in muscle, where it colocalizes at myofilaments with ARX-2, a component of the Arp2/3 actin-nucleation complex. Genetic studies suggest that SMN-1 promotes Arp2/3 activity important for localizing mitochondria to I-bands. Our results support the model that VAPB homologs are circulating hormones that pattern the striated muscle mitochondrial reticulum. This function is crucial in adults and requires SMN-1 in muscle, likely independent of its role in pre-mRNA splicing.
Collapse
Affiliation(s)
- Jessica Schultz
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Se-Jin Lee
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Tim Cole
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hieu D Hoang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jack Vibbert
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Pauline A Cottee
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michael A Miller
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sung Min Han
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
137
|
Klein Geltink RI, O'Sullivan D, Corrado M, Bremser A, Buck MD, Buescher JM, Firat E, Zhu X, Niedermann G, Caputa G, Kelly B, Warthorst U, Rensing-Ehl A, Kyle RL, Vandersarren L, Curtis JD, Patterson AE, Lawless S, Grzes K, Qiu J, Sanin DE, Kretz O, Huber TB, Janssens S, Lambrecht BN, Rambold AS, Pearce EJ, Pearce EL. Mitochondrial Priming by CD28. Cell 2017; 171:385-397.e11. [PMID: 28919076 DOI: 10.1016/j.cell.2017.08.018] [Citation(s) in RCA: 223] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 07/31/2017] [Accepted: 08/11/2017] [Indexed: 10/18/2022]
Abstract
T cell receptor (TCR) signaling without CD28 can elicit primary effector T cells, but memory T cells generated during this process are anergic, failing to respond to secondary antigen exposure. We show that, upon T cell activation, CD28 transiently promotes expression of carnitine palmitoyltransferase 1a (Cpt1a), an enzyme that facilitates mitochondrial fatty acid oxidation (FAO), before the first cell division, coinciding with mitochondrial elongation and enhanced spare respiratory capacity (SRC). microRNA-33 (miR33), a target of thioredoxin-interacting protein (TXNIP), attenuates Cpt1a expression in the absence of CD28, resulting in cells that thereafter are metabolically compromised during reactivation or periods of increased bioenergetic demand. Early CD28-dependent mitochondrial engagement is needed for T cells to remodel cristae, develop SRC, and rapidly produce cytokines upon restimulation-cardinal features of protective memory T cells. Our data show that initial CD28 signals during T cell activation prime mitochondria with latent metabolic capacity that is essential for future T cell responses.
Collapse
Affiliation(s)
- Ramon I Klein Geltink
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - David O'Sullivan
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Mauro Corrado
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Anna Bremser
- Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, 79106 Freiburg, Germany; Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Michael D Buck
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Joerg M Buescher
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Elke Firat
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Xuekai Zhu
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, 201210 Shanghai, People's Republic of China
| | - Gabriele Niedermann
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - George Caputa
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Beth Kelly
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Ursula Warthorst
- Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, 79106 Freiburg, Germany
| | - Anne Rensing-Ehl
- Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, 79106 Freiburg, Germany
| | - Ryan L Kyle
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Lana Vandersarren
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium
| | - Jonathan D Curtis
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Annette E Patterson
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Simon Lawless
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Katarzyna Grzes
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Jing Qiu
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - David E Sanin
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Oliver Kretz
- Department of Neuroanatomy, University of Freiburg, 79104 Freiburg, Germany; III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; Department of Medicine IV, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; BIOSS Center for Biological Signaling Studies and Center for Systems Biology (ZBSA), Albert-Ludwigs-University, 79104 Freiburg, Germany
| | - Sophie Janssens
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium
| | - Angelika S Rambold
- Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, 79106 Freiburg, Germany; Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Edward J Pearce
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Erika L Pearce
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany.
| |
Collapse
|
138
|
Abstract
The rise of follicle-stimulating hormone (FSH) is a hallmark of menopause associated with osteoporosis and visceral adiposity. In Nature, Zaidi and colleagues (Liu et al., 2017) report that blocking FSH action reduces body fat by promoting brown/beige fat thermogenesis, potentially providing a new intervention for the treatment of menopause-related metabolic diseases.
Collapse
Affiliation(s)
- Carlos Henrique Sponton
- Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Obesity and Comorbidities Research Center-OCRC, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Shingo Kajimura
- Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
139
|
Hill RA, Damisah EC, Chen F, Kwan AC, Grutzendler J. Targeted two-photon chemical apoptotic ablation of defined cell types in vivo. Nat Commun 2017. [PMID: 28621306 PMCID: PMC5501159 DOI: 10.1038/ncomms15837] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
A major bottleneck limiting understanding of mechanisms and consequences of cell death in complex organisms is the inability to induce and visualize this process with spatial and temporal precision in living animals. Here we report a technique termed two-photon chemical apoptotic targeted ablation (2Phatal) that uses focal illumination with a femtosecond-pulsed laser to bleach a nucleic acid-binding dye causing dose-dependent apoptosis of individual cells without collateral damage. Using 2Phatal, we achieve precise ablation of distinct populations of neurons, glia and pericytes in the mouse brain and in zebrafish. When combined with organelle-targeted fluorescent proteins and biosensors, we uncover previously unrecognized cell-type differences in patterns of apoptosis and associated dynamics of ribosomal disassembly, calcium overload and mitochondrial fission. 2Phatal provides a powerful and rapidly adoptable platform to investigate in vivo functional consequences and neural plasticity following cell death as well as apoptosis, cell clearance and tissue remodelling in diverse organs and species. Investigating cell death in living organisms is hampered by a lack of techniques to induce apoptosis with spatial and temporal precision without collateral damage. Here the authors develop two-photon chemical apoptotic targeted ablation (2Phatal), allowing studies of apoptosis and its functional consequences in vivo.
Collapse
Affiliation(s)
- Robert A Hill
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut 06511, USA.,Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Eyiyemisi C Damisah
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut 06511, USA.,Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut 06511, USA
| | - Fuyi Chen
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut 06511, USA.,Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Alex C Kwan
- Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut 06510, USA.,Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut 06511, USA
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut 06511, USA.,Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut 06510, USA
| |
Collapse
|
140
|
Blocking FSH induces thermogenic adipose tissue and reduces body fat. Nature 2017; 546:107-112. [PMID: 28538730 DOI: 10.1038/nature22342] [Citation(s) in RCA: 245] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 04/05/2017] [Indexed: 12/18/2022]
Abstract
Menopause is associated with bone loss and enhanced visceral adiposity. A polyclonal antibody that targets the β-subunit of the pituitary hormone follicle-stimulating hormone (Fsh) increases bone mass in mice. Here, we report that this antibody sharply reduces adipose tissue in wild-type mice, phenocopying genetic haploinsufficiency for the Fsh receptor gene Fshr. The antibody also causes profound beiging, increases cellular mitochondrial density, activates brown adipose tissue and enhances thermogenesis. These actions result from the specific binding of the antibody to the β-subunit of Fsh to block its action. Our studies uncover opportunities for simultaneously treating obesity and osteoporosis.
Collapse
|
141
|
Glancy B, Hartnell LM, Combs CA, Femnou A, Sun J, Murphy E, Subramaniam S, Balaban RS. Power Grid Protection of the Muscle Mitochondrial Reticulum. Cell Rep 2017; 19:487-496. [PMID: 28423313 DOI: 10.1016/j.celrep.2017.03.063] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/12/2017] [Accepted: 03/21/2017] [Indexed: 11/28/2022] Open
Abstract
Mitochondrial network connectivity enables rapid communication and distribution of potential energy throughout the cell. However, this connectivity puts the energy conversion system at risk, because damaged elements could jeopardize the entire network. Here, we demonstrate the mechanisms for mitochondrial network protection in heart and skeletal muscle (SKM). We find that the cardiac mitochondrial reticulum is segmented into subnetworks comprising many mitochondria linked through abundant contact sites at highly specific intermitochondrial junctions (IMJs). In both cardiac and SKM subnetworks, a rapid electrical and physical separation of malfunctioning mitochondria occurs, consistent with detachment of IMJs and retraction of elongated mitochondria into condensed structures. Regional mitochondrial subnetworks limit the cellular impact of local dysfunction while the dynamic disconnection of damaged mitochondria allows the remaining mitochondria to resume normal function within seconds. Thus, mitochondrial network security is comprised of both proactive and reactive mechanisms in striated muscle cells.
Collapse
Affiliation(s)
- Brian Glancy
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Lisa M Hartnell
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christian A Combs
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Armel Femnou
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Junhui Sun
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elizabeth Murphy
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sriram Subramaniam
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert S Balaban
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
142
|
Liu X, Yang L, Long Q, Weaver D, Hajnóczky G. Choosing proper fluorescent dyes, proteins, and imaging techniques to study mitochondrial dynamics in mammalian cells. BIOPHYSICS REPORTS 2017; 3:64-72. [PMID: 29238743 PMCID: PMC5719805 DOI: 10.1007/s41048-017-0037-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 09/05/2016] [Indexed: 11/30/2022] Open
Abstract
Mitochondrial dynamics refers to the processes maintaining mitochondrial homeostasis, including mitochondrial fission, fusion, transport, biogenesis, and mitophagy. Mitochondrial dynamics is essential for maintaining the metabolic function of mitochondria as well as their regulatory roles in cell signaling. In this review, we summarize the recently developed imaging techniques for studying mitochondrial dynamics including: mitochondrial-targeted fluorescent proteins and dyes, live-cell imaging using photoactivation, photoswitching and cell fusion, mitochondrial transcription and replication imaging by in situ hybridization, and imaging mitochondrial dynamics by super-resolution microscopy. Moreover, we discuss examples of how to choose and combine proper fluorescent dyes and/or proteins.
Collapse
Affiliation(s)
- Xingguo Liu
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Liang Yang
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Qi Long
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - David Weaver
- Department of Pathology, MitoCare Center, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - György Hajnóczky
- Department of Pathology, MitoCare Center, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107 USA
| |
Collapse
|
143
|
Bannerman P, Burns T, Xu J, Miers L, Pleasure D. Mice Hemizygous for a Pathogenic Mitofusin-2 Allele Exhibit Hind Limb/Foot Gait Deficits and Phenotypic Perturbations in Nerve and Muscle. PLoS One 2016; 11:e0167573. [PMID: 27907123 PMCID: PMC5132404 DOI: 10.1371/journal.pone.0167573] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 11/16/2016] [Indexed: 12/31/2022] Open
Abstract
Charcot-Marie-Tooth disease type 2A (CMT2A), the most common axonal form of hereditary sensory motor neuropathy, is caused by mutations of mitofusin-2 (MFN2). Mitofusin-2 is a GTPase required for fusion of mitochondrial outer membranes, repair of damaged mitochondria, efficient mitochondrial energetics, regulation of mitochondrial-endoplasmic reticulum calcium coupling and axonal transport of mitochondria. We knocked T105M MFN2 preceded by a loxP-flanked STOP sequence into the mouse Rosa26 locus to permit cell type-specific expression of this pathogenic allele. Crossing these mice with nestin-Cre transgenic mice elicited T105M MFN2 expression in neuroectoderm, and resulted in diminished numbers of mitochondria in peripheral nerve axons, an alteration in skeletal muscle fiber type distribution, and a gait abnormality.
Collapse
Affiliation(s)
- Peter Bannerman
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, Sacramento, California, United States of America
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, California, United States of America
- * E-mail:
| | - Travis Burns
- Department of Neurology, University of California Davis, Sacramento, California, United States of America
| | - Jie Xu
- Department of Neurology, University of California Davis, Sacramento, California, United States of America
| | - Laird Miers
- Department of Neurology, University of California Davis, Sacramento, California, United States of America
| | - David Pleasure
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, Sacramento, California, United States of America
- Department of Neurology, University of California Davis, Sacramento, California, United States of America
| |
Collapse
|
144
|
Rojansky R, Cha MY, Chan DC. Elimination of paternal mitochondria in mouse embryos occurs through autophagic degradation dependent on PARKIN and MUL1. eLife 2016; 5. [PMID: 27852436 PMCID: PMC5127638 DOI: 10.7554/elife.17896] [Citation(s) in RCA: 240] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 11/14/2016] [Indexed: 12/17/2022] Open
Abstract
A defining feature of mitochondria is their maternal mode of inheritance. However, little is understood about the cellular mechanism through which paternal mitochondria, delivered from sperm, are eliminated from early mammalian embryos. Autophagy has been implicated in nematodes, but whether this mechanism is conserved in mammals has been disputed. Here, we show that cultured mouse fibroblasts and pre-implantation embryos use a common pathway for elimination of mitochondria. Both situations utilize mitophagy, in which mitochondria are sequestered by autophagosomes and delivered to lysosomes for degradation. The E3 ubiquitin ligases PARKIN and MUL1 play redundant roles in elimination of paternal mitochondria. The process is associated with depolarization of paternal mitochondria and additionally requires the mitochondrial outer membrane protein FIS1, the autophagy adaptor P62, and PINK1 kinase. Our results indicate that strict maternal transmission of mitochondria relies on mitophagy and uncover a collaboration between MUL1 and PARKIN in this process. DOI:http://dx.doi.org/10.7554/eLife.17896.001 Mitochondria are commonly referred to as the 'powerhouses' of animal cells because these structures provide the majority of the energy in most cells. People inherit their mitochondria from their mother, and not their father. This is because the father's mitochondria, which are delivered by sperm to the egg, are degraded early on when the embryo starts to develop. Previous studies with model organisms, like nematode worms, showed that mitochondria delivered via sperm (also known as 'paternal mitochondria') were delivered to structures called lysosomes and broken down by the enzymes contained within. However, it remained controversial whether this process, named mitophagy, also occurred in mammalian cells, and the molecules involved were unknown. Now, Rojansky et al. have identified key molecules that are essential for the degradation of mitochondria in mouse cells and show that these same molecules are needed to degrade paternal mitochondria in early mouse embryos. These results indicate that paternal mitochondria are indeed degraded by mitophagy in mice. In addition, Rojansky et al. also note that one of the key molecules is a protein called PARKIN, which is mutated in many inherited cases of Parkinson's disease, a major neurodegenerative disorder. Even though these new findings provide a clearer idea as to how paternal mitochondria are degraded, the question of why remains unanswered. As a result, it is likely that this topic will continue to be heavily debated. Nevertheless, having identified the key molecules involved in degrading paternal mitochondria, it may now be possible to address this question more directly – for example by interfering with this process and then examining the consequences. DOI:http://dx.doi.org/10.7554/eLife.17896.002
Collapse
Affiliation(s)
- Rebecca Rojansky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Moon-Yong Cha
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| |
Collapse
|
145
|
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: 289] [Impact Index Per Article: 32.1] [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
|
146
|
Imaging of neuronal mitochondria in situ. Curr Opin Neurobiol 2016; 39:152-63. [PMID: 27454347 DOI: 10.1016/j.conb.2016.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/04/2016] [Accepted: 06/07/2016] [Indexed: 11/21/2022]
Abstract
Neuronal mitochondria are receiving a rapidly increasing level of attention. This is to a significant part due to the ability to visualize neuronal mitochondria in novel ways, especially in vivo. Such an approach allows studying neuronal mitochondria in an intact tissue context, during different developmental states and in various genetic backgrounds and disease conditions. Hence, in vivo imaging of mitochondria in the nervous system can reveal aspects of the 'mitochondrial life cycle' in neurons that hitherto have remained obscure or could only be inferred indirectly. In this survey of the current literature, we review the new insights that have emerged from studies using mitochondrial imaging in intact neural preparations ranging from worms to mice.
Collapse
|
147
|
Ron-Harel N, Santos D, Ghergurovich JM, Sage PT, Reddy A, Lovitch SB, Dephoure N, Satterstrom FK, Sheffer M, Spinelli JB, Gygi S, Rabinowitz JD, Sharpe AH, Haigis MC. Mitochondrial Biogenesis and Proteome Remodeling Promote One-Carbon Metabolism for T Cell Activation. Cell Metab 2016; 24:104-17. [PMID: 27411012 PMCID: PMC5330619 DOI: 10.1016/j.cmet.2016.06.007] [Citation(s) in RCA: 303] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/12/2016] [Accepted: 06/10/2016] [Indexed: 01/06/2023]
Abstract
Naive T cell stimulation activates anabolic metabolism to fuel the transition from quiescence to growth and proliferation. Here we show that naive CD4(+) T cell activation induces a unique program of mitochondrial biogenesis and remodeling. Using mass spectrometry, we quantified protein dynamics during T cell activation. We identified substantial remodeling of the mitochondrial proteome over the first 24 hr of T cell activation to generate mitochondria with a distinct metabolic signature, with one-carbon metabolism as the most induced pathway. Salvage pathways and mitochondrial one-carbon metabolism, fed by serine, contribute to purine and thymidine synthesis to enable T cell proliferation and survival. Genetic inhibition of the mitochondrial serine catabolic enzyme SHMT2 impaired T cell survival in culture and antigen-specific T cell abundance in vivo. Thus, during T cell activation, mitochondrial proteome remodeling generates specialized mitochondria with enhanced one-carbon metabolism that is critical for T cell activation and survival.
Collapse
Affiliation(s)
- Noga Ron-Harel
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel Santos
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Jonathan M Ghergurovich
- The Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Peter T Sage
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Anita Reddy
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Scott B Lovitch
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Noah Dephoure
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - F Kyle Satterstrom
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Michal Sheffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jessica B Spinelli
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Joshua D Rabinowitz
- The Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Arlene H Sharpe
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Marcia C Haigis
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
148
|
Nemet I, Ropelewski P, Imanishi Y. Applications of phototransformable fluorescent proteins for tracking the dynamics of cellular components. Photochem Photobiol Sci 2016; 14:1787-806. [PMID: 26345171 DOI: 10.1039/c5pp00174a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the past few decades, fluorescent proteins have revolutionized the field of cell biology. Phototransformable fluorescent proteins are capable of changing their excitation and emission spectra after being exposed to specific wavelength(s) of light. The majority of phototransformable fluorescent proteins have originated from marine organisms. Genetic engineering of these proteins has made available many choices for different colors, modes of conversion, and other biophysical properties. Their phototransformative property has allowed the highlighting and tracking of subpopulations of cells, organelles, and proteins in living systems. Furthermore, phototransformable fluorescent proteins have offered new methods for superresolution fluorescence microscopy and optogenetics manipulation of proteins. One of the major advantages of phototransformable fluorescent proteins is their applicability for visualizing newly synthesized proteins that are en route to their final destinations. In this paper, we will discuss the biological applications of phototransformable fluorescent proteins with special emphasis on the application of tracking membrane proteins in vertebrate photoreceptor cells.
Collapse
Affiliation(s)
- Ina Nemet
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA.
| | | | | |
Collapse
|
149
|
Exercise Promotes Healthy Aging of Skeletal Muscle. Cell Metab 2016; 23:1034-1047. [PMID: 27304505 PMCID: PMC5045036 DOI: 10.1016/j.cmet.2016.05.007] [Citation(s) in RCA: 324] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/18/2016] [Accepted: 05/24/2016] [Indexed: 02/08/2023]
Abstract
Primary aging is the progressive and inevitable process of bodily deterioration during adulthood. In skeletal muscle, primary aging causes defective mitochondrial energetics and reduced muscle mass. Secondary aging refers to additional deleterious structural and functional age-related changes caused by diseases and lifestyle factors. Secondary aging can exacerbate deficits in mitochondrial function and muscle mass, concomitant with the development of skeletal muscle insulin resistance. Exercise opposes deleterious effects of secondary aging by preventing the decline in mitochondrial respiration, mitigating aging-related loss of muscle mass and enhancing insulin sensitivity. This review focuses on mechanisms by which exercise promotes "healthy aging" by inducing modifications in skeletal muscle.
Collapse
|
150
|
Miro1 Regulates Activity-Driven Positioning of Mitochondria within Astrocytic Processes Apposed to Synapses to Regulate Intracellular Calcium Signaling. J Neurosci 2016; 35:15996-6011. [PMID: 26631479 DOI: 10.1523/jneurosci.2068-15.2015] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
It is fast emerging that maintaining mitochondrial function is important for regulating astrocyte function, although the specific mechanisms that govern astrocyte mitochondrial trafficking and positioning remain poorly understood. The mitochondrial Rho-GTPase 1 protein (Miro1) regulates mitochondrial trafficking and detachment from the microtubule transport network to control activity-dependent mitochondrial positioning in neurons. However, whether Miro proteins are important for regulating signaling-dependent mitochondrial dynamics in astrocytic processes remains unclear. Using live-cell confocal microscopy of rat organotypic hippocampal slices, we find that enhancing neuronal activity induces transient mitochondrial remodeling in astrocytes, with a concomitant, transient reduction in mitochondrial trafficking, mediated by elevations in intracellular Ca(2+). Stimulating neuronal activity also induced mitochondrial confinement within astrocytic processes in close proximity to synapses. Furthermore, we show that the Ca(2+)-sensing EF-hand domains of Miro1 are important for regulating mitochondrial trafficking in astrocytes and required for activity-driven mitochondrial confinement near synapses. Additionally, activity-dependent mitochondrial positioning by Miro1 reciprocally regulates the levels of intracellular Ca(2+) in astrocytic processes. Thus, the regulation of intracellular Ca(2+) signaling, dependent on Miro1-mediated mitochondrial positioning, could have important consequences for astrocyte Ca(2+) wave propagation, gliotransmission, and ultimately neuronal function.
Collapse
|