1
|
Woods PS, Mutlu GM. Differences in glycolytic metabolism between tissue-resident alveolar macrophages and recruited lung macrophages. Front Immunol 2025; 16:1535796. [PMID: 40092977 PMCID: PMC11906440 DOI: 10.3389/fimmu.2025.1535796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Immunometabolism has emerged as a key area of focus in immunology and has the potential to lead to new treatments for immune-related diseases. It is well-established that glycolytic metabolism is essential for adaptation to hypoxia and for macrophage inflammatory function. Macrophages have been shown to upregulate their glycolytic metabolism in response to pathogens and pathogen-associated molecular patterns such as LPS. As a direct link to the external environment, the lungs' distinctive nutrient composition and multiple macrophage subtypes provide a unique opportunity to study macrophage metabolism. This review aims to highlight how the steady-state airway and severely inflamed airway offer divergent environments for macrophage glycolytic metabolism. We describe the differences in glycolytic metabolism between tissue-resident alveolar macrophages, and other lung macrophages at steady-state and during inflammation/injury. We also provide an overview of experimental guidelines on how to assess metabolism at the cellular level using Seahorse-based bioenergetic analysis including a review of pharmacologic agents used to inhibit or activate glycolysis.
Collapse
Affiliation(s)
| | - Gökhan M. Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University
of Chicago, Chicago, IL, United States
| |
Collapse
|
2
|
Joly A, Schott A, Phadke I, Gonzalez-Menendez P, Kinet S, Taylor N. Beyond ATP: Metabolite Networks as Regulators of Physiological and Pathological Erythroid Differentiation. Physiology (Bethesda) 2025; 40:0. [PMID: 39226028 DOI: 10.1152/physiol.00035.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
Hematopoietic stem cells (HSCs) possess the capacity for self-renewal and the sustained production of all mature blood cell lineages. It has been well established that a metabolic rewiring controls the switch of HSCs from a self-renewal state to a more differentiated state, but it is only recently that we have appreciated the importance of metabolic pathways in regulating the commitment of progenitors to distinct hematopoietic lineages. In the context of erythroid differentiation, an extensive network of metabolites, including amino acids, sugars, nucleotides, fatty acids, vitamins, and iron, is required for red blood cell (RBC) maturation. In this review, we highlight the multifaceted roles via which metabolites regulate physiological erythropoiesis as well as the effects of metabolic perturbations on erythroid lineage commitment and differentiation. Of note, the erythroid differentiation process is associated with an exceptional breadth of solute carrier (SLC) metabolite transporter upregulation. Finally, we discuss how recent research, revealing the critical impact of metabolic reprogramming in diseases of disordered and ineffective erythropoiesis, has created opportunities for the development of novel metabolic-centered therapeutic strategies.
Collapse
Affiliation(s)
- Axel Joly
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | - Arthur Schott
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | - Ira Phadke
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
- Pediatric Oncology Branch, CCR, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Pedro Gonzalez-Menendez
- Departamento de Morfologia y Biologia Celular, Instituto Universitario de Oncologia del Principado de Asturias (IUOPA), Universidad de Oviedo, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Sandrina Kinet
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | - Naomi Taylor
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
- Pediatric Oncology Branch, CCR, NCI, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
3
|
Liu W, Liu X, Li L, Tai Z, Li G, Liu JX. EPC1/2 regulate hematopoietic stem and progenitor cell proliferation by modulating H3 acetylation and DLST. iScience 2024; 27:109263. [PMID: 38439957 PMCID: PMC10910311 DOI: 10.1016/j.isci.2024.109263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/21/2023] [Accepted: 02/14/2024] [Indexed: 03/06/2024] Open
Abstract
Enhancers of polycomb 1 (EPC1) and 2 (EPC2) are involved in multiple biological processes as components of histone acetyltransferases/deacetylase complexes and transcriptional cofactors, and their dysfunction was associated with developmental defects and diseases. However, it remains unknown how their dysfunction induces hematopoietic stem and progenitor cell (HSPC) defects. Here, we show that depletion of EPC1/2 significantly reduced the number of hematopoietic stem and progenitor cells (HSPCs) in the aorta-gonad mesonephros and caudal hematopoietic tissue regions by impairing HSPC proliferation, and consistently downregulated the expression of HSPC genes in K562 cells. This study demonstrates the functions of EPC1/2 in regulating histone H3 acetylation, and in regulating DLST (dihydrolipoamide S-succinyltransferase) via H3 acetylation and cooperating with transcription factors serum response factor and FOXR2 together, and in the subsequent HSPC emergence and proliferation. Our results demonstrate the essential roles of EPC1/2 in regulating H3 acetylation, and DLST as a linkage between EPC1 and EPC2 with mitochondria metabolism, in HSPC emergence and proliferation.
Collapse
Affiliation(s)
- WenYe Liu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China
| | - Xi Liu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China
| | - LingYa Li
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China
| | - ZhiPeng Tai
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China
| | - GuoLiang Li
- College of Informatics, Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, Huazhong Agricultural University, Wuhan 430070, China
| | - Jing-Xia Liu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
4
|
Xu Y, Chiang YH, Ho PC, Vannini N. Mitochondria Dictate Function and Fate of HSCs and T Cells. Cancer Immunol Res 2023; 11:1303-1313. [PMID: 37789763 DOI: 10.1158/2326-6066.cir-22-0685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 01/23/2023] [Accepted: 08/16/2023] [Indexed: 10/05/2023]
Abstract
Hematopoietic stem cells (HSC) and T cells are intimately related, lineage-dependent cell populations that are extensively used as therapeutic products for the treatment of hematologic malignancies and certain types of solid tumors. These cellular therapies can be life-saving treatments; however, their efficacies are often limited by factors influencing their activity and cellular properties. Among these factors is mitochondrial metabolism, which influences the function and fate commitment of both HSCs and T cells. Mitochondria, besides being the "cellular powerhouse," provide metabolic intermediates that are used as substrates for epigenetic modifications and chromatin remodeling, thus, driving cell fate decisions during differentiation. Moreover, mitochondrial fitness and mitochondrial quality control mechanisms are closely related to cellular function, and impairment of these mitochondrial properties associates with cellular dysfunction due to factors such as T-cell exhaustion and aging. Here, we give an overview of the role of mitochondria in shaping the behavior of these lineage-related cell populations. Moreover, we discuss the potential of novel mitochondria-targeting strategies for enhancing HSC- and T cell-based cancer immunotherapies and highlight how design and application of such approaches requires consideration of the metabolic similarities and differences between HSCs and T cells. See related article on p. 1302.
Collapse
Affiliation(s)
- Yingxi Xu
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Yi-Hsuan Chiang
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Nicola Vannini
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
5
|
Mistry JJ, Bowles K, Rushworth SA. HSC-derived fatty acid oxidation in steady-state and stressed hematopoiesis. Exp Hematol 2023; 117:1-8. [PMID: 36223830 DOI: 10.1016/j.exphem.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/20/2022] [Accepted: 10/04/2022] [Indexed: 01/10/2023]
Abstract
Metabolism impacts all cellular functions and plays a fundamental role in physiology. Metabolic regulation of hematopoiesis is dynamically regulated under steady-state and stress conditions. It is clear that hematopoietic stem cells (HSCs) impose different energy demands and flexibility during maintenance compared with stressed conditions. However, the cellular and molecular mechanisms underlying metabolic regulation in HSCs remain poorly understood. In this review, we focus on defining the role of fatty acid oxidation (FAO) in HSCs. We first review the existing literature describing FAO in HSCs under steady-state hematopoiesis. Next, we describe the models used to examine HSCs under stress conditions, and, finally, we describe how infection causes a shift toward FAO in HSCs and the impact of using this pathway on emergency hematopoiesis.
Collapse
Affiliation(s)
| | - Kristian Bowles
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, United Kingdom; Department of Haematology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Stuart A Rushworth
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, United Kingdom.
| |
Collapse
|
6
|
Arif T. Lysosomes and Their Role in Regulating the Metabolism of Hematopoietic Stem Cells. BIOLOGY 2022; 11:1410. [PMID: 36290314 PMCID: PMC9598322 DOI: 10.3390/biology11101410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/26/2022]
Abstract
Hematopoietic stem cells (HSCs) have the capacity to renew blood cells at all stages of life and are largely quiescent at a steady state. It is essential to understand the processes that govern quiescence in HSCs to enhance bone marrow transplantation. It is hypothesized that in their quiescent state, HSCs primarily use glycolysis for energy production rather than mitochondrial oxidative phosphorylation (OXPHOS). In addition, the HSC switch from quiescence to activation occurs along a continuous developmental path that is driven by metabolism. Specifying the metabolic regulation pathway of HSC quiescence will provide insights into HSC homeostasis for therapeutic application. Therefore, understanding the metabolic demands of HSCs at a steady state is key to developing innovative hematological therapeutics. Lysosomes are the major degradative organelle in eukaryotic cells. Catabolic, anabolic, and lysosomal function abnormalities are connected to an expanding list of diseases. In recent years, lysosomes have emerged as control centers of cellular metabolism, particularly in HSC quiescence, and essential regulators of cell signaling have been found on the lysosomal membrane. In addition to autophagic processes, lysosomal activities have been shown to be crucial in sustaining quiescence by restricting HSCs access to a nutritional reserve essential for their activation into the cell cycle. Lysosomal activity may preserve HSC quiescence by altering glycolysis-mitochondrial biogenesis. The understanding of HSC metabolism has significantly expanded over the decade, revealing previously unknown requirements of HSCs in both their dividing (active) and quiescent states. Therefore, understanding the role of lysosomes in HSCs will allow for the development of innovative treatment methods based on HSCs to fight clonal hematopoiesis and HSC aging.
Collapse
Affiliation(s)
- Tasleem Arif
- Department of Cell, Developmental & Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
7
|
Wang N, Chen C, Cheng Y, Fu Y, Zhong Z, Yang Y, Lv L, Chen H, Huang J, Duan Y. Hypoxia drives hematopoiesis with the enhancement of T lineage through eliciting arterial specification of hematopoietic endothelial progenitors from hESC. Stem Cell Res Ther 2022; 13:282. [PMID: 35765115 PMCID: PMC9241298 DOI: 10.1186/s13287-022-02967-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/19/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Hematopoietic stem cells are able to self-renew and differentiate into all blood cell lineages. Hematopoietic stem cell transplantation is a mainstay of life-saving therapy for hematopoietic malignancies and hypoproliferative disorders. In vitro hematopoietic differentiation of human pluripotent stem cells (hPSCs) is a promising approach for modeling hematopoietic development and cell replacement therapies. Although using hPSCs to derive hematopoietic progenitor cells has achieved some successes in the past, differentiation from hPSCs to produce all hematopoietic cells which can provide robust long-term multilineage engraftment is still very difficult. Here, we reported a novel culture system for hematopoietic differentiation from human embryonic stem cells (hESCs) with optimal cytokines combinations under hypoxia condition. METHODS In vitro production of T lineage hematopoietic stem/progenitor cells from hESCs by using hypoxia differentiation system, the effects and the potential mechanism of hypoxia promoting T lineage hematopoiesis were investigated by RT-qPCR validation, cell cycle assay and flow cytometry analysis. RESULTS Using our differentiation system, almost 80% CD45+ cells generated from hESCs were hematopoietic cells and particularly could be further induced into CD3+TCRαβ+ T cells in vitro. We detected more CD34+CD144+ hematopoietic endothelial progenitors (HEPs) induced from hESCs than those in normoxia conditions, and the early HEPs-related gene DLL4 was upregulated by enhancing the hypoxia signaling via potential HIF-1α/NOTCH1/DLL4 axis to enhance arterial feature, thus drove T lineage during the hematopoiesis. Strikingly, hematopoietic cells generated in our system exhibited the potential for all multilineage reconstruction including lymphoid, myeloid and erythroid lineages in vivo by transplantation assay. CONCLUSION Our results demonstrated that hypoxia plays an important role in T lineage hematopoiesis by promoting the expression of arterial endothelial gene DLL4 and upregulation of NOTCH1 through the activation of the HIF-1α signaling pathway. These results provide a significant approach for in vitro and in vivo production of fully functional hematopoietic stem/progenitor cells from hESCs.
Collapse
Affiliation(s)
- Ning Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, People's Republic of China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, Higher Education Mega Center, South China University of Technology, No.382 Waihuan East Road, Suite 406, Guangzhou, 510006, People's Republic of China
| | - Chuxin Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, People's Republic of China
| | - Yang Cheng
- Department of Gynaecology and Obstetrics, Guangzhou First People's Hospital, Guangzhou, 510180, People's Republic of China
| | - Yingjie Fu
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, Higher Education Mega Center, South China University of Technology, No.382 Waihuan East Road, Suite 406, Guangzhou, 510006, People's Republic of China
| | - Zhiyong Zhong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, People's Republic of China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, Higher Education Mega Center, South China University of Technology, No.382 Waihuan East Road, Suite 406, Guangzhou, 510006, People's Republic of China
| | - Yu Yang
- Department of Gynaecology and Obstetrics, Guangzhou First People's Hospital, Guangzhou, 510180, People's Republic of China
| | - Ling Lv
- Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guang Zhou Road, Nanjing, 210029, People's Republic of China.
| | - Honglin Chen
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, Higher Education Mega Center, South China University of Technology, No.382 Waihuan East Road, Suite 406, Guangzhou, 510006, People's Republic of China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, People's Republic of China.
| | - Jian Huang
- Coriell Institute for Medical Research, Camden, NJ, USA.
- Cooper Medical School of Rowan University, Camden, NJ, USA.
| | - Yuyou Duan
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, Higher Education Mega Center, South China University of Technology, No.382 Waihuan East Road, Suite 406, Guangzhou, 510006, People's Republic of China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
8
|
Gu LF, Chen JQ, Lin QY, Yang YZ. Roles of mitochondrial unfolded protein response in mammalian stem cells. World J Stem Cells 2021; 13:737-752. [PMID: 34367475 PMCID: PMC8316864 DOI: 10.4252/wjsc.v13.i7.737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/13/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
The mitochondrial unfolded protein response (UPRmt) is an evolutionarily conserved adaptive mechanism for improving cell survival under mitochondrial stress. Under physiological and pathological conditions, the UPRmt is the key to maintaining intracellular homeostasis and proteostasis. Important roles of the UPRmt have been demonstrated in a variety of cell types and in cell development, metabolism, and immune processes. UPRmt dysfunction leads to a variety of pathologies, including cancer, inflammation, neurodegenerative disease, metabolic disease, and immune disease. Stem cells have a special ability to self-renew and differentiate into a variety of somatic cells and have been shown to exist in a variety of tissues. These cells are involved in development, tissue renewal, and some disease processes. Although the roles and regulatory mechanisms of the UPRmt in somatic cells have been widely reported, the roles of the UPRmt in stem cells are not fully understood. The roles and functions of the UPRmt depend on stem cell type. Therefore, this paper summarizes the potential significance of the UPRmt in embryonic stem cells, tissue stem cells, tumor stem cells, and induced pluripotent stem cells. The purpose of this review is to provide new insights into stem cell differentiation and tumor pathogenesis.
Collapse
Affiliation(s)
- Li-Fang Gu
- Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, Key Laboratory of Reproduction and Genetics in Ningxia, Department of Histology and Embryology, School of Basic Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Jia-Qi Chen
- Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, Key Laboratory of Reproduction and Genetics in Ningxia, Department of Histology and Embryology, School of Basic Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Qing-Yin Lin
- Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, Key Laboratory of Reproduction and Genetics in Ningxia, Department of Histology and Embryology, School of Basic Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yan-Zhou Yang
- Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, Key Laboratory of Reproduction and Genetics in Ningxia, Department of Histology and Embryology, School of Basic Medicine, Ningxia Medical University, Yinchuan 750001, Ningxia Hui Autonomous Region, China.
| |
Collapse
|
9
|
Gonzalez-Menendez P, Romano M, Yan H, Deshmukh R, Papoin J, Oburoglu L, Daumur M, Dumé AS, Phadke I, Mongellaz C, Qu X, Bories PN, Fontenay M, An X, Dardalhon V, Sitbon M, Zimmermann VS, Gallagher PG, Tardito S, Blanc L, Mohandas N, Taylor N, Kinet S. An IDH1-vitamin C crosstalk drives human erythroid development by inhibiting pro-oxidant mitochondrial metabolism. Cell Rep 2021; 34:108723. [PMID: 33535038 PMCID: PMC9169698 DOI: 10.1016/j.celrep.2021.108723] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/26/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
The metabolic changes controlling the stepwise differentiation of hematopoietic stem and progenitor cells (HSPCs) to mature erythrocytes are poorly understood. Here, we show that HSPC development to an erythroid-committed proerythroblast results in augmented glutaminolysis, generating alpha-ketoglutarate (αKG) and driving mitochondrial oxidative phosphorylation (OXPHOS). However, sequential late-stage erythropoiesis is dependent on decreasing αKG-driven OXPHOS, and we find that isocitrate dehydrogenase 1 (IDH1) plays a central role in this process. IDH1 downregulation augments mitochondrial oxidation of αKG and inhibits reticulocyte generation. Furthermore, IDH1 knockdown results in the generation of multinucleated erythroblasts, a morphological abnormality characteristic of myelodysplastic syndrome and congenital dyserythropoietic anemia. We identify vitamin C homeostasis as a critical regulator of ineffective erythropoiesis; oxidized ascorbate increases mitochondrial superoxide and significantly exacerbates the abnormal erythroblast phenotype of IDH1-downregulated progenitors, whereas vitamin C, scavenging reactive oxygen species (ROS) and reprogramming mitochondrial metabolism, rescues erythropoiesis. Thus, an IDH1-vitamin C crosstalk controls terminal steps of human erythroid differentiation.
Collapse
Affiliation(s)
- Pedro Gonzalez-Menendez
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France.
| | - Manuela Romano
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Hongxia Yan
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; New York Blood Center, New York, NY, USA
| | - Ruhi Deshmukh
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Julien Papoin
- The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Leal Oburoglu
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Marie Daumur
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Anne-Sophie Dumé
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Ira Phadke
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France; Pediatric Oncology Branch, NCI, CCR, NIH, Bethesda, MD, USA
| | - Cédric Mongellaz
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Xiaoli Qu
- New York Blood Center, New York, NY, USA
| | - Phuong-Nhi Bories
- Service d'Hématologie Biologique, Assistance Publique-Hôpitaux de Paris, Institut Cochin, Paris, France
| | - Michaela Fontenay
- Laboratory of Excellence GR-Ex, Paris 75015, France; Service d'Hématologie Biologique, Assistance Publique-Hôpitaux de Paris, Institut Cochin, Paris, France
| | - Xiuli An
- New York Blood Center, New York, NY, USA
| | - Valérie Dardalhon
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Marc Sitbon
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Valérie S Zimmermann
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Patrick G Gallagher
- Departments of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Saverio Tardito
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Lionel Blanc
- The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | | | - Naomi Taylor
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France; Pediatric Oncology Branch, NCI, CCR, NIH, Bethesda, MD, USA.
| | - Sandrina Kinet
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France.
| |
Collapse
|
10
|
Rodriguez C, Simon V, Conget P, Vega IA. Both quiescent and proliferating cells circulate in the blood of the invasive apple snail Pomacea canaliculata. FISH & SHELLFISH IMMUNOLOGY 2020; 107:95-103. [PMID: 32966893 DOI: 10.1016/j.fsi.2020.09.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/13/2020] [Accepted: 09/17/2020] [Indexed: 06/11/2023]
Abstract
Gastropod hematopoiesis occurs at specialized tissues in some species, but the evidence also suggests that hemocyte generation is maybe widespread in the connective tissues or the blood system in others. In Ampullariidae (Caenogastropoda), both the kidney and the lung contain putative hematopoietic cells, which react to immune challenges. In the current study, we wanted to explore if hematopoiesis occurs in the blood of Pomacea canaliculata. Thus, we obtained circulating hemocytes from donor animals and tested their ability to proliferate in the blood of conspecific recipients. We tracked cell proliferation by labeling the donors' hemocytes with the fluorescent cell proliferation marker carboxyfluorescein diacetate succinimidyl ester (CFSE). Transferred CFSE-labeled hemocytes survived and proliferated into the recipients' circulation for at least 17 days. We also determined the cell cycle status of circulating hemocytes by using the propidium iodide (PI) and acridine orange (AO) staining methods. Flow cytometry analyses showed that most PI-stained hemocytes were in the G1 phase (~96%), while a lower proportion of cells were through the G2/S-M transition (~4%). When we instead used AO-staining, we further distinguished a subpopulation of cells (~5%) of low size, complexity-granularity, and RNA content. We regarded this subpopulation as quiescent cells. In separate experimental sets, we complemented these findings by assessing in circulating hemocytes two evolutionary conserved features of quiescent, undifferentiated cells. First, we used JC-1 staining to determine the mitochondrial membrane potential (Ψm) of circulating hemocytes, which is expected to be low in quiescent cells. Most hemocytes (~87%) showed high aggregation of JC-1, which indicates a high Ψm. Besides that, a small hemocyte subpopulation (~11%) showed low aggregation of the dye, thus indicating a low Ψm. It is known that the transition from a quiescent to a proliferating state associates with an increase of the Ψm. The specificity of these changes was here controlled by membrane depolarization with the Ψm disruptor CCCP. Second, we stained hemocytes with Hoechst33342 dye to determine the efflux activity of ABC transporters, which participate in the multixenobiotic resistance system characteristic of undifferentiated cells. Most hemocytes (>99%) showed a low dye-efflux activity, but a small proportion of cells (0.06-0.12%) showed a high dye-efflux activity, which was significantly inhibited by 100 and 500 μM verapamil, and thus is indicative of an undifferentiated subpopulation of circulating hemocytes. Taken together, our results suggest that, among circulating hemocytes, there are cells with the ability to proliferate or to stay in a quiescent state and behave as progenitor cells later, either in the circulation or the hematopoietic tissues/organs.
Collapse
Affiliation(s)
- Cristian Rodriguez
- IHEM, CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina; Universidad Nacional de Cuyo, Facultad de Ciencias Médicas, Instituto de Fisiología, Mendoza, Argentina; Universidad Nacional de Cuyo, Facultad de Ciencias Exactas y Naturales, Departamento de Biología, Mendoza, Argentina
| | - Valeska Simon
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, 7710162, Santiago, Chile
| | - Paulette Conget
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, 7710162, Santiago, Chile.
| | - Israel A Vega
- IHEM, CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina; Universidad Nacional de Cuyo, Facultad de Ciencias Médicas, Instituto de Fisiología, Mendoza, Argentina; Universidad Nacional de Cuyo, Facultad de Ciencias Exactas y Naturales, Departamento de Biología, Mendoza, Argentina.
| |
Collapse
|
11
|
Vlaski-Lafarge M, Labat V, Brandy A, Refeyton A, Duchez P, Rodriguez L, Gibson N, Brunet de la Grange P, Ivanovic Z. Normal Hematopoetic Stem and Progenitor Cells Can Exhibit Metabolic Flexibility Similar to Cancer Cells. Front Oncol 2020; 10:713. [PMID: 32528878 PMCID: PMC7247845 DOI: 10.3389/fonc.2020.00713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 04/15/2020] [Indexed: 02/01/2023] Open
Abstract
It is known that cancer stem cells (CSCs) with the largest proliferative capacity survive the anoxic and/or ischemic conditions present inside tumorous tissue. In this study we test whether normal stem cells can survive under the same conditions due to cancer cell-like metabolic adaptations. We cultivated a CD34+ population with a majority of hematopoietic progenitors, and a CD34+CD38lowCD133+CD90+CD45RA− population, highly enriched in hematopoietic stem cells (HSCs), under anoxic, anoxic/aglycemic (“ischemia-like”), or physiological conditions (3% O2). Results showed, despite a reduction in total cell fold expansion proportionate to the decrease in O2 concentration; CD34+ cells, aldehyde dehydrogenase-expressing primitive cells, and committed progenitors expanded, even in anoxia. Interestingly, under ischemia-like conditions, stem and CD34+ cell populations are maintained at day-0 level. Cell-cycle analysis further revealed an accumulation of cells in the G0/G1 phase in anoxia or anoxia/aglycemia, with a fraction of cells (~40%) actively cycling (SG2M phases). Also stem cell analysis showed that in these conditions a long-term Scid Repopulating activity was equal to that found with 3% O2. In addition stem cells with the highest proliferative capacity were maintained in anoxia/aglycemia and in anoxia. The estimated ATP profile, active mitochondrial content, and succinate accumulation are indicative of anaerobic mitochondrial respiration in both HSCs and CD34+ progenitors under ischemia-like conditions. We demonstrate here that primitive hematopoietic cells show similar metabolic flexibility to CSCs, allowing them to survive a lack of O2 and O2/glucose. Our study reveals that this feature is not the consequence of malignant transformation, but an attribute of stemness.
Collapse
Affiliation(s)
- Marija Vlaski-Lafarge
- R&D Department, Etablissement Français du Sang Nouvelle Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux, Bordeaux, France
| | - Veronique Labat
- R&D Department, Etablissement Français du Sang Nouvelle Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux, Bordeaux, France
| | - Alexandra Brandy
- R&D Department, Etablissement Français du Sang Nouvelle Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux, Bordeaux, France
| | - Alice Refeyton
- R&D Department, Etablissement Français du Sang Nouvelle Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux, Bordeaux, France
| | - Pascale Duchez
- R&D Department, Etablissement Français du Sang Nouvelle Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux, Bordeaux, France
| | - Laura Rodriguez
- R&D Department, Etablissement Français du Sang Nouvelle Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux, Bordeaux, France
| | - Nyere Gibson
- R&D Department, Etablissement Français du Sang Nouvelle Aquitaine, Bordeaux, France
| | - Philippe Brunet de la Grange
- R&D Department, Etablissement Français du Sang Nouvelle Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux, Bordeaux, France
| | - Zoran Ivanovic
- R&D Department, Etablissement Français du Sang Nouvelle Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux, Bordeaux, France
| |
Collapse
|
12
|
Vannini N, Campos V, Girotra M, Trachsel V, Rojas-Sutterlin S, Tratwal J, Ragusa S, Stefanidis E, Ryu D, Rainer PY, Nikitin G, Giger S, Li TY, Semilietof A, Oggier A, Yersin Y, Tauzin L, Pirinen E, Cheng WC, Ratajczak J, Canto C, Ehrbar M, Sizzano F, Petrova TV, Vanhecke D, Zhang L, Romero P, Nahimana A, Cherix S, Duchosal MA, Ho PC, Deplancke B, Coukos G, Auwerx J, Lutolf MP, Naveiras O. The NAD-Booster Nicotinamide Riboside Potently Stimulates Hematopoiesis through Increased Mitochondrial Clearance. Cell Stem Cell 2020; 24:405-418.e7. [PMID: 30849366 DOI: 10.1016/j.stem.2019.02.012] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/18/2018] [Accepted: 02/13/2019] [Indexed: 12/22/2022]
Abstract
It has been recently shown that increased oxidative phosphorylation, as reflected by increased mitochondrial activity, together with impairment of the mitochondrial stress response, can severely compromise hematopoietic stem cell (HSC) regeneration. Here we show that the NAD+-boosting agent nicotinamide riboside (NR) reduces mitochondrial activity within HSCs through increased mitochondrial clearance, leading to increased asymmetric HSC divisions. NR dietary supplementation results in a significantly enlarged pool of progenitors, without concurrent HSC exhaustion, improves survival by 80%, and accelerates blood recovery after murine lethal irradiation and limiting-HSC transplantation. In immune-deficient mice, NR increased the production of human leucocytes from hCD34+ progenitors. Our work demonstrates for the first time a positive effect of NAD+-boosting strategies on the most primitive blood stem cells, establishing a link between HSC mitochondrial stress, mitophagy, and stem-cell fate decision, and unveiling the potential of NR to improve recovery of patients suffering from hematological failure including post chemo- and radiotherapy.
Collapse
Affiliation(s)
- Nicola Vannini
- Laboratory of Regenerative Hematopoiesis, Swiss Institute for Experimental Cancer Research (ISREC) & Institute of Bioengineering (IBI), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Epalinges 1066, Switzerland.
| | - Vasco Campos
- Laboratory of Regenerative Hematopoiesis, Swiss Institute for Experimental Cancer Research (ISREC) & Institute of Bioengineering (IBI), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Mukul Girotra
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Epalinges 1066, Switzerland; Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Vincent Trachsel
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Shanti Rojas-Sutterlin
- Laboratory of Regenerative Hematopoiesis, Swiss Institute for Experimental Cancer Research (ISREC) & Institute of Bioengineering (IBI), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Josefine Tratwal
- Laboratory of Regenerative Hematopoiesis, Swiss Institute for Experimental Cancer Research (ISREC) & Institute of Bioengineering (IBI), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Simone Ragusa
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Epalinges 1066, Switzerland
| | - Evangelos Stefanidis
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Epalinges 1066, Switzerland; Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Dongryeol Ryu
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Pernille Y Rainer
- Laboratory of System Biology and Genetics, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Gena Nikitin
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sonja Giger
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Terytty Y Li
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Aikaterini Semilietof
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Epalinges 1066, Switzerland; Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Aurelien Oggier
- Laboratory of Regenerative Hematopoiesis, Swiss Institute for Experimental Cancer Research (ISREC) & Institute of Bioengineering (IBI), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Yannick Yersin
- Laboratory of Regenerative Hematopoiesis, Swiss Institute for Experimental Cancer Research (ISREC) & Institute of Bioengineering (IBI), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Loïc Tauzin
- Flow Cytometry Platform, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Eija Pirinen
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Wan-Chen Cheng
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Epalinges 1066, Switzerland
| | - Joanna Ratajczak
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Carles Canto
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Martin Ehrbar
- Department of Obstetrics, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Federico Sizzano
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Epalinges 1066, Switzerland; Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences. Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Dominique Vanhecke
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Epalinges 1066, Switzerland
| | - Lianjun Zhang
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Epalinges 1066, Switzerland
| | - Pedro Romero
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Epalinges 1066, Switzerland
| | - Aimable Nahimana
- Service and Central Laboratory of Hematology, Departments of Oncology and of Laboratories, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Stephane Cherix
- Service d'orthopédie et de traumatologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Michel A Duchosal
- Service and Central Laboratory of Hematology, Departments of Oncology and of Laboratories, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Ping-Chih Ho
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Epalinges 1066, Switzerland
| | - Bart Deplancke
- Laboratory of System Biology and Genetics, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - George Coukos
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Epalinges 1066, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; Institute of Chemical Sciences and Engineering, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Olaia Naveiras
- Laboratory of Regenerative Hematopoiesis, Swiss Institute for Experimental Cancer Research (ISREC) & Institute of Bioengineering (IBI), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; Service and Central Laboratory of Hematology, Departments of Oncology and of Laboratories, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland.
| |
Collapse
|
13
|
Injarabian L, Devin A, Ransac S, Marteyn BS. Neutrophil Metabolic Shift during their Lifecycle: Impact on their Survival and Activation. Int J Mol Sci 2019; 21:E287. [PMID: 31906243 PMCID: PMC6981538 DOI: 10.3390/ijms21010287] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 12/14/2022] Open
Abstract
Polymorphonuclear neutrophils (PMNs) are innate immune cells, which represent 50% to 70% of the total circulating leukocytes. How PMNs adapt to various microenvironments encountered during their life cycle, from the bone marrow, to the blood plasma fraction, and to inflamed or infected tissues remains largely unexplored. Metabolic shifts have been reported in other immune cells such as macrophages or lymphocytes, in response to local changes in their microenvironment, and in association with a modulation of their pro-inflammatory or anti-inflammatory functions. The potential contribution of metabolic shifts in the modulation of neutrophil activation or survival is anticipated even though it is not yet fully described. If neutrophils are considered to be mainly glycolytic, the relative importance of alternative metabolic pathways, such as the pentose phosphate pathway, glutaminolysis, or the mitochondrial oxidative metabolism, has not been fully considered during activation. This statement may be explained by the lack of knowledge regarding the local availability of key metabolites such as glucose, glutamine, and substrates, such as oxygen from the bone marrow to inflamed tissues. As highlighted in this review, the link between specific metabolic pathways and neutrophil activation has been outlined in many reports. However, the impact of neutrophil activation on metabolic shifts' induction has not yet been explored. Beyond its importance in neutrophil survival capacity in response to available metabolites, metabolic shifts may also contribute to neutrophil population heterogeneity reported in cancer (tumor-associated neutrophil) or auto-immune diseases (Low/High Density Neutrophils). This represents an active field of research. In conclusion, the characterization of neutrophil metabolic shifts is an emerging field that may provide important knowledge on neutrophil physiology and activation modulation. The related question of microenvironmental changes occurring during inflammation, to which neutrophils will respond to, will have to be addressed to fully appreciate the importance of neutrophil metabolic shifts in inflammatory diseases.
Collapse
Affiliation(s)
- Louise Injarabian
- Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire, CNRS, Architecture et Réactivité de l’ARN, UPR9002, F-67000 Strasbourg, France;
- Université de Bordeaux, IBGC, UMR 5095, 1 rue Camille Saint Saëns, 33077 Bordeaux Cedex, France; (A.D.); (S.R.)
| | - Anne Devin
- Université de Bordeaux, IBGC, UMR 5095, 1 rue Camille Saint Saëns, 33077 Bordeaux Cedex, France; (A.D.); (S.R.)
| | - Stéphane Ransac
- Université de Bordeaux, IBGC, UMR 5095, 1 rue Camille Saint Saëns, 33077 Bordeaux Cedex, France; (A.D.); (S.R.)
| | - Benoit S. Marteyn
- Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire, CNRS, Architecture et Réactivité de l’ARN, UPR9002, F-67000 Strasbourg, France;
- Institut Pasteur, Unité de Pathogenèse des Infections Vasculaires, 75724 Paris, France
| |
Collapse
|
14
|
Nakamura-Ishizu A, Matsumura T, Stumpf PS, Umemoto T, Takizawa H, Takihara Y, O'Neil A, Majeed ABBA, MacArthur BD, Suda T. Thrombopoietin Metabolically Primes Hematopoietic Stem Cells to Megakaryocyte-Lineage Differentiation. Cell Rep 2019; 25:1772-1785.e6. [PMID: 30428347 DOI: 10.1016/j.celrep.2018.10.059] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/07/2018] [Accepted: 10/15/2018] [Indexed: 02/07/2023] Open
Abstract
During acute myelosuppression or thrombocytopenia, bone marrow (BM) hematopoietic cells respond rapidly to replenish peripheral blood platelets. While the cytokine thrombopoietin (Thpo) both regulates platelet production and maintains HSC potential, whether Thpo controls megakaryocyte (Mk)-lineage differentiation of HSCs is unclear. Here, we show that Thpo rapidly upregulates mitochondrial activity in HSCs, an activity accompanied by differentiation to an Mk lineage. Moreover, in unperturbed hematopoiesis, HSCs with high mitochondrial activity exhibit Mk-lineage differentiation in vitro and myeloid lineage-biased reconstitution in vivo. Furthermore, Thpo skewed HSCs to express the tetraspanin CD9, a pattern correlated with mitochondrial activity. Mitochondria-active HSCs are resistant to apoptosis and oxidative stress upon Thpo stimulation. Thpo-regulated mitochondrial activity associated with mitochondrial translocation of STAT3 phosphorylated at serine 727. Overall, we report an important role for Thpo in regulating rapid Mk-lineage commitment. Thpo-dependent changes in mitochondrial metabolism prime HSCs to undergo direct differentiation to an Mk lineage.
Collapse
Affiliation(s)
- Ayako Nakamura-Ishizu
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, MD6, 117599 Singapore, Singapore; International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City 860-0811, Japan.
| | - Takayoshi Matsumura
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, MD6, 117599 Singapore, Singapore
| | - Patrick S Stumpf
- Centre for Human Development Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | - Terumasa Umemoto
- International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City 860-0811, Japan
| | - Hitoshi Takizawa
- International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City 860-0811, Japan
| | - Yuji Takihara
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, MD6, 117599 Singapore, Singapore
| | - Aled O'Neil
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, MD6, 117599 Singapore, Singapore
| | | | - Ben D MacArthur
- International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City 860-0811, Japan; Mathematical Sciences, University of Southampton, Southampton SO17 1BJ, UK; Centre for Human Development Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | - Toshio Suda
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, MD6, 117599 Singapore, Singapore; International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City 860-0811, Japan.
| |
Collapse
|
15
|
Ex vivo human HSC expansion requires coordination of cellular reprogramming with mitochondrial remodeling and p53 activation. Blood Adv 2019; 2:2766-2779. [PMID: 30348672 DOI: 10.1182/bloodadvances.2018024273] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 09/06/2018] [Indexed: 01/02/2023] Open
Abstract
The limited number of hematopoietic stem cells (HSCs) in umbilical cord blood (UCB) units restricts their use for stem cell transplantation. Ex vivo treatment of UCB-CD34+ cells with valproic acid (VPA) increases the number of transplantable HSCs. In this study, we demonstrate that HSC expansion is not merely a result of proliferation of the existing stem cells but, rather, a result of a rapid reprogramming of CD34+CD90- cells into CD34+CD90+ cells, which is accompanied by limited numbers of cell divisions. Beyond this phenotypic switch, the treated cells acquire and retain a transcriptomic and mitochondrial profile, reminiscent of primary HSCs. Single and bulk RNA-seq revealed a signature highly enriched for transcripts characteristic of primary HSCs. The acquisition of this HSC signature is linked to mitochondrial remodeling accompanied by a reduced activity and enhanced glycolytic potential. These events act in concert with a modest upregulation of p53 activity to limit the levels of reactive oxygen species (ROS). Inhibition of either glycolysis or p53 activity impairs HSC expansion. This study indicates that a complex interplay of events is required for effective ex vivo expansion of UCB-HSCs.
Collapse
|
16
|
Zhang J, Ghosh J, Mohamad SF, Zhang C, Huang X, Capitano ML, Gunawan AM, Cooper S, Guo B, Cai Q, Broxmeyer HE, Srour EF. CD166 Engagement Augments Mouse and Human Hematopoietic Progenitor Function via Activation of Stemness and Cell Cycle Pathways. Stem Cells 2019; 37:1319-1330. [PMID: 31260147 DOI: 10.1002/stem.3053] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 06/21/2019] [Indexed: 12/16/2022]
Abstract
Hematopoietic stem (HSC) and progenitor (HPC) cells are regulated by interacting signals and cellular and noncellular elements of the hematopoietic niche. We previously showed that CD166 is a functional marker of murine and human HSC and of cellular components of the murine niche. Selection of murine CD166+ engrafting HSC enriched for marrow repopulating cells. Here, we demonstrate that CD166-CD166 homophilic interactions enhance generation of murine and human HPC in vitro and augment hematopoietic function of these cells. Interactions between cultured CD166+ Lineage- Sca-1+ c-Kit+ (LSK) cells and CD166+ osteoblasts (OBs) significantly enhanced the expansion of colony-forming units (CFUs). Interactions between CD166+ LSK cells and immobilized CD166 protein generated more CFU in short-term cultures than between these cells and bovine serum albumin (BSA) or in cultures initiated with CD166- LSK cells. Similar results were obtained when LSK cells from wildtype (WT) or CD166 knockout (KO) (CD166-/- ) mice were used with immobilized CD166. Human cord blood CD34+ cells expressing CD166 produced significantly higher numbers of CFUs following interaction with immobilized CD166 than their CD166- counterparts. These data demonstrate the positive effects of CD166 homophilic interactions involving CD166 on the surface of murine and human HPCs. Single-cell RNA-seq analysis of CD150+ CD48- (signaling lymphocyte activation molecule (SLAM)) LSK cells from WT and CD166-/- mice incubated with immobilized CD166 protein revealed that engagement of CD166 on these cells activates cytokine, growth factor and hormone signaling, epigenetic pathways, and other genes implicated in maintenance of stem cell pluripotency-related and mitochondria-related signaling pathways. These studies provide tangible evidence implicating CD166 engagement in the maintenance of stem/progenitor cell function. Stem Cells 2019;37:1319-1330.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, AMMS, Beijing, People's Republic of China.,Experimental Hematology and Biochemistry Lab, Beijing Institute of Radiation Medicine, AMMS, Beijing, People's Republic of China.,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, People's Republic of China
| | - Joydeep Ghosh
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Safa F Mohamad
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Chi Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Xinxin Huang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Maegan L Capitano
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Andrea M Gunawan
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Scott Cooper
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Bin Guo
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Qingchun Cai
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Edward F Srour
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
17
|
Nakamura-Ishizu A, Suda T. Multifaceted roles of thrombopoietin in hematopoietic stem cell regulation. Ann N Y Acad Sci 2019; 1466:51-58. [PMID: 31292976 DOI: 10.1111/nyas.14169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/09/2019] [Accepted: 05/24/2019] [Indexed: 12/21/2022]
Abstract
Thrombopoietin (Thpo) and its receptor myeloid proliferative leukemia (Mpl) were initially identified as the cytokine signaling that stimulates megakaryopoiesis and platelet production. However, Thpo-Mpl signaling has also been widely characterized as one of the few cytokine systems that directly regulates hematopoietic stem and progenitor cells. The ability of Thpo signaling to stimulate hematopoietic stem cell (HSC) self-renewal has led to the development and utilization of Thpo mimetic drugs to treat hematopoietic diseases with restricted function of HSCs, such as aplastic anemia. This review will cover the mechanisms by which Thpo-Mpl signaling regulates HSCs.
Collapse
Affiliation(s)
| | - Toshio Suda
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto City, Japan
| |
Collapse
|
18
|
Roy IM, Biswas A, Verfaillie C, Khurana S. Energy Producing Metabolic Pathways in Functional Regulation of the Hematopoietic Stem Cells. IUBMB Life 2019; 70:612-624. [PMID: 29999238 DOI: 10.1002/iub.1870] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 04/20/2018] [Indexed: 02/06/2023]
Abstract
The hematopoietic system has a very well-studied hierarchy with the long-term (LT) hematopoietic stem cells (HSCs) taking the top position. The pool of quiescent adult LT-HSCs generated during the fetal and early postnatal life acts as a reservoir to supply all the blood cells. Therefore, the maintenance of this stem cell pool is pivotal to maintaining homeostasis in hematopoietic system. It has long been known that external cues, along with the internal genetic factors influence the status of HSCs in the bone marrow (BM). Hypoxia is one such factor that regulates the vascular as well as hematopoietic ontogeny from a very early time point in development. The metabolic outcomes of a hypoxic microenvironment play important roles in functional regulation of HSCs, especially in case of adult BM HSCs. Anaerobic metabolic pathways therefore perform prominent role in meeting energy demands. Increased oxidative pathways on the other hand result in loss of stemness. Recent studies have attributed the functional differences in HSCs across different life stages to their metabolic phenotypes regulated by respective niches. Indicating thus, that various energy production pathways could play distinct role in regulating HSC function at different developmental/physiological states. Here, we review the current status of our understanding over the role that energy production pathways play in regulating HSC stemness. © 2018 IUBMB Life, 70(7):612-624, 2018.
Collapse
Affiliation(s)
- Irene M Roy
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Kerala, India
| | - Atreyi Biswas
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Kerala, India
| | | | - Satish Khurana
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Kerala, India
| |
Collapse
|
19
|
Chapuis N, Poulain L, Birsen R, Tamburini J, Bouscary D. Rationale for Targeting Deregulated Metabolic Pathways as a Therapeutic Strategy in Acute Myeloid Leukemia. Front Oncol 2019; 9:405. [PMID: 31192118 PMCID: PMC6540604 DOI: 10.3389/fonc.2019.00405] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 04/30/2019] [Indexed: 12/22/2022] Open
Abstract
Metabolic reprogramming is a common cancer cell phenotype as it sustains growth and proliferation. Targeting metabolic activities offers a wide range of therapeutic possibilities which are applicable to acute myeloid leukemia (AML). Indeed, in addition to the IDH1/2-mutated AML model which established the proof-of-concept for specifically targeting metabolic adaptations in AML, several recent reports have expanded the scope of such strategies in these diseases. This review highlights recent findings on metabolic deregulation in AML and summarizes their implications in leukemogenesis.
Collapse
Affiliation(s)
- Nicolas Chapuis
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Service d'Hématologie Biologique, Paris, France
| | - Laury Poulain
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France
| | - Rudy Birsen
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France
| | - Jerome Tamburini
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Service d'Hématologie Clinique, Paris, France
| | - Didier Bouscary
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Service d'Hématologie Clinique, Paris, France
| |
Collapse
|
20
|
Mitochondrial Dynamics: Biogenesis, Fission, Fusion, and Mitophagy in the Regulation of Stem Cell Behaviors. Stem Cells Int 2019; 2019:9757201. [PMID: 31089338 PMCID: PMC6476046 DOI: 10.1155/2019/9757201] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 03/05/2019] [Indexed: 12/24/2022] Open
Abstract
Stem cells have the unique capacity to differentiate into many cell types during embryonic development and postnatal growth. Through coordinated cellular behaviors (self-renewal, proliferation, and differentiation), stem cells are also pivotal to the homeostasis, repair, and regeneration of many adult tissues/organs and thus of great importance in regenerative medicine. Emerging evidence indicates that mitochondria are actively involved in the regulation of stem cell behaviors. Mitochondria undergo specific dynamics (biogenesis, fission, fusion, and mitophagy) during stem cell self-renewal, proliferation, and differentiation. The alteration of mitochondrial dynamics, fine-tuned by stem cell niche factors and stress signaling, has considerable impacts on stem cell behaviors. Here, we summarize the recent research progress on (1) how mitochondrial dynamics controls stem cell behaviors, (2) intrinsic and extrinsic factors that regulate mitochondrial dynamics, and (3) pharmacological regulators of mitochondrial dynamics and their therapeutic potential. This review emphasizes the metabolic control of stemness and differentiation and may shed light on potential new applications in stem cell-based therapy.
Collapse
|
21
|
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: 61] [Impact Index Per Article: 10.2] [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
|
22
|
Vlaski-Lafarge M, Loncaric D, Perez L, Labat V, Debeissat C, Brunet de la Grange P, Rossignol R, Ivanovic Z, Bœuf H. Bioenergetic Changes Underline Plasticity of Murine Embryonic Stem Cells. Stem Cells 2019; 37:463-475. [PMID: 30599083 DOI: 10.1002/stem.2965] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/13/2018] [Accepted: 12/03/2018] [Indexed: 12/19/2022]
Abstract
Murine embryonic stem cells (mESCs) are endowed by a time-dependent window of plasticity during their early commitment steps. Indeed, while mESCs deprived of leukemia inhibitory factor (LIF) for 24 hours revert to their naive pluripotent state after subsequent LIF readdition, cells deprived of LIF for 48 hours are no longer efficient in reverting, upon LIF addition, and undergo irreversible differentiation. We investigated undisclosed bioenergetic profiles of early mESC-derived committed cells versus their undifferentiated states in order to reveal specific bioenergetic changes associated with mESC plasticity. Multiparametric bioenergetic analysis revealed that pluripotent (+LIF) and reversibly committed cells (-LIF24h) are energetically flexible, depending on both oxidative phosphorylation (OXPHOS) and glycolysis. They exhibit high mitochondrial respiration in the presence of the main energetic substrates and can also rely on glycolysis in the presence of OXPHOS inhibitor. Inhibition of the glycolysis or mitochondrial respiration does not change drastically the expression of pluripotency genes, which remain well expressed. In addition, cells treated with these inhibitors keep their capacity to differentiate efficiently upon embryoid bodies formation. Transition from metabolically active mESCs to irreversibly committed cells is associated with a clear change in mitochondrial network morphology, to an increase of adenosine triphosphate (ATP) produced from glycolysis and a decline of ATP turnover and of the mitochondrial activity without change in the mitochondrial mass. Our study pointed that plasticity window of mESCs is associated with the bivalent energetic metabolism and potency to shift to glycolysis or OXPHOS on demand. LIF removal provokes glycolytic metabolic orientation and consecutive loss of the LIF-dependent reversion of cells to the pluripotent state. Stem Cells 2019;37:463-475.
Collapse
Affiliation(s)
- Marija Vlaski-Lafarge
- R&D Department, Etablissement Français du Sang Nouvelle-Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux
| | - Darija Loncaric
- R&D Department, Etablissement Français du Sang Nouvelle-Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux
| | - Laura Perez
- R&D Department, Etablissement Français du Sang Nouvelle-Aquitaine, Bordeaux, France
| | - Véronique Labat
- R&D Department, Etablissement Français du Sang Nouvelle-Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux
| | - Christelle Debeissat
- R&D Department, Etablissement Français du Sang Nouvelle-Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux
| | - Philippe Brunet de la Grange
- R&D Department, Etablissement Français du Sang Nouvelle-Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux
| | | | - Zoran Ivanovic
- R&D Department, Etablissement Français du Sang Nouvelle-Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux
| | - Hélène Bœuf
- Inserm/U1026, University of Bordeaux, Bordeaux, France
| |
Collapse
|
23
|
Metabolic Regulations in Hematopoietic Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1143:59-74. [PMID: 31338815 DOI: 10.1007/978-981-13-7342-8_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
One of the bottlenecks of the treatments for malignant hematopoietic disorders is the unavailability of sufficient amount of hematopoietic stem cells (HSCs). HSCs are considered to be originated from the aorta-gonad-mesonephros and gradually migrates into fetal liver and resides in a unique microenvironment/niche of bone marrow. Although many intrinsic and extrinsic factors (niche components) are reported to be involved in the origination, maturation, migration, and localization of HSCs at different developmental stages, the detailed molecular mechanisms still remain largely unknown. Previous studies have shown that intrinsic metabolic networks may be critical for the cell fate determinations of HSCs. For example, HSCs mainly utilize glycolysis as the main energy sources; oxidative phosphorylation is required for the homeostasis of HSCs; lipid or amino acid metabolisms may also sustain HSC stemness. Mechanistically, lots of regulatory pathways, such as MEIS1/HIF1A and PI3K/AKT/mTOR signaling, are found to fine-tune the different nutrient metabolisms and cell fate commitments of HSCs. However, more efforts are required for the optimization and establishment of precise metabolic techniques specific for the HSCs with relatively rare cell frequency and understanding of the basic metabolic properties and their underlying regulatory mechanisms of different nutrients (such as glucose) during the different developmental stages of HSCs.
Collapse
|
24
|
Jin G, Xu C, Zhang X, Long J, Rezaeian AH, Liu C, Furth ME, Kridel S, Pasche B, Bian XW, Lin HK. Atad3a suppresses Pink1-dependent mitophagy to maintain homeostasis of hematopoietic progenitor cells. Nat Immunol 2017; 19:29-40. [PMID: 29242539 DOI: 10.1038/s41590-017-0002-1] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 10/04/2017] [Indexed: 01/13/2023]
Abstract
Although deletion of certain autophagy-related genes has been associated with defects in hematopoiesis, it remains unclear whether hyperactivated mitophagy affects the maintenance and differentiation of hematopoietic stem cells (HSCs) and committed progenitor cells. Here we report that targeted deletion of the gene encoding the AAA+-ATPase Atad3a hyperactivated mitophagy in mouse hematopoietic cells. Affected mice showed reduced survival, severely decreased bone-marrow cellularity, erythroid anemia and B cell lymphopenia. Those phenotypes were associated with skewed differentiation of stem and progenitor cells and an enlarged HSC pool. Mechanistically, Atad3a interacted with the mitochondrial channel components Tom40 and Tim23 and served as a bridging factor to facilitate appropriate transportation and processing of the mitophagy protein Pink1. Loss of Atad3a caused accumulation of Pink1 and activated mitophagy. Notably, deletion of Pink1 in Atad3a-deficient mice significantly 'rescued' the mitophagy defect, which resulted in restoration of the progenitor and HSC pools. Our data indicate that Atad3a suppresses Pink1-dependent mitophagy and thereby serves a key role in hematopoietic homeostasis.
Collapse
Affiliation(s)
- Guoxiang Jin
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chuan Xu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Oncology, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Xian Zhang
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jie Long
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Pathology School of Basic Medical Science, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Abdol Hossein Rezaeian
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chunfang Liu
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark E Furth
- Wake Forest Innovations, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Steven Kridel
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Boris Pasche
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA. .,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan. .,Department of Biotechnology, Asia University, Taichung, Taiwan.
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Hematopoietic stem cells (HSCs) possess two fundamental characteristics, the capacity for self-renewal and the sustained production of all blood cell lineages. The fine balance between HSC expansion and lineage specification is dynamically regulated by the interplay between external and internal stimuli. This review introduces recent advances in the roles played by the stem cell niche, regulatory transcriptional networks, and metabolic pathways in governing HSC self-renewal, commitment, and lineage differentiation. We will further focus on discoveries made by studying hematopoiesis at single-cell resolution. RECENT FINDINGS HSCs require the support of an interactive milieu with their physical position within the perivascular niche dynamically regulating HSC behavior. In these microenvironments, transcription factor networks and nutrient-mediated regulation of energy resources, signaling pathways, and epigenetic status govern HSC quiescence and differentiation. Once HSCs begin their lineage specification, single-cell analyses show that they do not become oligopotent but rather, differentiate directly into committed unipotent progenitors. SUMMARY The diversity of transcriptional networks and metabolic pathways in HSCs and their downstream progeny allows a high level of plasticity in blood differentiation. The intricate interactions between these pathways, within the perivascular niche, broaden the specification of HSCs in pathological and stressed conditions.
Collapse
|
26
|
Nguyen-McCarty M, Klein PS. Autophagy is a signature of a signaling network that maintains hematopoietic stem cells. PLoS One 2017; 12:e0177054. [PMID: 28486555 PMCID: PMC5423627 DOI: 10.1371/journal.pone.0177054] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/23/2017] [Indexed: 12/22/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are able to self-renew and to differentiate into all blood cells. HSCs reside in a low-perfusion niche and depend on local signals to survive and to maintain the capacity for self-renewal. HSCs removed from the niche are unable to survive without addition of hematopoietic cytokines and rapidly lose their ability to self-renew. We reported previously that inhibition of both GSK-3 and mTORC1 is essential to maintain long-term HSCs ex vivo. Although Wnt/β-catenin signaling downstream of GSK-3 is required for this response, the downstream effectors of mTORC1 remain undefined. We now report that HSCs express a pro-autophagic gene signature and accumulate LC3 puncta only when both mTORC1 and GSK-3 are inhibited, identifying autophagy as a signature for a signaling network that maintains HSCs ex vivo. In addition, these conditions sustain HSC repopulating function despite an increased rate of global translation. Together, these findings provide new insight into the relative contributions of various mTORC1 outputs toward the maintenance of HSC function and build upon the growing body of literature implicating autophagy and tightly controlled protein synthesis as important modulators of diverse stem cell populations.
Collapse
Affiliation(s)
- Michelle Nguyen-McCarty
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Peter S. Klein
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Medicine (Hematology/Oncology), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
27
|
Metabolic regulation of hematopoietic stem cell commitment and erythroid differentiation. Curr Opin Hematol 2016; 23:198-205. [PMID: 26871253 DOI: 10.1097/moh.0000000000000234] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Hematopoietic stem cell (HSC) renewal and lineage differentiation are finely tuned processes, regulated by cytokines, transcription factors and cell-cell contacts. However, recent studies have shown that fuel utilization also conditions HSC fate. This review focuses on our current understanding of the metabolic pathways that govern HSC self-renewal, commitment and specification to the erythroid lineage. RECENT FINDINGS HSCs reside in a hypoxic bone marrow niche that favors anaerobic glycolysis. Although this metabolic pathway is required for stem cell maintenance, other pathways also play critical roles. Fatty acid oxidation preserves HSC self-renewal by promoting asymmetric division, whereas oxidative phosphorylation induces lineage commitment. Committed erythroid progenitors support the production of 2.4 million erythrocytes per second in human adults via a synchronized regulation of iron, amino acid and glucose metabolism. Iron is indispensable for heme biosynthesis in erythroblasts; a process finely coordinated by at least two hormones, hepcidin and erythroferrone, together with multiple cell surface iron transporters. Furthermore, hemoglobin production is promoted by amino acid-induced mTOR signaling. Erythropoiesis is also strictly dependent on glutamine metabolism; under conditions where glutaminolysis is inhibited, erythropoietin-signaled progenitors are diverted to a myelomonocytic fate. Indeed, the utilization of both glutamine and glucose in de-novo nucleotide biosynthesis is a sine qua non for erythroid differentiation. SUMMARY Diverse metabolic networks function in concert with transcriptional, translational and epigenetic programs to regulate HSC potential and orient physiological as well as pathological erythroid differentiation.
Collapse
|
28
|
Sommerkamp P, Trumpp A. Metabolic cues for hematopoietic stem cells. Science 2016; 354:1103-1104. [PMID: 27934722 DOI: 10.1126/science.aal3466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Pia Sommerkamp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany. .,Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
29
|
Zhang Y, Dépond M, He L, Foudi A, Kwarteng EO, Lauret E, Plo I, Desterke C, Dessen P, Fujii N, Opolon P, Herault O, Solary E, Vainchenker W, Joulin V, Louache F, Wittner M. CXCR4/CXCL12 axis counteracts hematopoietic stem cell exhaustion through selective protection against oxidative stress. Sci Rep 2016; 6:37827. [PMID: 27886253 PMCID: PMC5122894 DOI: 10.1038/srep37827] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 11/02/2016] [Indexed: 01/17/2023] Open
Abstract
Hematopoietic stem cells (HSCs) undergo self-renewal to maintain hematopoietic homeostasis for lifetime, which is regulated by the bone marrow (BM) microenvironment. The chemokine receptor CXCR4 and its ligand CXCL12 are critical factors supporting quiescence and BM retention of HSCs. Here, we report an unknown function of CXCR4/CXCL12 axis in the protection of HSCs against oxidative stress. Disruption of CXCR4 receptor in mice leads to increased endogenous production of reactive oxygen species (ROS), resulting in p38 MAPK activation, increased DNA double-strand breaks and apoptosis leading to marked reduction in HSC repopulating potential. Increased ROS levels are directly responsible for exhaustion of the HSC pool and are not linked to loss of quiescence of CXCR4-deficient HSCs. Furthermore, we report that CXCL12 has a direct rescue effect on oxidative stress-induced HSC damage at the mitochondrial level. These data highlight the importance of CXCR4/CXCL12 axis in the regulation of lifespan of HSCs by limiting ROS generation and genotoxic stress.
Collapse
Affiliation(s)
- Yanyan Zhang
- Paris-Saclay University, UMRS-1170, Gustave Roussy, Villejuif, France
| | - Mallorie Dépond
- Paris-Saclay University, UMRS-1170, Gustave Roussy, Villejuif, France
| | - Liang He
- Paris-Saclay University, UMRS-1170, Gustave Roussy, Villejuif, France
| | - Adlen Foudi
- Paris-Saclay University, INSERM U935, Andre Lwoff Institute, Paul Brousse Hospital, Villejuif, France
| | | | - Evelyne Lauret
- Paris Descartes University, CNRS (UMR 8104), Inserm U1016, Institut Cochin, Paris, France
| | - Isabelle Plo
- Paris-Saclay University, UMRS-1170, Gustave Roussy, Villejuif, France
| | - Christophe Desterke
- Paris-Saclay University, UFR Medicine, INSERM UMS 33, Andre Lwoff Institute, Paul Brousse Hospital, Villejuif, France
| | - Philippe Dessen
- Bioinformatic platform, UMS AMMICA, INSERM US23/CNRS UMS3665, Gustave Roussy, Villejuif, France
| | - Nobutaka Fujii
- Kyoto University, Graduate School of Pharmaceutical Sciences, Kyoto, Japan
| | - Paule Opolon
- Laboratoire de pathologie expérimentale, Gustave Roussy, Villejuif, France
| | - Olivier Herault
- CNRS UMR 7292 GICC, Tours, France.,CNRS GDR 3697 MicroNiT, France
| | - Eric Solary
- Paris-Saclay University, UMRS-1170, Gustave Roussy, Villejuif, France
| | | | - Virginie Joulin
- Paris-Saclay University, UMRS-1170, Gustave Roussy, Villejuif, France
| | - Fawzia Louache
- Paris-Saclay University, UMRS-1170, Gustave Roussy, Villejuif, France.,CNRS GDR 3697 MicroNiT, France
| | - Monika Wittner
- Paris-Saclay University, UMRS-1170, Gustave Roussy, Villejuif, France.,CNRS GDR 3697 MicroNiT, France
| |
Collapse
|
30
|
Jalnapurkar S, Singh S, Devi MR, Limaye L, Kale V. Nitric oxide has contrasting age-dependent effects on the functionality of murine hematopoietic stem cells. Stem Cell Res Ther 2016; 7:171. [PMID: 27876094 PMCID: PMC5120451 DOI: 10.1186/s13287-016-0433-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 10/26/2016] [Accepted: 10/29/2016] [Indexed: 01/06/2023] Open
Abstract
Background The success of hematopoietic stem cell (HSC) transplantation is dependent on the quality of the donor HSCs. Some sources of HSCs display reduced engraftment efficiency either because of inadequate number (e.g., fetal liver and cord blood), or age-related dysfunction (e.g. in older individuals). Therefore, use of pharmacological compounds to improve functionality of HSCs is a forefront research area in hematology. Methods Lineage negative (Lin−) cells isolated from murine bone marrow or sort-purified Lin−Sca-1+c-Kit+CD34− (LSK-CD34−) were treated with a nitric oxide donor, sodium nitroprusside (SNP). The cells were subjected to various phenotypic and functional assays. Results We found that SNP treatment of Lin− cells leads to an increase in the numbers of LSK-CD34+ cells in them. Using sort-purified LSK CD34− HSCs, we show that this is related to acquisition of CD34 expression by LSK-CD34− cells, rather than proliferation of LSK-CD34+ cells. Most importantly, this upregulated expression of CD34 had age-dependent contrasting effects on HSC functionality. Increased CD34 expression significantly improved the engraftment of juvenile HSCs (6–8 weeks); in sharp contrast, it reduced the engraftment of adult HSCs (10–12 weeks). The molecular mechanism behind this phenomenon involved nitric oxide (NO)-mediated differential induction of various transcription factors involved in commitment with regard to self-renewal in adult and juvenile HSCs, respectively. Preliminary experiments performed on cord blood-derived and mobilized peripheral blood-derived cells revealed that NO exerts age-dependent contrasting effects on human HSCs as well. Conclusions This study demonstrates novel age-dependent contrasting effects of NO on HSC functionality and suggests that HSC age may be an important parameter in screening of various compounds for their use in manipulation of HSCs. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0433-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sapana Jalnapurkar
- Stem Cell Lab, National Centre for Cell Science, Ganeshkhind, Pune, 411007, India
| | - Shweta Singh
- Stem Cell Lab, National Centre for Cell Science, Ganeshkhind, Pune, 411007, India
| | | | - Lalita Limaye
- Stem Cell Lab, National Centre for Cell Science, Ganeshkhind, Pune, 411007, India
| | - Vaijayanti Kale
- Stem Cell Lab, National Centre for Cell Science, Ganeshkhind, Pune, 411007, India.
| |
Collapse
|
31
|
Specification of haematopoietic stem cell fate via modulation of mitochondrial activity. Nat Commun 2016; 7:13125. [PMID: 27731316 PMCID: PMC5064016 DOI: 10.1038/ncomms13125] [Citation(s) in RCA: 194] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 09/05/2016] [Indexed: 02/06/2023] Open
Abstract
Haematopoietic stem cells (HSCs) differ from their committed progeny by relying primarily on anaerobic glycolysis rather than mitochondrial oxidative phosphorylation for energy production. However, whether this change in the metabolic program is the cause or the consequence of the unique function of HSCs remains unknown. Here we show that enforced modulation of energy metabolism impacts HSC self-renewal. Lowering the mitochondrial activity of HSCs by chemically uncoupling the electron transport chain drives self-renewal under culture conditions that normally induce rapid differentiation. We demonstrate that this metabolic specification of HSC fate occurs through the reversible decrease of mitochondrial mass by autophagy. Our data thus reveal a causal relationship between mitochondrial metabolism and fate choice of HSCs and also provide a valuable tool to expand HSCs outside of their native bone marrow niches.
Collapse
|
32
|
SDF-1/CXCL12 modulates mitochondrial respiration of immature blood cells in a bi-phasic manner. Blood Cells Mol Dis 2016; 58:13-8. [PMID: 27067482 DOI: 10.1016/j.bcmd.2016.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 01/26/2016] [Indexed: 01/08/2023]
Abstract
SDF-1/CXCL12 is a potent chemokine required for the homing and engraftment of hematopoietic stem and progenitor cells. Previous data from our group has shown that in an SDF-1/CXCL12 transgenic mouse model, lineage(-) Sca-1(+) c-Kit(+) (LSK) bone marrow cells have reduced mitochondrial membrane potential versus wild-type. These results suggested that SDF-1/CXCL12 may function to keep mitochondrial respiration low in immature blood cells in the bone marrow. Low mitochondrial metabolism helps to maintain low levels of reactive oxygen species (ROS), which can influence differentiation. To test whether SDF-1/CXCL12 regulates mitochondrial metabolism, we employed the human leukemia cell line HL-60, that expresses high levels of the SDF-1/CXCL12 receptor, CXCR4, as a model of hematopoietic progenitor cells in vitro. We treated HL-60 cells with SDF-1/CXCL12 for 2 and 24h. Oxygen consumption rates (OCR), mitochondrial-associated ATP production, mitochondrial mass, and mitochondrial membrane potential of HL-60 cells were significantly reduced at 2h and increased at 24h as compared to untreated control cells. These biphasic effects of SDF-1/CXCL12 were reproduced with lineage negative primary mouse bone marrow cells, suggesting a novel function of SDF-1/CXCL12 in modulating mitochondrial respiration by regulating mitochondrial oxidative phosphorylation, ATP production and mitochondrial content.
Collapse
|
33
|
Wanet A, Arnould T, Najimi M, Renard P. Connecting Mitochondria, Metabolism, and Stem Cell Fate. Stem Cells Dev 2015; 24:1957-71. [PMID: 26134242 PMCID: PMC4543487 DOI: 10.1089/scd.2015.0117] [Citation(s) in RCA: 236] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
As sites of cellular respiration and energy production, mitochondria play a central role in cell metabolism. Cell differentiation is associated with an increase in mitochondrial content and activity and with a metabolic shift toward increased oxidative phosphorylation activity. The opposite occurs during reprogramming of somatic cells into induced pluripotent stem cells. Studies have provided evidence of mitochondrial and metabolic changes during the differentiation of both embryonic and somatic (or adult) stem cells (SSCs), such as hematopoietic stem cells, mesenchymal stem cells, and tissue-specific progenitor cells. We thus propose to consider those mitochondrial and metabolic changes as hallmarks of differentiation processes. We review how mitochondrial biogenesis, dynamics, and function are directly involved in embryonic and SSC differentiation and how metabolic and sensing pathways connect mitochondria and metabolism with cell fate and pluripotency. Understanding the basis of the crosstalk between mitochondria and cell fate is of critical importance, given the promising application of stem cells in regenerative medicine. In addition to the development of novel strategies to improve the in vitro lineage-directed differentiation of stem cells, understanding the molecular basis of this interplay could lead to the identification of novel targets to improve the treatment of degenerative diseases.
Collapse
Affiliation(s)
- Anaïs Wanet
- 1 Laboratory of Biochemistry and Cell Biology (URBC), NAmur Research Institute for LIfe Sciences (NARILIS), University of Namur (UNamur) , Namur, Belgium
| | - Thierry Arnould
- 1 Laboratory of Biochemistry and Cell Biology (URBC), NAmur Research Institute for LIfe Sciences (NARILIS), University of Namur (UNamur) , Namur, Belgium
| | - Mustapha Najimi
- 2 Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Clinique et Expérimentale (IREC), Université Catholique de Louvain , Brussels, Belgium
| | - Patricia Renard
- 1 Laboratory of Biochemistry and Cell Biology (URBC), NAmur Research Institute for LIfe Sciences (NARILIS), University of Namur (UNamur) , Namur, Belgium
| |
Collapse
|
34
|
Mantel CR, O'Leary HA, Chitteti BR, Huang X, Cooper S, Hangoc G, Brustovetsky N, Srour EF, Lee MR, Messina-Graham S, Haas DM, Falah N, Kapur R, Pelus LM, Bardeesy N, Fitamant J, Ivan M, Kim KS, Broxmeyer HE. Enhancing Hematopoietic Stem Cell Transplantation Efficacy by Mitigating Oxygen Shock. Cell 2015; 161:1553-65. [PMID: 26073944 PMCID: PMC4480616 DOI: 10.1016/j.cell.2015.04.054] [Citation(s) in RCA: 245] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 02/20/2015] [Accepted: 04/08/2015] [Indexed: 12/13/2022]
Abstract
Hematopoietic stem cells (HSCs) reside in hypoxic niches within bone marrow and cord blood. Yet, essentially all HSC studies have been performed with cells isolated and processed in non-physiologic ambient air. By collecting and manipulating bone marrow and cord blood in native conditions of hypoxia, we demonstrate that brief exposure to ambient oxygen decreases recovery of long-term repopulating HSCs and increases progenitor cells, a phenomenon we term extraphysiologic oxygen shock/stress (EPHOSS). Thus, true numbers of HSCs in the bone marrow and cord blood are routinely underestimated. We linked ROS production and induction of the mitochondrial permeability transition pore (MPTP) via cyclophilin D and p53 as mechanisms of EPHOSS. The MPTP inhibitor cyclosporin A protects mouse bone marrow and human cord blood HSCs from EPHOSS during collection in air, resulting in increased recovery of transplantable HSCs. Mitigating EPHOSS during cell collection and processing by pharmacological means may be clinically advantageous for transplantation.
Collapse
Affiliation(s)
- Charlie R Mantel
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Heather A O'Leary
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brahmananda R Chitteti
- Department of Medicine (Hematology/Oncology), Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - XinXin Huang
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Scott Cooper
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Giao Hangoc
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nickolay Brustovetsky
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Edward F Srour
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Medicine (Hematology/Oncology), Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Man Ryul Lee
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Soonchunhyang Institute of Medi-bio Science, Chungcheongnam-do 336-745, Korea
| | - Steven Messina-Graham
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - David M Haas
- Division of Clinical Pharmacology, Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nadia Falah
- Division of Clinical Pharmacology, Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Reuben Kapur
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biochemistry/Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Louis M Pelus
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nabeel Bardeesy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | - Julien Fitamant
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | - Mircea Ivan
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Medicine (Hematology/Oncology), Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 133-791, Korea
| | - Hal E Broxmeyer
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
35
|
Zöller M. CD44, Hyaluronan, the Hematopoietic Stem Cell, and Leukemia-Initiating Cells. Front Immunol 2015; 6:235. [PMID: 26074915 PMCID: PMC4443741 DOI: 10.3389/fimmu.2015.00235] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 04/30/2015] [Indexed: 12/14/2022] Open
Abstract
CD44 is an adhesion molecule that varies in size due to glycosylation and insertion of so-called variant exon products. The CD44 standard isoform (CD44s) is highly expressed in many cells and most abundantly in cells of the hematopoietic system, whereas expression of CD44 variant isoforms (CD44v) is more restricted. CD44s and CD44v are known as stem cell markers, first described for hematopoietic stem cells and later on confirmed for cancer- and leukemia-initiating cells. Importantly, both abundantly expressed CD44s as well as CD44v actively contribute to the maintenance of stem cell features, like generating and embedding in a niche, homing into the niche, maintenance of quiescence, and relative apoptosis resistance. This is surprising, as CD44 is not a master stem cell gene. I here will discuss that the functional contribution of CD44 relies on its particular communication skills with neighboring molecules, adjacent cells and, last not least, the surrounding matrix. In fact, it is the interaction of the hyaluronan receptor CD44 with its prime ligand, which strongly assists stem cells to fulfill their special and demanding tasks. Recent fundamental progress in support of this “old” hypothesis, which may soon pave the way for most promising new therapeutics, is presented for both hematopoietic stem cell and leukemia-initiating cell. The contribution of CD44 to the generation of a stem cell niche, to homing of stem cells in their niche, to stem cell quiescence and apoptosis resistance will be in focus.
Collapse
Affiliation(s)
- Margot Zöller
- Department of Tumor Cell Biology, University Hospital of Surgery , Heidelberg , Germany
| |
Collapse
|
36
|
|
37
|
Katajisto P, Döhla J, Chaffer CL, Pentinmikko N, Marjanovic N, Iqbal S, Zoncu R, Chen W, Weinberg RA, Sabatini DM. Stem cells. Asymmetric apportioning of aged mitochondria between daughter cells is required for stemness. Science 2015; 348:340-3. [PMID: 25837514 DOI: 10.1126/science.1260384] [Citation(s) in RCA: 385] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 03/12/2015] [Indexed: 12/19/2022]
Abstract
By dividing asymmetrically, stem cells can generate two daughter cells with distinct fates. However, evidence is limited in mammalian systems for the selective apportioning of subcellular contents between daughters. We followed the fates of old and young organelles during the division of human mammary stemlike cells and found that such cells apportion aged mitochondria asymmetrically between daughter cells. Daughter cells that received fewer old mitochondria maintained stem cell traits. Inhibition of mitochondrial fission disrupted both the age-dependent subcellular localization and segregation of mitochondria and caused loss of stem cell properties in the progeny cells. Hence, mechanisms exist for mammalian stemlike cells to asymmetrically sort aged and young mitochondria, and these are important for maintaining stemness properties.
Collapse
Affiliation(s)
- Pekka Katajisto
- Whitehead Institute for Biomedical Research, Boston, MA 02142, USA. Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA. Howard Hughes Medical Institute, MIT, Cambridge, MA 02139, USA. Institute of Biotechnology, University of Helsinki, P.O. Box 00014, Helsinki, Finland.
| | - Julia Döhla
- Institute of Biotechnology, University of Helsinki, P.O. Box 00014, Helsinki, Finland
| | | | - Nalle Pentinmikko
- Institute of Biotechnology, University of Helsinki, P.O. Box 00014, Helsinki, Finland
| | - Nemanja Marjanovic
- Whitehead Institute for Biomedical Research, Boston, MA 02142, USA. Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - Sharif Iqbal
- Institute of Biotechnology, University of Helsinki, P.O. Box 00014, Helsinki, Finland
| | - Roberto Zoncu
- Whitehead Institute for Biomedical Research, Boston, MA 02142, USA. Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA. Howard Hughes Medical Institute, MIT, Cambridge, MA 02139, USA
| | - Walter Chen
- Whitehead Institute for Biomedical Research, Boston, MA 02142, USA. Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA. Howard Hughes Medical Institute, MIT, Cambridge, MA 02139, USA
| | - Robert A Weinberg
- Whitehead Institute for Biomedical Research, Boston, MA 02142, USA. Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - David M Sabatini
- Whitehead Institute for Biomedical Research, Boston, MA 02142, USA. Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA. Howard Hughes Medical Institute, MIT, Cambridge, MA 02139, USA. Broad Institute, Cambridge, MA 02142, USA. The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
38
|
Mitochondrial function in murine skin epithelium is crucial for hair follicle morphogenesis and epithelial-mesenchymal interactions. J Invest Dermatol 2014; 135:679-689. [PMID: 25371971 DOI: 10.1038/jid.2014.475] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 07/31/2014] [Accepted: 08/12/2014] [Indexed: 12/21/2022]
Abstract
Here, we studied how epithelial energy metabolism impacts overall skin development by selectively deleting intraepithelial mtDNA in mice by ablating a key maintenance factor (Tfam(EKO)), which induces loss of function of the electron transport chain (ETC). Quantitative (immuno)histomorphometry demonstrated that Tfam(EKO) mice showed significantly reduced hair follicle (HF) density and morphogenesis, fewer intrafollicular keratin15+ epithelial progenitor cells, increased apoptosis, and reduced proliferation. Tfam(EKO) mice also displayed premature entry into (aborted) HF cycling by apoptosis-driven HF regression (catagen). Ultrastructurally, Tfam(EKO) mice exhibited severe HF dystrophy, pigmentary abnormalities, and telogen-like condensed dermal papillae. Epithelial HF progenitor cell differentiation (Plet1, Lrig1 Lef1, and β-catenin), sebaceous gland development (adipophilin, Scd1, and oil red), and key mediators/markers of epithelial-mesenchymal interactions during skin morphogenesis (NCAM, versican, and alkaline phosphatase) were all severely altered in Tfam(EKO) mice. Moreover, the number of mast cells, major histocompatibility complex class II+, or CD11b+ immunocytes in the skin mesenchyme was increased, and essentially no subcutis developed. Therefore, in contrast to their epidermal counterparts, pilosebaceous unit stem cells depend on a functional ETC. Most importantly, our findings point toward a frontier in skin biology: the coupling of HF keratinocyte mitochondrial function with the epithelial-mesenchymal interactions that drive overall development of the skin and its appendages.
Collapse
|
39
|
Tang AH, Rando TA. Induction of autophagy supports the bioenergetic demands of quiescent muscle stem cell activation. EMBO J 2014; 33:2782-97. [PMID: 25316028 DOI: 10.15252/embj.201488278] [Citation(s) in RCA: 222] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The exit of a stem cell out of quiescence into an activated state is characterized by major metabolic changes associated with increased biosynthesis of proteins and macromolecules. The regulation of this transition is poorly understood. Using muscle stem cells, or satellite cells (SCs), we found that autophagy, which catabolizes intracellular contents to maintain proteostasis and to produce energy during nutrient deprivation, was induced during SC activation. Inhibition of autophagy suppressed the increase in ATP levels and delayed SC activation, both of which could be partially rescued by exogenous pyruvate as an energy source, suggesting that autophagy may provide nutrients necessary to meet bioenergetic demands during this critical transition from quiescence to activation. We found that SIRT1, a known nutrient sensor, regulates autophagic flux in SC progeny. A deficiency of SIRT1 led to a delay in SC activation that could also be partially rescued by exogenous pyruvate. These studies suggest that autophagy, regulated by SIRT1, may play an important role during SC activation to meet the high bioenergetic demands of the activation process.
Collapse
Affiliation(s)
- Ann H Tang
- Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas A Rando
- Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA Neurology Service and Rehabilitation Research and Developmental Center of Excellence, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| |
Collapse
|
40
|
Abstract
SIGNIFICANCE The effect of redox signaling on hematopoietic stem cell (HSC) function is not clearly understood. RECENT ADVANCES A growing body of evidence suggests that adult HSCs reside in the hypoxic bone marrow microenvironment or niche during homeostasis. It was recently shown that primitive HSCs in the bone marrow prefer to utilize anaerobic glycolysis to meet their energy demands and have lower rates of oxygen consumption and lower ATP levels. Hypoxia-inducible factor-α (Hif-1α) is a master regulator of cellular metabolism. With hundreds of downstream target genes and crosstalk with other signaling pathways, it regulates various aspects of metabolism from the oxidative stress response to glycolysis and mitochondrial respiration. Hif-1α is highly expressed in HSCs, where it regulates their function and metabolic phenotype. However, the regulation of Hif-1α in HSCs is not entirely understood. The homeobox transcription factor myeloid ecotropic viral integration site 1 (Meis1) is expressed in the most primitive HSCs populations, and it is required for primitive hematopoiesis. Recent reports suggest that Meis1 is required for normal adult HSC function by regulating the metabolism and redox state of HSCs transcriptionally through Hif-1α and Hif-2α. CRITICAL ISSUES Given the profound effect of redox status on HSC function, it is critical to fully characterize the intrinsic, and microenvironment-related mechanisms of metabolic and redox regulation in HSCs. FUTURE DIRECTIONS Future studies will be needed to elucidate the link between HSC metabolism and HSC fates, including quiescence, self-renewal, differentiation, apoptosis, and migration.
Collapse
Affiliation(s)
- Cheng Cheng Zhang
- Division of Cardiology, Departments of Physiology and Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hesham A. Sadek
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
41
|
Strandkvist C, Juul J, Bendtsen KM. Asymmetric segregation of damaged cellular components in spatially structured multicellular organisms. PLoS One 2014; 9:e87917. [PMID: 24551071 PMCID: PMC3923766 DOI: 10.1371/journal.pone.0087917] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 01/01/2014] [Indexed: 11/18/2022] Open
Abstract
The asymmetric distribution of damaged cellular components has been observed in species ranging from fission yeast to humans. To study the potential advantages of damage segregation, we have developed a mathematical model describing ageing mammalian tissue, that is, a multicellular system of somatic cells that do not rejuvenate at cell division. To illustrate the applicability of the model, we specifically consider damage incurred by mutations to mitochondrial DNA, which are thought to be implicated in the mammalian ageing process. We show analytically that the asymmetric distribution of damaged cellular components reduces the overall damage level and increases the longevity of the cell population. Motivated by the experimental reports of damage segregation in human embryonic stem cells, dividing symmetrically with respect to cell-fate, we extend the model to consider spatially structured systems of cells. Imposing spatial structure reduces, but does not eliminate, the advantage of asymmetric division over symmetric division. The results suggest that damage partitioning could be a common strategy for reducing the accumulation of damage in a wider range of cell types than previously thought.
Collapse
Affiliation(s)
| | - Jeppe Juul
- University of Copenhagen, Niels Bohr Institute, Copenhagen, Denmark
| | | |
Collapse
|
42
|
Piccoli C, Agriesti F, Scrima R, Falzetti F, Di Ianni M, Capitanio N. To breathe or not to breathe: the haematopoietic stem/progenitor cells dilemma. Br J Pharmacol 2014; 169:1652-71. [PMID: 23714011 DOI: 10.1111/bph.12253] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Revised: 05/11/2013] [Accepted: 05/16/2013] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Adult haematopoietic stem/progenitor cells (HSPCs) constitute the lifespan reserve for the generation of all the cellular lineages in the blood. Although massive progress in identifying the cluster of master genes controlling self-renewal and multipotency has been achieved in the past decade, some aspects of the physiology of HSPCs still need to be clarified. In particular, there is growing interest in the metabolic profile of HSPCs in view of their emerging role as determinants of cell fate. Indeed, stem cells and progenitors have distinct metabolic profiles, and the transition from stem to progenitor cell corresponds to a critical metabolic change, from glycolysis to oxidative phosphorylation. In this review, we summarize evidence, reported in the literature and provided by our group, highlighting the peculiar ability of HSPCs to adapt their mitochondrial oxidative/bioenergetic metabolism to survive in the hypoxic microenvironment of the endoblastic niche and to exploit redox signalling in controlling the balance between quiescence versus active cycling and differentiation. Especial prominence is given to the interplay between hypoxia inducible factor-1, globins and NADPH oxidases in managing the mitochondrial dioxygen-related metabolism and biogenesis in HSPCs under different ambient conditions. A mechanistic model is proposed whereby 'mitochondrial differentiation' is a prerequisite in uncommitted stem cells, paving the way for growth/differentiation factor-dependent processes. Advancing the understanding of stem cell metabolism will, hopefully, help to (i) improve efforts to maintain, expand and manipulate HSPCs ex vivo and realize their potential therapeutic benefits in regenerative medicine; (ii) reprogramme somatic cells to generate stem cells; and (iii) eliminate, selectively, malignant stem cells. LINKED ARTICLES This article is part of a themed section on Emerging Therapeutic Aspects in Oncology. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.169.issue-8.
Collapse
Affiliation(s)
- C Piccoli
- Department of Medical and Experimental Medicine, University of Foggia, Foggia, Italy.
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW Hematopoietic stem cells (HSCs) residing in the hypoxic niches can both self-renew and give rise to progeny. Multiple regulatory mechanisms for these cellular processes have been identified. Emerging evidence has revealed that metabolism and bioenergetics play important roles in determining stem cell fate in concert with other regulatory networks. In this review, we will discuss recent advances in this field. RECENT FINDINGS Recent studies have helped define and redefine metabolic regulation of HSCs. Resting quiescent stem cells use primarily anaerobic glycolysis for energy production and this metabolic program is required to maintain a functional quiescent state. However, when they exit this state and rapidly proliferate and differentiate into different blood cell types, a robust up-regulation of energy metabolism is expected to meet the quickly rising energy demand. Dysregulation of metabolism in HSCs results in various blood disorders, including leukemia. SUMMARY Energy metabolism and HSC activity influence and interlink each other in a highly sophisticated and orchestrated manner. Understanding metabolic regulation of HSC function has significant implications for HSC-based therapies and leukemogenesis research.
Collapse
|
44
|
Abstract
Hematopoietic stem cells (HSCs) are inherently quiescent and self-renewing, yet can differentiate and commit to multiple blood cell types. Intracellular mitochondrial content is dynamic, and there is an increase in mitochondrial content during differentiation and lineage commitment in HSCs. HSCs reside in a hypoxic niche within the bone marrow and rely heavily on glycolysis, while differentiated and committed progenitors rely on oxidative phosphorylation. Increased oxidative phosphorylation during differentiation and commitment is not only due to increased mitochondrial content but also due to changes in mitochondrial cytosolic distribution and efficiency. These changes in the intracellular mitochondrial landscape contribute signals toward regulating differentiation and commitment. Thus, a functional relationship exists between the mitochondria in HSCs and the state of the HSCs (i.e., stemness vs. differentiated). This review focuses on how autophagy-mediated mitochondrial clearance (i.e., mitophagy) may affect HSC mitochondrial content, thereby influencing the fate of HSCs and maintenance of hematopoietic homeostasis.
Collapse
Affiliation(s)
- Aashish Joshi
- Department of Pathology; St. Jude Children's Research Hospital; Memphis, TN USA
| | | |
Collapse
|
45
|
FLVCR is necessary for erythroid maturation, may contribute to platelet maturation, but is dispensable for normal hematopoietic stem cell function. Blood 2013; 122:2903-10. [PMID: 24021674 DOI: 10.1182/blood-2012-10-465104] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Heme is a pleiotropic molecule that is important for oxygen and oxidative metabolism, most notably as the prosthetic group of hemoglobin and cytochromes. Because excess free intracellular heme is toxic, organisms have developed mechanisms to tightly regulate its concentration. One mechanism is through active heme export by the group C feline leukemia virus receptor (FLVCR). Previously, we have shown that FLVCR is necessary for embryonic and postnatal erythropoiesis. However, FLVCR is also expressed in numerous other tissues, including hematopoietic stem cells (HSCs). To explore a possible role for FLVCR in HSC function, we performed serial, competitive repopulation transplant experiments using FLVCR-deleted and control bone marrow cells, along with wild-type competitor cells. Loss of FLVCR did not impact HSC function under steady-state or myelotoxic stress conditions (such as arsenic or radiation exposure), nor did FLVCR deletion result in alterations in the various progenitor compartments. However, even when 95% of the donor bone marrow cells lacked FLVCR, all red cells in recipient mice were wild type. This is due to the increased apoptosis of FLVCR-deleted proerythroblasts. Also, remarkably, loss of FLVCR increased megakaryocyte ploidy. Together, these findings show FLVCR is redundant in stem cells but has critical and contrasting stage-specific roles in discrete hematopoietic lineages.
Collapse
|
46
|
Yamaguchi M, Kashiwakura I. Role of reactive oxygen species in the radiation response of human hematopoietic stem/progenitor cells. PLoS One 2013; 8:e70503. [PMID: 23936220 PMCID: PMC3723682 DOI: 10.1371/journal.pone.0070503] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 06/20/2013] [Indexed: 01/10/2023] Open
Abstract
Hematopoietic stem/progenitor cells (HSPCs), which are present in small numbers in hematopoietic tissues, can differentiate into all hematopoietic lineages and self-renew to maintain their undifferentiated phenotype. HSPCs are extremely sensitive to oxidative stressors such as anti-cancer agents, radiation, and the extensive accumulation of reactive oxygen species (ROS). The quiescence and stemness of HSPCs are maintained by the regulation of mitochondrial biogenesis, ROS, and energy homeostasis in a special microenvironment called the stem cell niche. The present study evaluated the relationship between the production of intracellular ROS and mitochondrial function during the proliferation and differentiation of X-irradiated CD34+ cells prepared from human placental/umbilical cord blood HSPCs. Highly purified CD34+ HSPCs exposed to X-rays were cultured in liquid and semi-solid medium supplemented with hematopoietic cytokines. X-irradiated CD34+ HSPCs treated with hematopoietic cytokines, which promote their proliferation and differentiation, exhibited dramatically suppressed cell growth and clonogenic potential. The amount of intracellular ROS in X-irradiated CD34+ HSPCs was significantly higher than that in non-irradiated cells during the culture period. However, neither the intracellular mitochondrial content nor the mitochondrial superoxide production was elevated in X-irradiated CD34+ HSPCs compared with non-irradiated cells. Radiation-induced gamma-H2AX expression was observed immediately following exposure to 4 Gy of X-rays and gradually decreased during the culture period. This study reveals that X-irradiation can increase persistent intracellular ROS in human CD34+ HSPCs, which may not result from mitochondrial ROS due to mitochondrial dysfunction, and indicates that substantial DNA double-strand breakage can critically reduce the stem cell function.
Collapse
Affiliation(s)
- Masaru Yamaguchi
- Department of Radiological Life Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori, Japan
| | - Ikuo Kashiwakura
- Department of Radiological Life Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori, Japan
- * E-mail:
| |
Collapse
|
47
|
Romero-Moya D, Bueno C, Montes R, Navarro-Montero O, Iborra FJ, López LC, Martin M, Menendez P. Cord blood-derived CD34+ hematopoietic cells with low mitochondrial mass are enriched in hematopoietic repopulating stem cell function. Haematologica 2013; 98:1022-9. [PMID: 23349299 DOI: 10.3324/haematol.2012.079244] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The homeostasis of the hematopoietic stem/progenitor cell pool relies on a fine-tuned balance between self-renewal, differentiation and proliferation. Recent studies have proposed that mitochondria regulate these processes. Although recent work has contributed to understanding the role of mitochondria during stem cell differentiation, it remains unclear whether the mitochondrial content/function affects human hematopoietic stem versus progenitor function. We found that mitochondrial mass correlates strongly with mitochondrial membrane potential in CD34(+) hematopoietic stem/progenitor cells. We, therefore, sorted cord blood CD34(+) cells on the basis of their mitochondrial mass and analyzed the in vitro homeostasis and clonogenic potential as well as the in vivo repopulating potential of CD34(+) cells with high (CD34(+) Mito(High)) versus low (CD34(+) Mito(Low)) mitochondrial mass. The CD34(+) Mito(Low) fraction contained 6-fold more CD34(+)CD38(-) primitive cells and was enriched in hematopoietic stem cell function, as demonstrated by its significantly greater hematopoietic reconstitution potential in immuno-deficient mice. In contrast, the CD34(+) Mito(High) fraction was more enriched in hematopoietic progenitor function with higher in vitro clonogenic capacity. In vitro differentiation of CD34(+) Mito(Low) cells was significantly delayed as compared to that of CD34(+) Mito(High) cells. The eventual complete differentiation of CD34(+) Mito(Low) cells, which coincided with a robust expansion of the CD34(-) differentiated progeny, was accompanied by mitochondrial adaptation, as shown by significant increases in ATP production and expression of the mitochondrial genes ND1 and COX2. In conclusion, cord blood CD34(+) cells with low levels of mitochondrial mass are enriched in hematopoietic repopulating stem cell function whereas high levels of mitochondrial mass identify hematopoietic progenitors. A mitochondrial response underlies hematopoietic stem/progenitor cell differentiation and proliferation of lineage-committed CD34(-) cells.
Collapse
Affiliation(s)
- Damia Romero-Moya
- GENyO-Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Government, Granada, Spain
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Broxmeyer HE, Mantel C. A ROSy future for metabolic regulation of HSC division. Nat Med 2012; 18:1334-6. [PMID: 22961159 DOI: 10.1038/nm.2917] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
49
|
Broxmeyer HE. Enhancing engraftment of cord blood cells via insight into the biology of stem/progenitor cell function. Ann N Y Acad Sci 2012; 1266:151-60. [PMID: 22901266 PMCID: PMC3428743 DOI: 10.1111/j.1749-6632.2012.06509.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cord blood (CB) transplantation has been used over the last 24 years to treat patients with malignant and nonmalignant disorders. CB has its advantages and disadvantages compared with other sources of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) for transplantation. More knowledge of the cytokines and intracellular signaling molecules regulating HSCs and HPCs could be used to modulate these regulators for clinical benefit. This review provides information about the general field of CB transplantation and about studies from the author's laboratory that focus on regulation of HSCs and HPCs by CD26/DPPIV, SDF-1/CXCL12, the Rheb2-mTOR pathway, SIRT1, DEK, cyclin-dependent kinase inhibitors, and cytokines/growth factors. Cryopreservation of CB HSCs and HPCs is also briefly discussed.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
50
|
Mouse hematopoietic cell-targeted STAT3 deletion: stem/progenitor cell defects, mitochondrial dysfunction, ROS overproduction, and a rapid aging-like phenotype. Blood 2012; 120:2589-99. [PMID: 22665934 DOI: 10.1182/blood-2012-01-404004] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Nuclear transcription factor Stat3 is important for proper regulation of hematopoietic stem cell (HSC) and hematopoietic progenitor cell (HPC) proliferation, survival, and cytokine signaling responses. A new, noncanonical role for Stat3 in mitochondrial function has been discovered recently. However, there is little information on the role(s) of mitochondrial Stat3 in HSC/HPC function, especially potential effects of Stat3/mitochondrial dysregulation in human diseases. We investigated hematopoietic cell-targeted deletion of the STAT3 gene in HSCs/HPCs with a focus on mitochondrial function. We found that STAT3(-/-) mice, which have a very shortened lifespan, dysfunctional/dysregulated mitochondrial function and excessive reactive oxygen species production in HSCs/HPCs that coincides with pronounced defects in function. These animals have a blood phenotype with similarities to premature aging and to human diseases of myelodysplastic syndrome and myeloproliferative neoplasms such as erythroid dysplasia, anemia, excessive myeloproliferation, and lymphomyeloid ratio shifts. We show herein that the lifespan of STAT3(-/-) animals is lengthened by treatment with a reactive oxygen species scavenger, which lessened the severity of the blood phenotype. These data suggest a need for more detailed studies of role(s) of Stat3 in HSC/HPC mitochondrial function in human diseases and raise the idea that mitochondrial Stat3 could be used as a potential therapeutic target.
Collapse
|