201
|
The initiation of puberty in Atlantic salmon brings about large changes in testicular gene expression that are modulated by the energy status. BMC Genomics 2019; 20:475. [PMID: 31185904 PMCID: PMC6558769 DOI: 10.1186/s12864-019-5869-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 05/31/2019] [Indexed: 02/08/2023] Open
Abstract
Background When puberty starts before males reach harvest size, animal welfare and sustainability issues occur in Atlantic salmon (Salmo salar) aquaculture. Hallmarks of male puberty are an increased proliferation activity in the testis and elevated androgen production. Examining transcriptional changes in salmon testis during the transition from immature to maturing testes may help understanding the regulation of puberty, potentially leading to procedures to modulate its start. Since differences in body weight influence, via unknown mechanisms, the chances for entering puberty, we used two feed rations to create body weight differences. Results Maturing testes were characterized by an elevated proliferation activity of Sertoli cells and of single undifferentiated spermatogonia. Pituitary gene expression data suggest increased Gnrh receptor and gonadotropin gene expression, potentially responsible for the elevated circulating androgen levels in maturing fish. Transcriptional changes in maturing testes included a broad variety of signaling systems (e.g. Tgfβ, Wnt, insulin/Igf, nuclear receptors), but also, activation of metabolic pathways such as anaerobic metabolism and protection against ROS. Feed restriction lowered the incidence of puberty. In males maturing despite feed restriction, plasma androgen levels were higher than in maturing fish receiving the full ration. A group of 449 genes that were up-regulated in maturing fully fed fish, was up-regulated more prominently in testis from fish maturing under caloric restriction. Moreover, 421 genes were specifically up-regulated in testes from fish maturing under caloric restriction, including carbon metabolism genes, a pathway relevant for nucleotide biosynthesis and for placing epigenetic marks. Conclusions Undifferentiated spermatogonia and Sertoli cell populations increased at the beginning of puberty, which was associated with the up-regulation of metabolic pathways (e.g. anaerobic and ROS pathways) known from other stem cell systems. The higher androgen levels in males maturing under caloric restriction may be responsible for the stronger up-regulation of a common set of (449) maturation-associated genes, and the specific up-regulation of another set of (421) genes. The latter opened regulatory and/or metabolic options for initiating puberty despite feed restriction. As a means to reduce the incidence of male puberty in salmon, however, caloric restriction seems unsuitable. Electronic supplementary material The online version of this article (10.1186/s12864-019-5869-9) contains supplementary material, which is available to authorized users.
Collapse
|
202
|
Liu X, Wang M, Jiang T, He J, Fu X, Xu Y. IDO1 Maintains Pluripotency of Primed Human Embryonic Stem Cells by Promoting Glycolysis. Stem Cells 2019; 37:1158-1165. [PMID: 31145821 DOI: 10.1002/stem.3044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 04/22/2019] [Accepted: 05/14/2019] [Indexed: 12/26/2022]
Abstract
Human embryonic stem cells (hESCs) depend on glycolysis for energy supply and pluripotency and switch to oxidative phosphorylation upon differentiation. The underlying mechanisms remain unclear. Here, we demonstrate that indoleamine 2,3-dioxygenase 1 (IDO1) is expressed in primed hESCs and its expression rapidly downregulated upon hESC differentiation. IDO1 is required to maintain pluripotency by suppressing mitochondria activity and promoting glycolysis through the increase of NAD+ /NADH ratio. The upregulation of IDO1 during hESC differentiation suppresses the differentiation of hESCs into certain lineages of cells such as cardiomyocytes, which depend on oxidative phosphorylation to satisfy their high energy demand. Therefore, IDO1 plays important roles in maintaining the pluripotency of hESCs. Stem Cells 2019;37:1158-1165.
Collapse
Affiliation(s)
- Xin Liu
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Division of Biological Sciences, University of California, San Diego, La Jolla, California, USA
| | - Meiyan Wang
- Division of Biological Sciences, University of California, San Diego, La Jolla, California, USA
| | - Tao Jiang
- Division of Biological Sciences, University of California, San Diego, La Jolla, California, USA.,The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Jingjin He
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Xuemei Fu
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Yang Xu
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Division of Biological Sciences, University of California, San Diego, La Jolla, California, USA.,The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| |
Collapse
|
203
|
Larijani B, Goodarzi P, Payab M, Alavi-Moghadam S, Rahim F, Bana N, Abedi M, Arabi M, Adibi H, Gilany K, Arjmand B. Metabolomics and Cell Therapy in Diabetes Mellitus. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2019; 8:41-48. [PMID: 32351908 PMCID: PMC7175613 DOI: 10.22088/ijmcm.bums.8.2.41] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/30/2019] [Indexed: 12/22/2022]
Abstract
Diabetes with a broad spectrum of complications has become a global epidemic metabolic disorder. Till now, several pharmaceutical and non-pharmaceutical therapeutic approaches were applied for its treatment. Cell-based therapies have become promising methods for diabetes treatment. Better understanding of diabetes pathogenesis and identification of its specific biomarkers along with evaluation of different treatments efficacy, can be possible by clarification of specific metabolic modifications during the diabetes progression. Subsequently, metabolomics technology can support this goal as an effective tool. The present review tried to show how metabolomics quantifications can be useful for diabetic monitoring before and after cell therapy. Cell therapy is an alternative approach to achieve diabetes treatments goals including insulin resistance amelioration, insulin independence reparation, and control of glycemia. OMICs approaches provide a comprehensive insight into the molecular mechanisms of cells features and functional mechanism of their genomics, transcriptomics, proteomics, and metabolomics profile which can be useful for their therapeutic application. As a modern technology for the detection and analysis of metabolites in biological samples, metabolomica can identify many of the metabolic and molecular pathways associated with diabetes and its following complications.
Collapse
Affiliation(s)
- Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fakher Rahim
- Health Research Institute, Thalassemia and Hemoglobinopathies Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nikoo Bana
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Abedi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Arabi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Adibi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Kambiz Gilany
- Department of Biomedical Sciences, University of Antwerp, Belgium.,Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
204
|
Rastogi A, Joshi P, Contreras E, Gama V. Remodeling of mitochondrial morphology and function: an emerging hallmark of cellular reprogramming. Cell Stress 2019; 3:181-194. [PMID: 31225513 PMCID: PMC6558935 DOI: 10.15698/cst2019.06.189] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Research in the stem cell field has traditionally focused on understanding key transcriptional factors that provide pluripotent cell identity. However, much less is known about other critical non-transcriptional signaling networks that govern stem cell identity. Although we continue to gain critical insights into the mechanisms underlying mitochondrial morphology and function during cellular reprogramming – the process of reverting the fate of a differentiated cell into a stem cell, many uncertainties remain. Recent studies suggest an emerging landscape in which mitochondrial morphology and function have an active role in maintaining and regulating changes in cell identity. In this review, we will focus on these emerging concepts as crucial modulators of cellular reprogramming. Recognition of the widespread applicability of these concepts will increase our understanding of the mitochondrial mechanisms involved in cell identity, cell fate and disease.
Collapse
Affiliation(s)
- Anuj Rastogi
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37240
| | - Piyush Joshi
- Neuroscience Program, Vanderbilt University Medical Center, Nashville, TN 37240.,Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN 37240
| | - Ela Contreras
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37240
| | - Vivian Gama
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37240.,Neuroscience Program, Vanderbilt University Medical Center, Nashville, TN 37240.,Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN 37240.,Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37240
| |
Collapse
|
205
|
Verovskaya EV, Dellorusso PV, Passegué E. Losing Sense of Self and Surroundings: Hematopoietic Stem Cell Aging and Leukemic Transformation. Trends Mol Med 2019; 25:494-515. [PMID: 31109796 DOI: 10.1016/j.molmed.2019.04.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/29/2019] [Accepted: 04/11/2019] [Indexed: 02/07/2023]
Abstract
Aging leads to functional decline of the hematopoietic system, manifested by an increased incidence of hematological disease in the elderly. Deterioration of hematopoietic integrity with age originates in part from the degraded functionality of hematopoietic stem cells (HSCs). Here, we review recent findings identifying changes in metabolic programs and loss of epigenetic identity as major drivers of old HSC dysfunction and their role in promoting leukemia onset in the context of age-related clonal hematopoiesis (ARCH). We discuss how inflammatory and growth signals from the aged bone marrow (BM) microenvironment contribute to cell-intrinsic HSC aging phenotypes and favor leukemia development. Finally, we address how metabolic, epigenetic, and inflammatory pathways could be targeted to enhance old HSC fitness and prevent leukemic transformation.
Collapse
Affiliation(s)
- Evgenia V Verovskaya
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Paul V Dellorusso
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
206
|
Suppressing Mitochondrial Respiration Is Critical for Hypoxia Tolerance in the Fetal Growth Plate. Dev Cell 2019; 49:748-763.e7. [PMID: 31105007 DOI: 10.1016/j.devcel.2019.04.029] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 02/27/2019] [Accepted: 04/17/2019] [Indexed: 12/14/2022]
Abstract
Oxygen (O2) is both an indispensable metabolic substrate and a regulatory signal that controls the activity of Hypoxia-Inducible Factor 1α (Hif1a), a mediator of the cellular adaptation to low O2 tension (hypoxia). Hypoxic cells require Hif1a to survive. Additionally, Hif1a is an inhibitor of mitochondrial respiration. Hence, we hypothesized that enhancing mitochondrial respiration is detrimental to the survival of hypoxic cells in vivo. We tested this hypothesis in the fetal growth plate, which is hypoxic. Our findings show that mitochondrial respiration is dispensable for survival of growth plate chondrocytes. Furthermore, its impairment prevents the extreme hypoxia and the massive chondrocyte death observed in growth plates lacking Hif1a. Consequently, augmenting mitochondrial respiration affects the survival of hypoxic chondrocytes by, at least in part, increasing intracellular hypoxia. We thus propose that partial suppression of mitochondrial respiration is crucial during development to protect the tissues that are physiologically hypoxic from lethal intracellular anoxia.
Collapse
|
207
|
Fujita J, Tohyama S, Kishino Y, Okada M, Morita Y. Concise Review: Genetic and Epigenetic Regulation of Cardiac Differentiation from Human Pluripotent Stem Cells. Stem Cells 2019; 37:992-1002. [PMID: 31021504 DOI: 10.1002/stem.3027] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 04/15/2019] [Indexed: 12/28/2022]
Abstract
Human pluripotent stem cells (hPSCs), including both embryonic stem cells and induced pluripotent stem cells, are the ideal cell sources for disease modeling, drug discovery, and regenerative medicine. In particular, regenerative therapy with hPSC-derived cardiomyocytes (CMs) is an unmet medical need for the treatment of severe heart failure. Cardiac differentiation protocols from hPSCs are made on the basis of cardiac development in vivo. However, current protocols have yet to yield 100% pure CMs, and their maturity is low. Cardiac development is regulated by the cardiac gene network, including transcription factors (TFs). According to our current understanding of cardiac development, cardiac TFs are sequentially expressed during cardiac commitment in hPSCs. Expression levels of each gene are strictly regulated by epigenetic modifications. DNA methylation, histone modification, and noncoding RNAs significantly influence cardiac differentiation. These complex circuits of genetic and epigenetic factors dynamically affect protein expression and metabolic changes in cardiac differentiation and maturation. Here, we review cardiac differentiation protocols and their molecular machinery, closing with a discussion of the future challenges for producing hPSC-derived CMs. Stem Cells 2019;37:992-1002.
Collapse
Affiliation(s)
- Jun Fujita
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Marina Okada
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Yuika Morita
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
208
|
Ni Z, Deng J, Potter CMF, Nowak WN, Gu W, Zhang Z, Chen T, Chen Q, Hu Y, Zhou B, Xu Q, Zhang L. Recipient c-Kit Lineage Cells Repopulate Smooth Muscle Cells of Transplant Arteriosclerosis in Mouse Models. Circ Res 2019; 125:223-241. [PMID: 31079549 PMCID: PMC6615935 DOI: 10.1161/circresaha.119.314855] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Supplemental Digital Content is available in the text. Rationale: Transplantation-accelerated arteriosclerosis is one of the major challenges for long-term survival of patients with solid organ transplantation. Although stem/progenitor cells have been implicated to participate in this process, the cells of origin and underlying mechanisms have not been fully defined. Objective: The objective of our study was to investigate the role of c-Kit lineage cells in allograft-induced neointima formation and to explore the mechanisms underlying this process. Methods and Results: Using an inducible lineage tracing Kit-CreER;Rosa26-tdTomato mouse model, we observed that c-Kit is expressed in multiple cell types in the blood vessels, rather than a specific stem/progenitor cell marker. We performed allograft transplantation between different donor and recipient mice, as well as bone marrow transplantation experiments, demonstrating that recipient c-Kit+ cells repopulate neointimal smooth muscle cells (SMCs) and leukocytes, and contribute to neointima formation in an allograft transplantation model. c-Kit–derived SMCs originate from nonbone marrow tissues, whereas bone marrow-derived c-Kit+ cells mainly generate CD45+ leukocytes. However, the exact identity of c-Kit lineage cells contributing to neointimal SMCs remains unclear. ACK2 (anti-c-Kit antibody), which specifically binds and blocks c-Kit function, ameliorates allograft-induced arteriosclerosis. Stem cell factor and TGF (transforming growth factor)-β1 levels were significantly increased in blood and neointimal lesions after allograft transplantation, by which stem cell factor facilitated c-Kit+ cell migration through the stem cell factor/c-Kit axis and downstream activation of small GTPases, MEK (mitogen-activated protein kinase kinase)/ERK (extracellular signal–regulated kinase)/MLC (myosin light chain), and JNK (c-Jun N-terminal kinase)/c-Jun signaling pathways, whereas TGF-β1 induces c-Kit+ cell differentiation into SMCs via HK (hexokinase)-1–dependent metabolic reprogramming and a possible downstream O-GlcNAcylation of myocardin and serum response factor. Conclusions: Our findings provide evidence that recipient c-Kit lineage cells contribute to vascular remodeling in an allograft transplantation model, in which the stem cell factor/c-Kit axis is responsible for cell migration and HK-1–dependent metabolic reprogramming for SMC differentiation.
Collapse
Affiliation(s)
- Zhichao Ni
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (Z.N., J.D., C.M.F.P., W.N.N., W.G., Z.Z., Y.H., Q.X.)
| | - Jiacheng Deng
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (Z.N., J.D., C.M.F.P., W.N.N., W.G., Z.Z., Y.H., Q.X.)
| | - Claire M F Potter
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (Z.N., J.D., C.M.F.P., W.N.N., W.G., Z.Z., Y.H., Q.X.)
| | - Witold N Nowak
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (Z.N., J.D., C.M.F.P., W.N.N., W.G., Z.Z., Y.H., Q.X.)
| | - Wenduo Gu
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (Z.N., J.D., C.M.F.P., W.N.N., W.G., Z.Z., Y.H., Q.X.)
| | - Zhongyi Zhang
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (Z.N., J.D., C.M.F.P., W.N.N., W.G., Z.Z., Y.H., Q.X.)
| | - Ting Chen
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China (T.C., Q.C., Q.X., L.Z.)
| | - Qishan Chen
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China (T.C., Q.C., Q.X., L.Z.)
| | - Yanhua Hu
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (Z.N., J.D., C.M.F.P., W.N.N., W.G., Z.Z., Y.H., Q.X.)
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, China (B.Z.)
| | - Qingbo Xu
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (Z.N., J.D., C.M.F.P., W.N.N., W.G., Z.Z., Y.H., Q.X.).,Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China (T.C., Q.C., Q.X., L.Z.)
| | - Li Zhang
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China (T.C., Q.C., Q.X., L.Z.)
| |
Collapse
|
209
|
Yuan X, Logan TM, Ma T. Metabolism in Human Mesenchymal Stromal Cells: A Missing Link Between hMSC Biomanufacturing and Therapy? Front Immunol 2019; 10:977. [PMID: 31139179 PMCID: PMC6518338 DOI: 10.3389/fimmu.2019.00977] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/16/2019] [Indexed: 12/21/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) are the most commonly-tested adult stem cells in cell therapy. While the initial focus for hMSC clinical applications was to exploit their multi-potentiality for cell replacement therapies, it is now apparent that hMSCs empower tissue repair primarily by secretion of immuno-regulatory and pro-regenerative factors. A growing trend in hMSC clinical trials is the use of allogenic and culture-expanded cells because they are well-characterized and can be produced in large scale from specific donors to compensate for the donor pathological condition(s). However, donor morbidity and large-scale expansion are known to alter hMSC secretory profile and reduce therapeutic potency, which are significant barriers in hMSC clinical translation. Therefore, understanding the regulatory mechanisms underpinning hMSC phenotypic and functional property is crucial for developing novel engineering protocols that maximize yield while preserving therapeutic potency. hMSC are heterogenous at the level of primary metabolism and that energy metabolism plays important roles in regulating hMSC functional properties. This review focuses on energy metabolism in regulating hMSC immunomodulatory properties and its implication in hMSC sourcing and biomanufacturing. The specific characteristics of hMSC metabolism will be discussed with a focus on hMSC metabolic plasticity and donor- and culture-induced changes in immunomodulatory properties. Potential strategies of modulating hMSC metabolism to enhance their immunomodulation and therapeutic efficacy in preclinical models will be reviewed.
Collapse
Affiliation(s)
- Xuegang Yuan
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL, United States
| | - Timothy M Logan
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, United States.,Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL, United States
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL, United States.,Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
210
|
Mohammad K, Dakik P, Medkour Y, Mitrofanova D, Titorenko VI. Quiescence Entry, Maintenance, and Exit in Adult Stem Cells. Int J Mol Sci 2019; 20:ijms20092158. [PMID: 31052375 PMCID: PMC6539837 DOI: 10.3390/ijms20092158] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/24/2019] [Accepted: 04/28/2019] [Indexed: 12/13/2022] Open
Abstract
Cells of unicellular and multicellular eukaryotes can respond to certain environmental cues by arresting the cell cycle and entering a reversible state of quiescence. Quiescent cells do not divide, but can re-enter the cell cycle and resume proliferation if exposed to some signals from the environment. Quiescent cells in mammals and humans include adult stem cells. These cells exhibit improved stress resistance and enhanced survival ability. In response to certain extrinsic signals, adult stem cells can self-renew by dividing asymmetrically. Such asymmetric divisions not only allow the maintenance of a population of quiescent cells, but also yield daughter progenitor cells. A multistep process of the controlled proliferation of these progenitor cells leads to the formation of one or more types of fully differentiated cells. An age-related decline in the ability of adult stem cells to balance quiescence maintenance and regulated proliferation has been implicated in many aging-associated diseases. In this review, we describe many traits shared by different types of quiescent adult stem cells. We discuss how these traits contribute to the quiescence, self-renewal, and proliferation of adult stem cells. We examine the cell-intrinsic mechanisms that allow establishing and sustaining the characteristic traits of adult stem cells, thereby regulating quiescence entry, maintenance, and exit.
Collapse
Affiliation(s)
- Karamat Mohammad
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Paméla Dakik
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Younes Medkour
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Darya Mitrofanova
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Vladimir I Titorenko
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| |
Collapse
|
211
|
Abstract
Mitochondria are customarily acknowledged as the powerhouse of the cell by virtue of their indispensable role in cellular energy production. In addition, it plays an important role in pluripotency, differentiation, and reprogramming. This review describes variation in the stem cells and their mitochondrial heterogeneity. The mitochondrial variation can be described in terms of structure, function, and subcellular distribution. The mitochondria cristae development status and their localization patterns determine the oxygen consumption rate and ATP production which is a central controller of stem cell maintenance and differentiation. Generally, stem cells show spherical, immature mitochondria with perinuclear distribution. Such mitochondria are metabolically less energetic and low polarized. Moreover, mostly glycolytic energy production is found in pluripotent stem cells with a variation in naïve stem cells which perform oxidative phosphorylation (OXPHOS). This article also describes the structural and functional journey of mitochondria during development. Future insight into underlying mechanisms associated with such alternation in mitochondria of stem cells during embryonic stages could uncover mitochondrial adaptability on cellular demands. Moreover, investigating the importance of mitochondria in pluripotency maintenance might unravel the cause of mitochondrial diseases, aging, and regenerative therapies.
Collapse
|
212
|
Abstract
Over the past decade, advances in systems biology or 'omics techniques have enabled unprecedented insights into the biological processes that occur in cells, tissues, and on the organism level. One of these technologies is the metabolomics, which examines the whole content of the metabolites in a given sample. In a biological system, a stem cell for instance, there are thousands of single components, such as genes, RNA, proteins, and metabolites. These multiple molecular species interact with each other and these interactions may change over the life-time of a cell or in response to specific stimuli, adding to the complexity of the system. Using metabolomics, we can obtain an instantaneous snapshot of the biological status of a cell, tissue, or organism and gain insights on the pattern(s) of numerous analytes, both known and unknown, that result because of a given biological condition. Here, we outline the main methods to study the metabolism of stem cells, including a relatively recent technology of mass spectrometry imaging. Given the abundant and increasing interest in stem cell metabolism in both physiological and pathological conditions, we hope that this chapter will provide incentives for more research in these areas to ultimately reach wide network of applications in biomedical, pharmaceutical, and nutritional research and clinical medicine.
Collapse
|
213
|
Seruggia D, Oti M, Tripathi P, Canver MC, LeBlanc L, Di Giammartino DC, Bullen MJ, Nefzger CM, Sun YBY, Farouni R, Polo JM, Pinello L, Apostolou E, Kim J, Orkin SH, Das PP. TAF5L and TAF6L Maintain Self-Renewal of Embryonic Stem Cells via the MYC Regulatory Network. Mol Cell 2019; 74:1148-1163.e7. [PMID: 31005419 DOI: 10.1016/j.molcel.2019.03.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 01/24/2019] [Accepted: 03/21/2019] [Indexed: 12/16/2022]
Abstract
Self-renewal and pluripotency of the embryonic stem cell (ESC) state are established and maintained by multiple regulatory networks that comprise transcription factors and epigenetic regulators. While much has been learned regarding transcription factors, the function of epigenetic regulators in these networks is less well defined. We conducted a CRISPR-Cas9-mediated loss-of-function genetic screen that identified two epigenetic regulators, TAF5L and TAF6L, components or co-activators of the GNAT-HAT complexes for the mouse ESC (mESC) state. Detailed molecular studies demonstrate that TAF5L/TAF6L transcriptionally activate c-Myc and Oct4 and their corresponding MYC and CORE regulatory networks. Besides, TAF5L/TAF6L predominantly regulate their target genes through H3K9ac deposition and c-MYC recruitment that eventually activate the MYC regulatory network for self-renewal of mESCs. Thus, our findings uncover a role of TAF5L/TAF6L in directing the MYC regulatory network that orchestrates gene expression programs to control self-renewal for the maintenance of mESC state.
Collapse
Affiliation(s)
- Davide Seruggia
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute (DFCI), Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Martin Oti
- Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Wellington Road, Clayton, VIC 3800, Australia
| | - Pratibha Tripathi
- Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Wellington Road, Clayton, VIC 3800, Australia
| | - Matthew C Canver
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute (DFCI), Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Lucy LeBlanc
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, USA
| | - Dafne C Di Giammartino
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Michael J Bullen
- Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Wellington Road, Clayton, VIC 3800, Australia
| | - Christian M Nefzger
- Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Wellington Road, Clayton, VIC 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton, VIC 3800, Australia
| | - Yu Bo Yang Sun
- Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Wellington Road, Clayton, VIC 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton, VIC 3800, Australia
| | - Rick Farouni
- Molecular Pathology & Cancer Center, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114, USA
| | - Jose M Polo
- Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Wellington Road, Clayton, VIC 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton, VIC 3800, Australia
| | - Luca Pinello
- Molecular Pathology & Cancer Center, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114, USA
| | - Effie Apostolou
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jonghwan Kim
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, USA
| | - Stuart H Orkin
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute (DFCI), Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA.
| | - Partha Pratim Das
- Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Wellington Road, Clayton, VIC 3800, Australia.
| |
Collapse
|
214
|
Jeong GJ, Kang D, Kim AK, Han KH, Jeon HR, Kim DI. Metabolites can regulate stem cell behavior through the STAT3/AKT pathway in a similar trend to that under hypoxic conditions. Sci Rep 2019; 9:6112. [PMID: 30992510 PMCID: PMC6468014 DOI: 10.1038/s41598-019-42669-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 11/30/2018] [Indexed: 11/16/2022] Open
Abstract
Stem cell therapy has long been considered a promising mode of treatment for many incurable diseases. Human mesenchymal stem cells (hMSCs) have provided the most promising results to date for regenerative medicine. Nevertheless, due to several obstacles such as difficulty in sourcing and characterizing hMSCs, they remain largely unavailable for clinical use. The signaling requirements for maintaining stem cell function have been studied widely, but little is known about how metabolism contributes to stem cell function. hMSCs have been shown to promote therapeutic efficacy in hypoxic conditions through metabolic conversion. According to published studies, certain metabolites are able to convert stem cell metabolism from oxidative phosphorylation to glycolysis. In this study, we selected several metabolites (fructose-1,6-bisphosphate (FBP), Phosphoenolpyruvic acid (PEP) and sodium oxalate (OXA)) to examine the relation between metabolites and stem cell functions. In addition, we investigated the ability of selected metabolites to induce rapid expansion of this cell population. Our results indicate that selected metabolites stimulate stem cell proliferation by induce glycolytic metabolism via AKT/STAT signaling.
Collapse
Affiliation(s)
- Gun-Jae Jeong
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Donglim Kang
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ae-Kyeong Kim
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyu-Hyun Han
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hye Ran Jeon
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dong-Ik Kim
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
215
|
Otten ABC, Sallevelt SCEH, Carling PJ, Dreesen JCFM, Drüsedau M, Spierts S, Paulussen ADC, de Die-Smulders CEM, Herbert M, Chinnery PF, Samuels DC, Lindsey P, Smeets HJM. Mutation-specific effects in germline transmission of pathogenic mtDNA variants. Hum Reprod 2019; 33:1331-1341. [PMID: 29850888 DOI: 10.1093/humrep/dey114] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 05/15/2018] [Indexed: 12/31/2022] Open
Abstract
STUDY QUESTION Does germline selection (besides random genetic drift) play a role during the transmission of heteroplasmic pathogenic mitochondrial DNA (mtDNA) mutations in humans? SUMMARY ANSWER We conclude that inheritance of mtDNA is mutation-specific and governed by a combination of random genetic drift and negative and/or positive selection. WHAT IS KNOWN ALREADY mtDNA inherits maternally through a genetic bottleneck, but the underlying mechanisms are largely unknown. Although random genetic drift is recognized as an important mechanism, selection mechanisms are thought to play a role as well. STUDY DESIGN, SIZE, DURATION We determined the mtDNA mutation loads in 160 available oocytes, zygotes, and blastomeres of five carriers of the m.3243A>G mutation, one carrier of the m.8993T>G mutation, and one carrier of the m.14487T>C mutation. PARTICIPANTS/MATERIALS, SETTING, METHODS Mutation loads were determined in PGD samples using PCR assays and analysed mathematically to test for random sampling effects. In addition, a meta-analysis has been performed on mutation load transmission data in the literature to confirm the results of the PGD samples. MAIN RESULTS AND THE ROLE OF CHANCE By applying the Kimura distribution, which assumes random mechanisms, we found that mtDNA segregations patterns could be explained by variable bottleneck sizes among all our carriers (moment estimates ranging from 10 to 145). Marked differences in the bottleneck size would determine the probability that a carrier produces offspring with mutations markedly different than her own. We investigated whether bottleneck sizes might also be influenced by non-random mechanisms. We noted a consistent absence of high mutation loads in all our m.3243A>G carriers, indicating non-random events. To test this, we fitted a standard and a truncated Kimura distribution to the m.3243A>G segregation data. A Kimura distribution truncated at 76.5% heteroplasmy has a significantly better fit (P-value = 0.005) than the standard Kimura distribution. For the m.8993T>G mutation, we suspect a skewed mutation load distribution in the offspring. To test this hypothesis, we performed a meta-analysis on published blood mutation levels of offspring-mother (O-M) transmission for the m.3243A>G and m.8993T>G mutations. This analysis revealed some evidence that the O-M ratios for the m.8993T>G mutation are different from zero (P-value <0.001), while for the m.3243A>G mutation there was little evidence that the O-M ratios are non-zero. Lastly, for the m.14487T>G mutation, where the whole range of mutation loads was represented, we found no indications for selective events during its transmission. LARGE SCALE DATA All data are included in the Results section of this article. LIMITATIONS, REASON FOR CAUTION The availability of human material for the mutations is scarce, requiring additional samples to confirm our findings. WIDER IMPLICATIONS OF THE FINDINGS Our data show that non-random mechanisms are involved during mtDNA segregation. We aimed to provide the mechanisms underlying these selection events. One explanation for selection against high m.3243A>G mutation loads could be, as previously reported, a pronounced oxidative phosphorylation (OXPHOS) deficiency at high mutation loads, which prohibits oogenesis (e.g. progression through meiosis). No maximum mutation loads of the m.8993T>G mutation seem to exist, as the OXPHOS deficiency is less severe, even at levels close to 100%. In contrast, high mutation loads seem to be favoured, probably because they lead to an increased mitochondrial membrane potential (MMP), a hallmark on which healthy mitochondria are being selected. This hypothesis could provide a possible explanation for the skewed segregation pattern observed. Our findings are corroborated by the segregation pattern of the m.14487T>C mutation, which does not affect OXPHOS and MMP significantly, and its transmission is therefore predominantly determined by random genetic drift. Our conclusion is that mutation-specific selection mechanisms occur during mtDNA inheritance, which has implications for PGD and mitochondrial replacement therapy. STUDY FUNDING/COMPETING INTEREST(S) This work has been funded by GROW-School of Oncology and Developmental Biology. The authors declare no competing interests.
Collapse
Affiliation(s)
- Auke B C Otten
- Department of Genetics and Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, the Netherlands
| | - Suzanne C E H Sallevelt
- Department of Clinical Genetics, Maastricht University Medical Centre+ (MUMC+), Maastricht, the Netherlands
| | - Phillippa J Carling
- Department of Neuroscience, Sheffield institute for translational neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Joseph C F M Dreesen
- Department of Clinical Genetics, Maastricht University Medical Centre+ (MUMC+), Maastricht, the Netherlands
| | - Marion Drüsedau
- Department of Clinical Genetics, Maastricht University Medical Centre+ (MUMC+), Maastricht, the Netherlands
| | - Sabine Spierts
- Department of Clinical Genetics, Maastricht University Medical Centre+ (MUMC+), Maastricht, the Netherlands
| | - Aimee D C Paulussen
- Department of Clinical Genetics, Maastricht University Medical Centre+ (MUMC+), Maastricht, the Netherlands
| | | | - Mary Herbert
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Patrick F Chinnery
- Department of Clinical Neuroscience, School of Clinical Medicine, University of Cambridge, Cambridge, UK.,Medical Research Council Mitochondrial Biology Unit, Cambridge, Biomedical Campus, Cambridge, UK
| | - David C Samuels
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Patrick Lindsey
- Department of Genetics and Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, the Netherlands
| | - Hubert J M Smeets
- Department of Genetics and Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
216
|
Princz A, Kounakis K, Tavernarakis N. Mitochondrial contributions to neuronal development and function. Biol Chem 2019; 399:723-739. [PMID: 29476663 DOI: 10.1515/hsz-2017-0333] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 02/20/2018] [Indexed: 12/17/2022]
Abstract
Mitochondria are critical to tissues and organs characterized by high-energy demands, such as the nervous system. They provide essential energy and metabolites, and maintain Ca2+ balance, which is imperative for proper neuronal function and development. Emerging findings further underline the role of mitochondria in neurons. Technical advances in the last decades made it possible to investigate key mechanisms in neuronal development and the contribution of mitochondria therein. In this article, we discuss the latest findings relevant to the involvement of mitochondria in neuronal development, placing emphasis on mitochondrial metabolism and dynamics. In addition, we survey the role of mitochondrial energy metabolism and Ca2+ homeostasis in proper neuronal function, and the involvement of mitochondria in axon myelination.
Collapse
Affiliation(s)
- Andrea Princz
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, N. Plastira 100, Vassilika Vouton, Heraklion 70013, Crete, Greece
- Department of Biology, University of Crete, N. Plastira 100, Vassilika Vouton, Heraklion 70013, Crete, Greece
| | - Konstantinos Kounakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, N. Plastira 100, Vassilika Vouton, Heraklion 70013, Crete, Greece
- Department of Basic Sciences, Faculty of Medicine, University of Crete, N. Plastira 100, Vassilika Vouton, Heraklion 70013, Crete, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, N. Plastira 100, Vassilika Vouton, Heraklion 70013, Crete, Greece
- Department of Basic Sciences, Faculty of Medicine, University of Crete, N. Plastira 100, Vassilika Vouton, Heraklion 70013, Crete, Greece
| |
Collapse
|
217
|
Izumi S, Otsuru S, Adachi N, Akabudike N, Enomoto-Iwamoto M. Control of glucose metabolism is important in tenogenic differentiation of progenitors derived from human injured tendons. PLoS One 2019; 14:e0213912. [PMID: 30883580 PMCID: PMC6422258 DOI: 10.1371/journal.pone.0213912] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 03/04/2019] [Indexed: 02/06/2023] Open
Abstract
Glucose metabolism is altered in injured and healing tendons. However, the mechanism by which the glucose metabolism is involved in the pathogenesis of tendon healing process remains unclear. Injured tendons do not completely heal, and often induce fibrous scar and chondroid lesion. Because previous studies have shown that tendon progenitors play roles in tendon repair, we asked whether connective tissue progenitors appearing in injured tendons alter glucose metabolism during tendon healing process. We isolated connective tissue progenitors from the human injured tendons, obtained at the time of primary surgical repair of rupture or laceration. We first characterized the change in glucose metabolism by metabolomics analysis using [1,2-13C]-glucose using the cells isolated from the lacerated flexor tendon. The flux of glucose to the glycolysis pathway was increased in the connective tissue progenitors when they proceeded toward tenogenic and chondrogenic differentiation. The influx of glucose to the tricarboxylic acid (TCA) cycle and biosynthesis of amino acids from the intermediates of the TCA cycle were strongly stimulated toward chondrogenic differentiation. When we treated the cultures with 2-deoxy-D-glucose (2DG), an inhibitor of glycolysis, 2DG inhibited chondrogenesis as characterized by accumulation of mucopolysaccharides and expression of AGGRECAN. Interestingly, 2DG strongly stimulated expression of tenogenic transcription factor genes, SCLERAXIS and MOHAWK under both chondrogenic and tenogenic differentiation conditions. The findings suggest that control of glucose metabolism is beneficial for tenogenic differentiation of connective tissue progenitors.
Collapse
Affiliation(s)
- Soutarou Izumi
- Department of Orthopaedics, University of Maryland, Baltimore, Baltimore, Maryland, United States of America
- Department of Orthopaedic Surgery, Hiroshima University, Hiroshima, Japan
| | - Satoru Otsuru
- Department of Orthopaedics, University of Maryland, Baltimore, Baltimore, Maryland, United States of America
| | - Nobuo Adachi
- Department of Orthopaedic Surgery, Hiroshima University, Hiroshima, Japan
| | - Ngozi Akabudike
- Department of Orthopaedics, University of Maryland, Baltimore, Baltimore, Maryland, United States of America
- * E-mail: (MEI); (NA)
| | - Motomi Enomoto-Iwamoto
- Department of Orthopaedics, University of Maryland, Baltimore, Baltimore, Maryland, United States of America
- * E-mail: (MEI); (NA)
| |
Collapse
|
218
|
Relevance of Oxygen Concentration in Stem Cell Culture for Regenerative Medicine. Int J Mol Sci 2019; 20:ijms20051195. [PMID: 30857245 PMCID: PMC6429522 DOI: 10.3390/ijms20051195] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 02/28/2019] [Accepted: 03/04/2019] [Indexed: 01/10/2023] Open
Abstract
The key hallmark of stem cells is their ability to self-renew while keeping a differentiation potential. Intrinsic and extrinsic cell factors may contribute to a decline in these stem cell properties, and this is of the most importance when culturing them. One of these factors is oxygen concentration, which has been closely linked to the maintenance of stemness. The widely used environmental 21% O2 concentration represents a hyperoxic non-physiological condition, which can impair stem cell behaviour by many mechanisms. The goal of this review is to understand these mechanisms underlying the oxygen signalling pathways and their negatively-associated consequences. This may provide a rationale for culturing stem cells under physiological oxygen concentration for stem cell therapy success, in the field of tissue engineering and regenerative medicine.
Collapse
|
219
|
Abnosi MH, Pari S. Exogenous Nitric Oxide Induced Early Mineralization in
Rat Bone Marrow Mesenchymal Stem Cells via
Activation of Alkaline Phosphatase. IRANIAN BIOMEDICAL JOURNAL 2019; 23. [PMID: 30376703 PMCID: PMC6707106 DOI: 10.29252/.23.2.142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background Since the low concentration and short-time treatment with sodium nitroprusside (SNP), a nitric oxide (NO)–donor, cause no harm to rat bone marrow mesenchymal stem cells (MSCs), we studied the impact of SNP on MSCs differentiation. Methods MSCs were treated with 100 and 1000 µM of SNP for 1 hour in every 48 hours and after 5, 10, 15, and 21 days in osteogenic media. The viability and the level of mineralization were determined using MTT assay and alizarin red staining, respectively. Morphology of the cells was studied using fluorescent dye. Concentration of calcium and the activity of alanine transaminase (ALT), aspartate transaminase (AST), lactate dehydrogenase (LDH), and alkaline phosphatase (ALP) were evaluated by commercial kits. Results SNP with the concentration of 1000 µM significantly reduced viability from day 5 to day 20, but 100 µM did not affect the viability until the day 15. The low concentration of SNP increased matrix deposition from day 10 and reached almost its maximum (4.40 ± 2.4) at the day 15. Also, increasing the activity of ALP (419 ± 2.2), due to low concentration of SNP, started at day 10 and continued till the day 20, while LDH (2026 ± 11) and AST (25.6 ± 0.4) elevations were observed from day 5 onwards. In case of ALT, we observed a significant decrease (36%) from day 5 till day 20. Conclusion Based on our findings, low concentrations of SNP might be useful in the promotion of bone repair.
Collapse
Affiliation(s)
- Mohammad Hussein Abnosi
- Corresponding Author: Mohammad Hussein Abnosi , Department of Biology, Faculty of Sciences, Arak University, Markazi, Iran; Mobile: (+98-918) 9630568; E-mail:
| | | |
Collapse
|
220
|
Mitochondrial Role in Stemness and Differentiation of Hematopoietic Stem Cells. Stem Cells Int 2019; 2019:4067162. [PMID: 30881461 PMCID: PMC6381553 DOI: 10.1155/2019/4067162] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/24/2018] [Indexed: 01/07/2023] Open
Abstract
Quiescent and self-renewing hematopoietic stem cells (HSCs) rely on glycolysis rather than on mitochondrial oxidative phosphorylation (OxPHOS) for energy production. HSC reliance on glycolysis is considered an adaptation to the hypoxic environment of the bone marrow (BM) and reflects the low energetic demands of HSCs. Metabolic rewiring from glycolysis to mitochondrial-based energy generation accompanies HSC differentiation and lineage commitment. Recent evidence, however, highlights that alterations in mitochondrial metabolism and activity are not simply passive consequences but active drivers of HSC fate decisions. Modulation of mitochondrial activity and metabolism is therefore critical for maintaining the self-renewal potential of primitive HSCs and might be beneficial for ex vivo expansion of transplantable HSCs. In this review, we emphasize recent advances in the emerging role of mitochondria in hematopoiesis, cellular reprograming, and HSC fate decisions.
Collapse
|
221
|
Non-thermal plasma-activated medium modified metabolomic profiles in the glycolysis of U251SP glioblastoma. Arch Biochem Biophys 2019; 662:83-92. [DOI: 10.1016/j.abb.2018.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/19/2018] [Accepted: 12/01/2018] [Indexed: 12/19/2022]
|
222
|
Hameury S, Borderie L, Monneuse JM, Skorski G, Pradines D. Prediction of skin anti-aging clinical benefits of an association of ingredients from marine and maritime origins: Ex vivo evaluation using a label-free quantitative proteomic and customized data processing approach. J Cosmet Dermatol 2019; 18:355-370. [PMID: 29797450 DOI: 10.1111/jocd.12528] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2018] [Indexed: 01/18/2023]
Abstract
BACKGROUND The application of ingredients from marine and maritime origins is increasingly common in skin care products, driven by consumer expectations for natural ingredients. However, these ingredients are typically studied for a few isolated in vitro activities. OBJECTIVES The purpose of this study was to carry out a comprehensive evaluation of the activity on the skin of an association of ingredients from marine and maritime origins using label-free quantitative proteomic analysis, in order to predict the clinical benefits if used in a skin care product. METHODS An aqueous gel containing 6.1% of ingredients from marine and maritime origins (amino acid-enriched giant kelp extract, trace element-enriched seawater, dedifferentiated sea fennel cells) was topically applied on human skin explants. The skin explants' proteome was analyzed in a label-free manner by high-performance liquid nano-chromatography coupled with tandem mass spectrometry. A specific data processing pipeline (CORAVALID) providing an objective and comprehensive interpretation of the statistically relevant biological activities processed the results. RESULTS Compared to untreated skin explants, 64 proteins were significantly regulated by the gel treatment (q-value ≤ 0.05). Computer data processing revealed an activity of the ingredients on the epidermis and the dermis. These significantly regulated proteins are involved in gene expression, cell survival and metabolism, inflammatory processes, dermal extracellular matrix synthesis, melanogenesis and keratinocyte proliferation, migration, and differentiation. CONCLUSIONS These results suggest that the tested ingredients could help to preserve a healthy epidermis and dermis, and possibly to prevent the visible signs of skin aging.
Collapse
Affiliation(s)
- Sebastien Hameury
- Research & Development Department, Laboratoires B.L.C. Thalgo Cosmetic S.A., Roquebrune-sur-Argens, France
| | | | | | | | - Dominique Pradines
- Research & Development Department, Laboratoires B.L.C. Thalgo Cosmetic S.A., Roquebrune-sur-Argens, France
| |
Collapse
|
223
|
Kourti M, Westwell A, Jiang W, Cai J. Repurposing old carbon monoxide-releasing molecules towards the anti-angiogenic therapy of triple-negative breast cancer. Oncotarget 2019; 10:1132-1148. [PMID: 30800223 PMCID: PMC6383690 DOI: 10.18632/oncotarget.26638] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 01/16/2019] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is defined by the lack of expression of the oestrogen and progesterone receptors and HER-2. Recently, carbon monoxide (CO) was found to behave as an important endogenous signalling molecule and to suppress VEGF receptor-2 (VEGFR-2) and protein kinase B phosphorylation. Given that anti-angiogenic drugs exist as one of the few available targeted therapies against TNBC, the aim of this project was to study the effects of CO-releasing molecules (CORMs) on TNBC cell lines and the associated endothelial cells and characterise their anti-angiogenic properties that can be used for the reduction of cancer-driven angiogenesis. Four commercially available CORMs were screened for their cytotoxicity, their effects on cell metabolism, migration, VEGF expression, tube formation and VEGFR-2 activation. The most important result was the reduction in VEGF levels expressed by CORM-treated TNBC cells, along with the inhibition of phosphorylation of VEGFR2 and downstream proteins. The migration and tube formation ability of endothelial cells was also decreased by CORMs, justifying a potential re-purposing of old CORMs towards the anti-angiogenic therapy of TNBC. The additional favourable low cytotoxicity, reduction in the glycolysis levels and downregulation of haem oxygenase-1 in TNBC cells enhance the potential of CORMs against TNBC. In this study, CORM-2 remained the most effective CORM and we propose that CORM-2 may be pursued further as an additional agent in combination with existing anti-angiogenic therapies for a more successful targeting of malignant angiogenesis in TNBC.
Collapse
Affiliation(s)
- Malamati Kourti
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK.,School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, CF10 3NB, UK
| | - Andrew Westwell
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, CF10 3NB, UK
| | - Wen Jiang
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Jun Cai
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| |
Collapse
|
224
|
Evano B, Tajbakhsh S. Skeletal muscle stem cells in comfort and stress. NPJ Regen Med 2018; 3:24. [PMID: 30588332 PMCID: PMC6303387 DOI: 10.1038/s41536-018-0062-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/28/2018] [Indexed: 12/21/2022] Open
Abstract
Investigations on developmental and regenerative myogenesis have led to major advances in decrypting stem cell properties and potential, as well as their interactions within the evolving niche. As a consequence, regenerative myogenesis has provided a forum to investigate intrinsic regulators of stem cell properties as well as extrinsic factors, including stromal cells, during normal growth and following injury and disease. Here we review some of the latest advances in the field that have exposed fundamental processes including regulation of stress following trauma and ageing, senescence, DNA damage control and modes of symmetric and asymmetric cell divisions. Recent studies have begun to explore the nature of the niche that is distinct in different muscle groups, and that is altered from prenatal to postnatal stages, and during ageing. We also discuss heterogeneities among muscle stem cells and how distinct properties within the quiescent and proliferating cell states might impact on homoeostasis and regeneration. Interestingly, cellular quiescence, which was thought to be a passive cell state, is regulated by multiple mechanisms, many of which are deregulated in various contexts including ageing. These and other factors including metabolic activity and genetic background can impact on the efficiency of muscle regeneration.
Collapse
Affiliation(s)
- Brendan Evano
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 75015 Paris, France
- CNRS UMR 3738, Institut Pasteur, 75015 Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 75015 Paris, France
- CNRS UMR 3738, Institut Pasteur, 75015 Paris, France
| |
Collapse
|
225
|
Therapeutic targeting of lipid synthesis metabolism for selective elimination of cancer stem cells. Arch Pharm Res 2018; 42:25-39. [PMID: 30536027 DOI: 10.1007/s12272-018-1098-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/01/2018] [Indexed: 02/06/2023]
Abstract
Cancer stem cells (CSCs) are believed to have an essential role in tumor resistance and metastasis; however, no therapeutic strategy for the selective elimination of CSCs has been established. Recently, several studies have shown that the metabolic regulation for ATP synthesis and biological building block generation in CSCs are different from that in bulk cancer cells and rather similar to that in normal tissue stem cells. To take advantage of this difference for CSC elimination therapy, many studies have tested the effect of blocking these metabolism. Two specific processes for lipid biosynthesis, i.e., fatty acid unsaturation and cholesterol biosynthesis, have been shown to be very effective and selective for CSC targets. In this review, lipid metabolism specific to CSCs are summarized. In addition, how monounsaturated fatty acid and cholesterol synthesis may contribute to CSC maintenance are discussed. Specifically, the molecular mechanism required for lipid synthesis and essential for stem cell biology is highlighted. The limit and preview of the lipid metabolism targeting for CSCs are also discussed.
Collapse
|
226
|
Son MJ, Jeong JK, Kwon Y, Ryu JS, Mun SJ, Kim HJ, Kim SW, Yoo S, Kook J, Lee H, Kim J, Chung KS. A novel and safe small molecule enhances hair follicle regeneration by facilitating metabolic reprogramming. Exp Mol Med 2018; 50:1-15. [PMID: 30523246 PMCID: PMC6283868 DOI: 10.1038/s12276-018-0185-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/15/2018] [Accepted: 09/11/2018] [Indexed: 02/08/2023] Open
Abstract
Targeting hair follicle regeneration has been investigated for the treatment of hair loss, and fundamental studies investigating stem cells and their niche have been described. However, knowledge of stem cell metabolism and the specific regulation of bioenergetics during the hair regeneration process is currently insufficient. Here, we report the hair regrowth-promoting effect of a newly synthesized novel small molecule, IM176OUT05 (IM), which activates stem cell metabolism. IM facilitated stemness induction and maintenance during an induced pluripotent stem cell generation process. IM treatment mildly inhibited mitochondrial oxidative phosphorylation and concurrently increased glycolysis, which accelerated stemness induction during the early phase of reprogramming. More importantly, the topical application of IM accelerated hair follicle regeneration by stimulating the progression of the hair follicle cycle to the anagen phase and increased the hair follicle number in mice. Furthermore, the stem cell population with a glycolytic metabotype appeared slightly earlier in the IM-treated mice. Stem cell and niche signaling involved in the hair regeneration process was also activated by the IM treatment during the early phase of hair follicle regeneration. Overall, these results show that the novel small molecule IM promotes tissue regeneration, specifically in hair regrowth, by restructuring the metabolic configuration of stem cells. A compound that establishes metabolic conditions favorable for sustaining stem cells may also offer a safe drug for promoting hair regrowth. Drugs that inhibit mitochondrial activity help lock stem cells into a pluripotent state that allows them to actively divide and repair various tissues, but many of these drugs are toxic. Researchers led by Myung Jin Son of the Korea Research Institute of Bioscience and Biotechnology, Daejeon and ImmunoMet, USA have identified a new compound that safely achieves the same effect. This potential drug helped promote stemness in diverse stem cell types, including the highly proliferative cells that comprise hair follicles. Topical application proved more effective at promoting hair regrowth in female mice than the baldness drug minoxidil, and matched its performance in males, demonstrating its potency as a stem cell modulator.
Collapse
Affiliation(s)
- Myung Jin Son
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea. .,Department of Functional Genomics, Korea University of Science & Technology (UST), 217 Gajungro, Yuseong-gu, Daejeon, 34113, Republic of Korea.
| | - Jae Kap Jeong
- HanAll Biopharma, Bongeunsaro114-gil 12, 9th Floor, Kangnam-gu, Seoul, Republic of Korea.,SCAS-BTT Bioanalysis Co., Ltd, Ochang Scientific Complex 53, Yengudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungcheongbuk-do, 28115, Republic of Korea
| | - Youjeong Kwon
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.,Department of Functional Genomics, Korea University of Science & Technology (UST), 217 Gajungro, Yuseong-gu, Daejeon, 34113, Republic of Korea.,Center for Biomolecular Sciences, University of Illinois at Chicago, 900 South Ashland Ave. 3018, Chicago, IL, 60607, USA
| | - Jae-Sung Ryu
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Seon Ju Mun
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.,Department of Functional Genomics, Korea University of Science & Technology (UST), 217 Gajungro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Hye Jin Kim
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.,Eco-Friendly and New Materials Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Sung-Wuk Kim
- HanAll Biopharma, Bongeunsaro114-gil 12, 9th Floor, Kangnam-gu, Seoul, Republic of Korea.,ImmunoMet Therapeutics Inc., JLABS at Texas Medical Center, 2450 Holcombe Blvd, Houston, TX, 77021, USA
| | - Sanghee Yoo
- ImmunoMet Therapeutics Inc., JLABS at Texas Medical Center, 2450 Holcombe Blvd, Houston, TX, 77021, USA
| | - Jiae Kook
- ImmunoMet Therapeutics Inc., JLABS at Texas Medical Center, 2450 Holcombe Blvd, Houston, TX, 77021, USA
| | - Hongbum Lee
- ImmunoMet Therapeutics Inc., JLABS at Texas Medical Center, 2450 Holcombe Blvd, Houston, TX, 77021, USA
| | - Janghwan Kim
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.,Department of Functional Genomics, Korea University of Science & Technology (UST), 217 Gajungro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Kyung-Sook Chung
- Department of Functional Genomics, Korea University of Science & Technology (UST), 217 Gajungro, Yuseong-gu, Daejeon, 34113, Republic of Korea. .,Biomedical Translational Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
227
|
Tanis SP, Colca JR, Parker TT, Artman GD, Larsen SD, McDonald WG, Gadwood RC, Kletzien RF, Zeller JB, Lee PH, Adams WJ. PPARγ-sparing thiazolidinediones as insulin sensitizers. Design, synthesis and selection of compounds for clinical development. Bioorg Med Chem 2018; 26:5870-5884. [DOI: 10.1016/j.bmc.2018.10.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/20/2018] [Accepted: 10/27/2018] [Indexed: 01/09/2023]
|
228
|
He Y, Wang Y, Zhang B, Li Y, Diao L, Lu L, Yao J, Liu Z, Li D, He F. Revealing the metabolic characteristics of human embryonic stem cells by genome-scale metabolic modeling. FEBS Lett 2018; 592:3670-3682. [PMID: 30223296 DOI: 10.1002/1873-3468.13255] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/29/2018] [Accepted: 09/12/2018] [Indexed: 12/19/2022]
Abstract
Embryonic stem cells (ESCs) are characterized by a dual capacity, self-renewal and pluripotency, which can be regulated by metabolism. A better understanding of ESC metabolism and regulatory mechanisms is pivotal for research into development, ageing, and cancer treatment. However, a systematic and comprehensive delineation of human ESC metabolism is still lacking. Here, we reconstructed the first genome-scale metabolic model (GEM) of human ESCs (hESCs). By GEM simulation and analyses, hESC global metabolic characteristics including essential metabolites and network motifs were identified. Potential metabolic subsystems responsible for self-renewal and pluripotency were also identified by analyses and experiments. This first GEM of hESCs provides a novel view and resource for stem cell metabolism research and will contribute to the elucidation of their metabolic characteristics.
Collapse
Affiliation(s)
- Yangzhige He
- School of Life Sciences, Tsinghua University, Beijing, China.,State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, China.,Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yan Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, China
| | - Boya Zhang
- School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yang Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, China
| | - Lihong Diao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, China
| | - Liang Lu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, China
| | - Jingwen Yao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, China
| | - Zhongyang Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, China
| | - Dong Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, China
| | - Fuchu He
- School of Life Sciences, Tsinghua University, Beijing, China.,State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, China
| |
Collapse
|
229
|
Hazim RA, Volland S, Yen A, Burgess BL, Williams DS. Rapid differentiation of the human RPE cell line, ARPE-19, induced by nicotinamide. Exp Eye Res 2018; 179:18-24. [PMID: 30336127 DOI: 10.1016/j.exer.2018.10.009] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 09/17/2018] [Accepted: 10/13/2018] [Indexed: 12/31/2022]
Abstract
Human RPE cell lines, especially the ARPE-19 cell line, are widely-used in eye research, as well as general epithelial cell studies. In comparison with primary RPE cells, they offer relative convenience and consistency, but cultures derived from these lines are typically not well differentiated. We describe a simple, rapid method to establish cultures from ARPE-19 cells, with significantly improved epithelial cell morphology and cytoskeletal organization, and RPE-related functions. We identify the presence of nicotinamide, a member of the vitamin B family, as an essential factor in promoting the observed differentiation, indicating the importance of metabolism in RPE cell differentiation.
Collapse
Affiliation(s)
- Roni A Hazim
- Department of Ophthalmology and Stein Eye Institute, University of California, Los Angeles, CA, USA
| | - Stefanie Volland
- Department of Ophthalmology and Stein Eye Institute, University of California, Los Angeles, CA, USA
| | - Alice Yen
- Department of Ophthalmology and Stein Eye Institute, University of California, Los Angeles, CA, USA
| | - Barry L Burgess
- Department of Ophthalmology and Stein Eye Institute, University of California, Los Angeles, CA, USA
| | - David S Williams
- Department of Ophthalmology and Stein Eye Institute, University of California, Los Angeles, CA, USA; Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, CA, USA; Brain Research Institute, University of California, Los Angeles, CA, USA.
| |
Collapse
|
230
|
Augustyniak J, Lenart J, Gaj P, Kolanowska M, Jazdzewski K, Stepien PP, Buzanska L. Bezafibrate Upregulates Mitochondrial Biogenesis and Influence Neural Differentiation of Human-Induced Pluripotent Stem Cells. Mol Neurobiol 2018; 56:4346-4363. [PMID: 30315479 PMCID: PMC6505510 DOI: 10.1007/s12035-018-1368-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 09/27/2018] [Indexed: 01/12/2023]
Abstract
Bezafibrate (BZ) regulates mitochondrial biogenesis by activation of PPAR’s receptors and enhancing the level of PGC-1α coactivator. In this report, we investigated the effect of BZ on the expression of genes (1) that are linked to different pathways involved in mitochondrial biogenesis, e.g., regulated by PPAR’s receptors or PGC-1α coactivator, and (2) involved in neuronal or astroglial fate, during neural differentiation of hiPSC. The tested cell populations included hiPSC-derived neural stem cells (NSC), early neural progenitors (eNP), and neural progenitors (NP). RNA-seq analysis showed the expression of PPARA, PPARD receptors and excluded PPARG in all tested populations. The expression of PPARGC1A encoding PGC-1α was dependent on the stage of differentiation: NSC, eNP, and NP differed significantly as compared to hiPSC. In addition, BZ-evoked upregulation of PPARGC1A, GFAP, S100B, and DCX genes coexist with downregulation of MAP2 gene only at the eNP stage of differentiation. In the second task, we investigated the cell sensitivity and mitochondrial biogenesis upon BZ treatment. BZ influenced the cell viability, ROS level, mitochondrial membrane potential, and total cell number in concentration- and stage of differentiation-dependent manner. Induction of mitochondrial biogenesis evoked by BZ determined by the changes in the level of SDHA and COX-1 protein, and mtDNA copy number, as well as the expression of NRF1, PPARGC1A, and TFAM genes, was detected only at NP stage for all tested markers. Thus, developmental stage-specific sensitivity to BZ of neurally differentiating hiPSC can be linked to mitochondrial biogenesis, while fate commitment decisions to PGC-1α (encoded by PPARGC1A) pathway.
Collapse
Affiliation(s)
- Justyna Augustyniak
- Stem Cell Bioengineering Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Jacek Lenart
- Department of Neurochemistry, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Paweł Gaj
- Laboratory of Human Cancer Genetics, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | | | - Krystian Jazdzewski
- Laboratory of Human Cancer Genetics, Centre of New Technologies, University of Warsaw, Warsaw, Poland.,Genomic Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Pawel Stepien
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.,Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.,Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Leonora Buzanska
- Stem Cell Bioengineering Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
231
|
Permissiveness to form pluripotent stem cells may be an evolutionarily derived characteristic in Mus musculus. Sci Rep 2018; 8:14706. [PMID: 30279419 PMCID: PMC6168588 DOI: 10.1038/s41598-018-32116-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 09/03/2018] [Indexed: 01/06/2023] Open
Abstract
Mus musculus is the only known species from which embryonic stem cells (ESC) can be isolated under conditions requiring only leukemia inhibitory factor (LIF). Other species are non-permissive in LIF media, and form developmentally primed epiblast stem cells (EpiSC) similar to cells derived from post-implantation, egg cylinders. To evaluate whether non-permissiveness extends to induced pluripotent stem cells (iPSC), we derived iPSC from the eight founder strains of the mouse Collaborative Cross. Two strains, NOD/ShiLtJ and the WSB/EiJ, were non-permissive, consistent with the previous classification of NOD/ShiLtJ as non-permissive to ESC derivation. We determined non-permissiveness is recessive, and that non-permissive genomes do not compliment. We overcame iPSC non-permissiveness by using GSK3B and MEK inhibitors with serum, a technique we termed 2iS reprogramming. Although used for ESC derivation, GSK3B and MEK inhibitors have not been used during iPSC reprogramming because they inhibit survival of progenitor differentiated cells. iPSC derived in 2iS are more transcriptionally similar to ESC than EpiSC, indicating that 2iS reprogramming acts to overcome genetic background constraints. Finally, of species tested for ESC or iPSC derivation, only some M. musculus strains are permissive under LIF culture conditions suggesting that this is an evolutionarily derived characteristic in the M. musculus lineage.
Collapse
|
232
|
Cell-specific proteome analyses of human bone marrow reveal molecular features of age-dependent functional decline. Nat Commun 2018; 9:4004. [PMID: 30275468 PMCID: PMC6167374 DOI: 10.1038/s41467-018-06353-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 08/30/2018] [Indexed: 01/25/2023] Open
Abstract
Diminishing potential to replace damaged tissues is a hallmark for ageing of somatic stem cells, but the mechanisms remain elusive. Here, we present proteome-wide atlases of age-associated alterations in human haematopoietic stem and progenitor cells (HPCs) and five other cell populations that constitute the bone marrow niche. For each, the abundance of a large fraction of the ~12,000 proteins identified is assessed in 59 human subjects from different ages. As the HPCs become older, pathways in central carbon metabolism exhibit features reminiscent of the Warburg effect, where glycolytic intermediates are rerouted towards anabolism. Simultaneously, altered abundance of early regulators of HPC differentiation reveals a reduced functionality and a bias towards myeloid differentiation. Ageing causes alterations in the bone marrow niche too, and diminishes the functionality of the pathways involved in HPC homing. The data represent a valuable resource for further analyses, and for validation of knowledge gained from animal models. Ageing causes an inability to replace damaged tissue. Here, the authors perform proteomics analyses of human haematopoietic stem cells and other cells in the bone marrow niche at different ages and show changes in central carbon metabolism, reduced bone marrow niche function, and enhanced myeloid differentiation.
Collapse
|
233
|
Miyazawa H, Aulehla A. Revisiting the role of metabolism during development. Development 2018; 145:145/19/dev131110. [DOI: 10.1242/dev.131110] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
ABSTRACT
An emerging view emphasizes that metabolism is highly regulated in both time and space. In addition, it is increasingly being recognized that metabolic pathways are tightly connected to specific biological processes such as cell signaling, proliferation and differentiation. As we obtain a better view of this spatiotemporal regulation of metabolism, and of the molecular mechanisms that connect metabolism and signaling, we can now move from largely correlative to more functional studies. It is, therefore, a particularly promising time to revisit how metabolism can affect multiple aspects of animal development. In this Review, we discuss how metabolism is mechanistically linked to cellular and developmental programs through both its bioenergetic and metabolic signaling functions. We highlight how metabolism is regulated across various spatial and temporal scales, and discuss how this regulation can influence cellular processes such as cell signaling, gene expression, and epigenetic and post-translational modifications during embryonic development.
Collapse
Affiliation(s)
- Hidenobu Miyazawa
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, 69117, Germany
| | - Alexander Aulehla
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, 69117, Germany
| |
Collapse
|
234
|
Ito K, Bonora M, Ito K. Metabolism as master of hematopoietic stem cell fate. Int J Hematol 2018; 109:18-27. [PMID: 30219988 DOI: 10.1007/s12185-018-2534-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 09/10/2018] [Indexed: 12/13/2022]
Abstract
HSCs have a fate choice when they divide; they can self-renew, producing new HSCs, or produce daughter cells that will mature to become committed cells. Technical challenges, however, have long obscured the mechanics of these choices. Advances in flow-sorting have made possible the purification of HSC populations, but available HSC-enriched fractions still include substantial heterogeneity, and single HSCs have proven extremely difficult to track and observe. Advances in single-cell approaches, however, have led to the identification of a highly purified population of hematopoietic stem cells (HSCs) that make a critical contribution to hematopoietic homeostasis through a preference for self-renewing division. Metabolic cues are key regulators of this cell fate choice, and the importance of controlling the population and quality of mitochondria has recently been highlighted to maintain the equilibrium of HSC populations. Leukemic cells also demand tightly regulated metabolism, and shifting the division balance of leukemic cells toward commitment has been considered as a promising therapeutic strategy. A deeper understanding of precisely how specific modes of metabolism control HSC fate is, therefore, of great biological interest, and more importantly will be critical to the development of new therapeutic strategies that target HSC division balance for the treatment of hematological disease.
Collapse
Affiliation(s)
- Kyoko Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Massimo Bonora
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA.
- Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
- Albert Einstein Cancer Center and Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
235
|
Transferrin receptor 2 is a potential novel therapeutic target for β-thalassemia: evidence from a murine model. Blood 2018; 132:2286-2297. [PMID: 30209118 DOI: 10.1182/blood-2018-05-852277] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/31/2018] [Indexed: 12/15/2022] Open
Abstract
β-thalassemias are genetic disorders characterized by anemia, ineffective erythropoiesis, and iron overload. Current treatment of severe cases is based on blood transfusion and iron chelation or allogeneic bone marrow (BM) transplantation. Novel approaches are explored for nontransfusion-dependent patients (thalassemia intermedia) who develop anemia and iron overload. Here, we investigated the erythropoietin (EPO) receptor partner, transferrin receptor 2 (TFR2), as a novel potential therapeutic target. We generated a murine model of thalassemia intermedia specifically lacking BM Tfr2: because their erythroid cells are more susceptible to EPO stimulation, mice show improved erythropoiesis and red blood cell morphology as well as partial correction of anemia and iron overload. The beneficial effects become attenuated over time, possibly due to insufficient iron availability to sustain the enhanced erythropoiesis. Germ line deletion of Tfr2, including haploinsufficiency, had a similar effect in the thalassemic model. Because targeting TFR2 enhances EPO-mediated effects exclusively in cells expressing both receptors, this approach may have advantages over erythropoiesis-stimulating agents in the treatment of other anemias.
Collapse
|
236
|
Spit M, Koo BK, Maurice MM. Tales from the crypt: intestinal niche signals in tissue renewal, plasticity and cancer. Open Biol 2018; 8:rsob.180120. [PMID: 30209039 PMCID: PMC6170508 DOI: 10.1098/rsob.180120] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/17/2018] [Indexed: 02/06/2023] Open
Abstract
Rapidly renewing tissues such as the intestinal epithelium critically depend on the activity of small-sized stem cell populations that continuously generate new progeny to replace lost and damaged cells. The complex and tightly regulated process of intestinal homeostasis is governed by a variety of signalling pathways that balance cell proliferation and differentiation. Accumulating evidence suggests that stem cell control and daughter cell fate determination is largely dictated by the microenvironment. Here, we review recent developments in the understanding of intestinal stem cell dynamics, focusing on the roles, mechanisms and interconnectivity of prime signalling pathways that regulate stem cell behaviour in intestinal homeostasis. Furthermore, we discuss how mutational activation of these signalling pathways endows colorectal cancer cells with niche-independent growth advantages during carcinogenesis.
Collapse
Affiliation(s)
- Maureen Spit
- Cell Biology, Center for Molecular Medicine, UMC Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Bon-Kyoung Koo
- IMBA - Institute of Molecular Biotechnology, Dr Bohr-Gasse 3, 1030 Vienna, Austria
| | - Madelon M Maurice
- Cell Biology, Center for Molecular Medicine, UMC Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands .,Oncode Institute, The Netherlands
| |
Collapse
|
237
|
Theeuwes W, Gosker H, Langen R, Pansters N, Schols A, Remels A. Inactivation of glycogen synthase kinase 3β (GSK-3β) enhances mitochondrial biogenesis during myogenesis. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2913-2926. [DOI: 10.1016/j.bbadis.2018.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/18/2018] [Accepted: 06/04/2018] [Indexed: 12/31/2022]
|
238
|
Zhang C, Skamagki M, Liu Z, Ananthanarayanan A, Zhao R, Li H, Kim K. Biological Significance of the Suppression of Oxidative Phosphorylation in Induced Pluripotent Stem Cells. Cell Rep 2018; 21:2058-2065. [PMID: 29166598 PMCID: PMC5841608 DOI: 10.1016/j.celrep.2017.10.098] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 08/31/2017] [Accepted: 10/25/2017] [Indexed: 12/15/2022] Open
Abstract
We discovered that induced pluripotent stem cell (iPSC) clones generated from aged tissue donors (A-iPSCs) fail to suppress oxidative phosphorylation. Compared to embryonic stem cells (ESCs) and iPSCs generated from young donors (Y-iPSCs), A-iPSCs show poor expression of the pluripotent stem cell-specific glucose transporter 3 (GLUT3) and impaired glucose uptake, making them unable to support the high glucose demands of glycolysis. Persistent oxidative phosphorylation in A-iPSCs generates higher levels of reactive oxygen species (ROS), which leads to excessive elevation of glutathione (a ROS-scavenging metabolite) and a blunted DNA damage response. These phenotypes were recapitulated in Y-iPSCs by inhibiting pyruvate dehydrogenase kinase (PDK) or supplying citrate to activate oxidative phosphorylation. In addition, oxidative phosphorylation in A-iPSC clones depletes citrate, a nuclear source of acetyl group donors for histone acetylation; this consequently alters histone acetylation status. Expression of GLUT3 in A-iPSCs recovers the metabolic defect, DNA damage response, and histone acetylation status.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Molecular Pharmacology & Experimental Therapeutics, Center for Individualized Medicine, Mayo Clinic College of Medicine, Rochester, MN 55902, USA
| | - Maria Skamagki
- Cancer Biology and Genetics Program, The Center for Cell Engineering, The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Zhong Liu
- Department of Biochemistry and Molecular Genetics, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Aparna Ananthanarayanan
- Cancer Biology and Genetics Program, The Center for Cell Engineering, The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Rui Zhao
- Department of Biochemistry and Molecular Genetics, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Hu Li
- Department of Molecular Pharmacology & Experimental Therapeutics, Center for Individualized Medicine, Mayo Clinic College of Medicine, Rochester, MN 55902, USA.
| | - Kitai Kim
- Cancer Biology and Genetics Program, The Center for Cell Engineering, The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY 10065, USA.
| |
Collapse
|
239
|
Rothan HA, Fang S, Mahesh M, Byrareddy SN. Zika Virus and the Metabolism of Neuronal Cells. Mol Neurobiol 2018; 56:2551-2557. [PMID: 30043260 DOI: 10.1007/s12035-018-1263-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 07/18/2018] [Indexed: 02/07/2023]
Abstract
Zika virus (ZIKV) infection is associated with abnormal functions of neuronal cells causing neurological disorders such as microcephaly in the newborns and Guillain-Barré syndrome in the adults. Typically, healthy brain growth is associated with normal neural stem cell proliferation, differentiation, and maturation. This process requires a controlled cellular metabolism that is essential for normal migration, axonal elongation, and dendrite morphogenesis of newly generated neurons. Thus, the remarkable changes in the cellular metabolism during early stages of neuronal stem cell differentiation are crucial for brain development. Recent studies show that ZIKV directly infects neuronal stem cells in the fetus and impairs brain growth. In this review, we highlighted the fact that the activation of P53 and inhibition of the mTOR pathway by ZIKV infection to neuronal stem cells induces early shifting from glycolysis to oxidative phosphorylation (OXPHOS) may induce immature differentiation, apoptosis, and stem cell exhaustion. We hypothesize that ZIKV infection to mature myelin-producing cells and resulting metabolic shift may lead to the development of neurological diseases, such as Guillain-Barré syndrome. Thus, the effects of ZIKV on the cellular metabolism of neuronal cells may lead to the incidence of neurological disorders as observed recently during ZIKV infection.
Collapse
Affiliation(s)
- Hussin A Rothan
- Center for Biomedical Engineering & Technology, School of Medicine, University of Maryland, Baltimore, MD, USA. .,Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA.
| | - Shengyun Fang
- Center for Biomedical Engineering & Technology, School of Medicine, University of Maryland, Baltimore, MD, USA.,Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Mohan Mahesh
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA
| | - Siddappa N Byrareddy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Centre (UNMC), Omaha, NE, 68198-5800, USA.
| |
Collapse
|
240
|
Srivillibhuthur M, Warder BN, Toke NH, Shah PP, Feng Q, Gao N, Bonder EM, Verzi MP. TFAM is required for maturation of the fetal and adult intestinal epithelium. Dev Biol 2018; 439:92-101. [PMID: 29684311 PMCID: PMC5978755 DOI: 10.1016/j.ydbio.2018.04.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 04/18/2018] [Accepted: 04/18/2018] [Indexed: 12/14/2022]
Abstract
During development, the embryo transitions from a metabolism favoring glycolysis to a metabolism favoring mitochondrial respiration. How metabolic shifts regulate developmental processes, or how developmental processes regulate metabolic shifts, remains unclear. To test the requirement of mitochondrial function in developing endoderm-derived tissues, we genetically inactivated the mitochondrial transcription factor, Tfam, using the Shh-Cre driver. Tfam mutants did not survive postnatally, exhibiting defects in lung development. In the developing intestine, TFAM-loss was tolerated until late fetal development, during which the process of villus elongation was compromised. While progenitor cell populations appeared unperturbed, markers of enterocyte maturation were diminished and villi were blunted. Loss of TFAM was also tested in the adult intestinal epithelium, where enterocyte maturation was similarly dependent upon the mitochondrial transcription factor. While progenitor cells in the transit amplifying zone of the adult intestine remained proliferative, intestinal stem cell renewal was dependent upon TFAM, as indicated by molecular profiling and intestinal organoid formation assays. Taken together, these studies point to critical roles for the mitochondrial regulator TFAM for multiple aspects of intestinal development and maturation, and highlight the importance of mitochondrial regulators in tissue development and homeostasis.
Collapse
Affiliation(s)
- Manasa Srivillibhuthur
- Rutgers University, Department of Genetics, Human Genetics Institute of New Jersey (HGINJ), 145 Bevier Road, Piscataway Township, NJ 08854, USA; Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Bailey N Warder
- Rutgers University, Department of Genetics, Human Genetics Institute of New Jersey (HGINJ), 145 Bevier Road, Piscataway Township, NJ 08854, USA
| | - Natalie H Toke
- Rutgers University, Department of Genetics, Human Genetics Institute of New Jersey (HGINJ), 145 Bevier Road, Piscataway Township, NJ 08854, USA
| | - Pooja P Shah
- Rutgers University, Department of Genetics, Human Genetics Institute of New Jersey (HGINJ), 145 Bevier Road, Piscataway Township, NJ 08854, USA
| | - Qiang Feng
- Department of Biological Sciences, Rutgers, The State University of New Jersey, Newark, NJ 07102, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers, The State University of New Jersey, Newark, NJ 07102, USA
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers, The State University of New Jersey, Newark, NJ 07102, USA
| | - Michael P Verzi
- Rutgers University, Department of Genetics, Human Genetics Institute of New Jersey (HGINJ), 145 Bevier Road, Piscataway Township, NJ 08854, USA; Rutgers Cancer Institute of New Jersey (CINJ), 195 Little Albany Street, New Brunswick, NJ 08903, USA.
| |
Collapse
|
241
|
Wei P, Dove KK, Bensard C, Schell JC, Rutter J. The Force Is Strong with This One: Metabolism (Over)powers Stem Cell Fate. Trends Cell Biol 2018; 28:551-559. [PMID: 29555207 PMCID: PMC6005741 DOI: 10.1016/j.tcb.2018.02.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 02/18/2018] [Accepted: 02/20/2018] [Indexed: 12/19/2022]
Abstract
Compared to their differentiated progeny, stem cells are often characterized by distinct metabolic landscapes that emphasize anaerobic glycolysis and a lower fraction of mitochondrial carbohydrate oxidation. Until recently, the metabolic program of stem cells had been thought to be a byproduct of the environment, rather than an intrinsic feature determined by the cell itself. However, new studies highlight the impact of metabolic behavior on the maintenance and function of intestinal stem cells and hair follicle stem cells. This Review summarizes and discusses the evidence that metabolism is not a mere consequence of, but rather influential on stem cell fate.
Collapse
Affiliation(s)
- Peng Wei
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Katja K Dove
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Claire Bensard
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - John C Schell
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Jared Rutter
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| |
Collapse
|
242
|
Induction of quiescence (G0) in bone marrow stromal stem cells enhances their stem cell characteristics. Stem Cell Res 2018; 30:69-80. [DOI: 10.1016/j.scr.2018.05.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 04/22/2018] [Accepted: 05/16/2018] [Indexed: 12/14/2022] Open
|
243
|
Marcu R, Choi YJ, Xue J, Fortin CL, Wang Y, Nagao RJ, Xu J, MacDonald JW, Bammler TK, Murry CE, Muczynski K, Stevens KR, Himmelfarb J, Schwartz SM, Zheng Y. Human Organ-Specific Endothelial Cell Heterogeneity. iScience 2018; 4:20-35. [PMID: 30240741 PMCID: PMC6147238 DOI: 10.1016/j.isci.2018.05.003] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 04/24/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022] Open
Abstract
The endothelium first forms in the blood islands in the extra-embryonic yolk sac and then throughout the embryo to establish circulatory networks that further acquire organ-specific properties during development to support diverse organ functions. Here, we investigated the properties of endothelial cells (ECs), isolated from four human major organs-the heart, lung, liver, and kidneys-in individual fetal tissues at three months' gestation, at gene expression, and at cellular function levels. We showed that organ-specific ECs have distinct expression patterns of gene clusters, which support their specific organ development and functions. These ECs displayed distinct barrier properties, angiogenic potential, and metabolic rate and support specific organ functions. Our findings showed the link between human EC heterogeneity and organ development and can be exploited therapeutically to contribute in organ regeneration, disease modeling, as well as guiding differentiation of tissue-specific ECs from human pluripotent stem cells.
Collapse
Affiliation(s)
- Raluca Marcu
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Yoon Jung Choi
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jun Xue
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Chelsea L Fortin
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Yuliang Wang
- Department of Computer Science & Engineering, University of Washington, Seattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Ryan J Nagao
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jin Xu
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - James W MacDonald
- Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Theo K Bammler
- Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Charles E Murry
- Department of Bioengineering, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Kelly R Stevens
- Department of Bioengineering, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Jonathan Himmelfarb
- Department of Medicine, University of Washington, Seattle, WA, USA; Kidney Research Institute, University of Washington, Seattle, WA, USA
| | | | - Ying Zheng
- Department of Bioengineering, University of Washington, Seattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Kidney Research Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
244
|
Method for measuring extracellular flux from intact polarized epithelial monolayers. Mol Vis 2018; 24:425-433. [PMID: 30034209 PMCID: PMC6031101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 06/23/2018] [Indexed: 12/03/2022] Open
Abstract
PURPOSE The Seahorse XFp platform is widely used for metabolic assessment of cultured cells. Current methods require replating of cells into specialized plates. This is problematic for certain cell types, such as primary human fetal RPE (hfRPE) cells, which must be cultured for months to become properly differentiated. Our goal was to overcome this limitation by devising a method for assaying intact cell monolayers with the Seahorse XFp, without the need for replating. METHODS Primary hfRPE cells were differentiated by prolonged culture on filter inserts. Triangular sections of filters with differentiated cells attached were excised, transferred to XFp cell culture miniplate wells, immobilized at the bottoms, and subjected to mitochondrial stress tests. Replated cells were measured for comparison. Differentiated hfRPE cells were challenged or not with bovine photoreceptor outer segments (POS), and mitochondrial stress tests were performed 3.5 h later, after filter excision and transfer to assay plates. RESULTS Differentiated hfRPE cells assayed following filter excision demonstrated increased maximal respiration, increased spare respiration capacity, and increased extracellular acidification rate (ECAR) relative to replated controls. hfRPE cells challenged with POS exhibited increased maximal respiration and spare capacity, with no apparent change in the ECAR, relative to untreated controls. CONCLUSIONS We have developed a method to reproducibly assay intact, polarized monolayers of hfRPE cells with the Seahorse XFp platform and have shown that the method yields more robust metabolic measurements compared to standard methods and is suitable for assessing the consequences of prolonged perturbations of differentiated cells. We expect our approach to be useful for a variety of studies involving metabolic assessment of adherent cells cultured on filters.
Collapse
|
245
|
Zaveri L, Dhawan J. Cycling to Meet Fate: Connecting Pluripotency to the Cell Cycle. Front Cell Dev Biol 2018; 6:57. [PMID: 29974052 PMCID: PMC6020794 DOI: 10.3389/fcell.2018.00057] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 05/14/2018] [Indexed: 01/26/2023] Open
Abstract
Pluripotent stem cells are characterized by their high proliferative rates, their ability to self-renew and their potential to differentiate to all the three germ layers. This rapid proliferation is brought about by a highly modified cell cycle that allows the cells to quickly shuttle from DNA synthesis to cell division, by reducing the time spent in the intervening gap phases. Many key regulators that define the somatic cell cycle are either absent or exhibit altered behavior, allowing the pluripotent cell to bypass cell cycle checkpoints typical of somatic cells. Experimental analysis of this modified stem cell cycle has been challenging due to the strong link between rapid proliferation and pluripotency, since perturbations to the cell cycle or pluripotency factors result in differentiation. Despite these hurdles, our understanding of this unique cell cycle has greatly improved over the past decade, in part because of the availability of new technologies that permit the analysis of single cells in heterogeneous populations. This review aims to highlight some of the recent discoveries in this area with a special emphasis on different states of pluripotency. We also discuss the highly interlinked network that connects pluripotency factors and key cell cycle genes and review evidence for how this interdependency may promote the rapid cell cycle. This issue gains translational importance since disruptions in stem cell proliferation and differentiation can impact disorders at opposite ends of a spectrum, from cancer to degenerative disease.
Collapse
Affiliation(s)
- Lamuk Zaveri
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India.,CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India.,Manipal Academy of Higher Education, Manipal, India
| | - Jyotsna Dhawan
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India.,CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India
| |
Collapse
|
246
|
Yi M, Li J, Chen S, Cai J, Ban Y, Peng Q, Zhou Y, Zeng Z, Peng S, Li X, Xiong W, Li G, Xiang B. Emerging role of lipid metabolism alterations in Cancer stem cells. J Exp Clin Cancer Res 2018; 37:118. [PMID: 29907133 PMCID: PMC6003041 DOI: 10.1186/s13046-018-0784-5] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/28/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) or tumor-initiating cells (TICs) represent a small population of cancer cells with self-renewal and tumor-initiating properties. Unlike the bulk of tumor cells, CSCs or TICs are refractory to traditional therapy and are responsible for relapse or disease recurrence in cancer patients. Stem cells have distinct metabolic properties compared to differentiated cells, and metabolic rewiring contributes to self-renewal and stemness maintenance in CSCs. MAIN BODY Recent advances in metabolomic detection, particularly in hyperspectral-stimulated raman scattering microscopy, have expanded our knowledge of the contribution of lipid metabolism to the generation and maintenance of CSCs. Alterations in lipid uptake, de novo lipogenesis, lipid droplets, lipid desaturation, and fatty acid oxidation are all clearly implicated in CSCs regulation. Alterations on lipid metabolism not only satisfies the energy demands and biomass production of CSCs, but also contributes to the activation of several important oncogenic signaling pathways, including Wnt/β-catenin and Hippo/YAP signaling. In this review, we summarize the current progress in this attractive field and describe some recent therapeutic agents specifically targeting CSCs based on their modulation of lipid metabolism. CONCLUSION Increased reliance on lipid metabolism makes it a promising therapeutic strategy to eliminate CSCs. Targeting key players of fatty acids metabolism shows promising to anti-CSCs and tumor prevention effects.
Collapse
Affiliation(s)
- Mei Yi
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, 410013 Hunan China
- Department of Dermatology, Xiangya hospital of Central South University, Changsha, 410008 China
| | - Junjun Li
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, 410013 Hunan China
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078 China
| | - Shengnan Chen
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, 410013 Hunan China
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078 China
| | - Jing Cai
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, 410013 Hunan China
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078 China
| | - Yuanyuan Ban
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, 410013 Hunan China
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078 China
| | - Qian Peng
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, 410013 Hunan China
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078 China
| | - Ying Zhou
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, 410013 Hunan China
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078 China
| | - Zhaoyang Zeng
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, 410013 Hunan China
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078 China
| | - Shuping Peng
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, 410013 Hunan China
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078 China
| | - Xiaoling Li
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, 410013 Hunan China
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078 China
| | - Wei Xiong
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, 410013 Hunan China
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078 China
| | - Guiyuan Li
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, 410013 Hunan China
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078 China
| | - Bo Xiang
- Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, The Central South University, Changsha, 410013 Hunan China
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078 China
| |
Collapse
|
247
|
Ito K, Ito K. Hematopoietic stem cell fate through metabolic control. Exp Hematol 2018; 64:1-11. [PMID: 29807063 DOI: 10.1016/j.exphem.2018.05.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/18/2018] [Accepted: 05/19/2018] [Indexed: 01/02/2023]
Abstract
Hematopoietic stem cells maintain a quiescent state in the bone marrow to preserve their self-renewal capacity, but also undergo cell divisions as required. Organelles such as the mitochondria sustain cumulative damage during these cell divisions and this damage may eventually compromise the cells' self-renewal capacity. Hematopoietic stem cell divisions result in either self-renewal or differentiation, with the balance between the two affecting hematopoietic homeostasis directly; however, the heterogeneity of available hematopoietic stem cell-enriched fractions, together with the technical challenges of observing hematopoietic stem cell behavior, has long hindered the analysis of individual hematopoietic stem cells and prevented the elucidation of this process. Recent advances in genetic models, metabolomics analyses, and single-cell approaches have revealed the contributions made to hematopoietic stem cell self-renewal by metabolic cues, mitochondrial biogenesis, and autophagy/mitophagy, which have highlighted mitochondrial quality control as a key factor in the equilibrium of hematopoietic stem cells. A deeper understanding of precisely how specific modes of metabolism control hematopoietic stem cells fate at the single-cell level is therefore not only of great biological interest, but will also have clear clinical implications for the development of therapies for hematological diseases.
Collapse
Affiliation(s)
- Kyoko Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA; Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA; Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Albert Einstein Cancer Center and Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
248
|
Abstract
Purpose of Review Functional decline of hematopoiesis that occurs in the elderly, or in patients who receive therapies that trigger cellular senescence effects, results in a progressive reduction in the immune response and an increased incidence of myeloid malignancy. Intracellular signals in hematopoietic stem cells and progenitors (HSC/P) mediate systemic, microenvironment, and cell-intrinsic effector aging signals that induce their decline. This review intends to summarize and critically review our advances in the understanding of the intracellular signaling pathways responsible for HSC decline during aging and opportunities for intervention. Recent Findings For a long time, aging of HSC has been thought to be an irreversible process imprinted in stem cells due to the cell intrinsic nature of aging. However, recent murine models and human correlative studies provide evidence that aging is associated with molecular signaling pathways, including oxidative stress, metabolic dysfunction, loss of polarity and an altered epigenome. These signaling pathways provide potential targets for prevention or reversal of age-related changes. Summary Here we review our current understanding of the signalling pathways that are differentially activated or repressed during HSC/P aging, focusing on the oxidative, metabolic, biochemical and structural consequences downstream, and cell-intrinsic, systemic, and environmental influences.
Collapse
|
249
|
Khacho M, Slack RS. Mitochondrial and Reactive Oxygen Species Signaling Coordinate Stem Cell Fate Decisions and Life Long Maintenance. Antioxid Redox Signal 2018; 28:1090-1101. [PMID: 28657337 DOI: 10.1089/ars.2017.7228] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Significance: Recent discoveries in mitochondrial biology have transformed and further solidified the importance of mitochondria in development, aging, and disease. Within the realm of regenerative and stem cell research, these recent advances have brought forth new concepts that revolutionize our understanding of metabolic and redox states in the establishment of cellular identity and fate decisions. Recent Advances: Mitochondrial metabolism, morphology, and cellular redox states are dynamic characteristics that undergo shifts during stem cell differentiation. Although it was once thought that this was solely because of changing metabolic needs of differentiating cells, it is now clear that these events are driving forces in the regulation of stem cell identity and fate decisions. Critical Issues: Although recent discoveries have placed mitochondrial function and physiological reactive oxygen species (ROS) at the forefront for the regulation of stem cell self-renewal, how this may impact tissue homeostasis and regenerative capacity is poorly understood. In addition, the role of mitochondria and ROS on the maintenance of a stem cell population in many degenerative diseases and during aging is not clear, despite the fact that mitochondrial dysfunction and elevated ROS levels are commonly observed in these conditions. Future Directions: Given the newly established role for mitochondria and ROS in stem cell self-renewal capacity, special attention should now be directed in understanding how this would impact the development and progression of aging and diseases, whereby mitochondrial and ROS defects are a prominent factor. Antioxid. Redox Signal. 28, 1090-1101.
Collapse
Affiliation(s)
- Mireille Khacho
- Department of Cellular and Molecular Medicine, Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| | - Ruth S Slack
- Department of Cellular and Molecular Medicine, Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| |
Collapse
|
250
|
Heber-Katz E, Messersmith P. Drug delivery and epimorphic salamander-type mouse regeneration: A full parts and labor plan. Adv Drug Deliv Rev 2018. [PMID: 29524586 DOI: 10.1016/j.addr.2018.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The capacity to regenerate entire body parts, tissues, and organs had generally been thought to be lost in evolution with very few exceptions (e.g. the liver) surviving in mammals. The discovery of the MRL mouse and the elucidation of the underlying molecular pathway centering around hypoxia inducible factor, HIF-1α, has allowed a drug and materials approach to regeneration in mice and hopefully humans. The HIF-1α pathway is ancient and permitted the transition from unicellular to multicellular organisms. Furthermore, HIF-1α and its regulation by PHDs, important oxygen sensors in the cell, provides a perfect drug target. We review the historical background of regeneration biology, the discovery of the MRL mouse, and its underlying biology, and novel approaches to drugs, targets, and delivery systems (see Fig. 1).
Collapse
|