101
|
Lin W, Chen S, Wang Y, Wang M, Lee WYW, Jiang X, Li G. Dynamic regulation of mitochondrial-endoplasmic reticulum crosstalk during stem cell homeostasis and aging. Cell Death Dis 2021; 12:794. [PMID: 34400615 PMCID: PMC8368094 DOI: 10.1038/s41419-021-03912-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 02/07/2023]
Abstract
Cellular therapy exerts profound therapeutic potential for curing a broad spectrum of diseases. Adult stem cells reside within a specified dynamic niche in vivo, which is essential for continuous tissue homeostatic maintenance through balancing self-renewal with lineage selection. Meanwhile, adult stem cells may be multipotent or unipotent, and are present in both quiescent and actively dividing states in vivo of the mammalians, which may switch to each other state in response to biophysical cues through mitochondria-mediated mechanisms, such as alterations in mitochondrial respiration and metabolism. In general, stem cells facilitate tissue repair after tissue-specific homing through various mechanisms, including immunomodulation of local microenvironment, differentiation into functional cells, cell "empowerment" via paracrine secretion, immunoregulation, and intercellular mitochondrial transfer. Interestingly, cell-source-specific features have been reported between different tissue-derived adult stem cells with distinct functional properties due to the different microenvironments in vivo, as well as differential functional properties in different tissue-derived stem cell-derived extracellular vehicles, mitochondrial metabolism, and mitochondrial transfer capacity. Here, we summarized the current understanding on roles of mitochondrial dynamics during stem cell homeostasis and aging, and lineage-specific differentiation. Also, we proposed potential unique mitochondrial molecular signature features between different source-derived stem cells and potential associations between stem cell aging and mitochondria-endoplasmic reticulum (ER) communication, as well as potential novel strategies for anti-aging intervention and healthy aging.
Collapse
Affiliation(s)
- Weiping Lin
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China.
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Shuxun Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China
| | - Yan Wang
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ming Wang
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wayne Yuk-Wai Lee
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Xiaohua Jiang
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
- Faculty of Medicine, MOE Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Gang Li
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
- Faculty of Medicine, MOE Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
102
|
Tang X, Wu H, Xie J, Wang N, Chen Q, Zhong Z, Qiu Y, Wang J, Li X, Situ P, Lai L, Zern MA, Chen H, Duan Y. The combination of dextran sulphate and polyvinyl alcohol prevents excess aggregation and promotes proliferation of pluripotent stem cells in suspension culture. Cell Prolif 2021; 54:e13112. [PMID: 34390064 PMCID: PMC8450127 DOI: 10.1111/cpr.13112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/13/2021] [Accepted: 07/24/2021] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES For clinical applications of cell-based therapies, a large quantity of human pluripotent stem cells (hPSCs) produced in standardized and scalable culture processes is required. Currently, microcarrier-free suspension culture shows potential for large-scale expansion of hPSCs; however, hPSCs tend to aggregate during culturing leading to a negative effect on cell yield. To overcome this problem, we developed a novel protocol to effectively control the sizes of cell aggregates and enhance the cell proliferation during the expansion of hPSCs in suspension. MATERIALS AND METHODS hPSCs were expanded in suspension culture supplemented with polyvinyl alcohol (PVA) and dextran sulphate (DS), and 3D suspension culture of hPSCs formed cell aggregates under static or dynamic conditions. The sizes of cell aggregates and the cell proliferation as well as the pluripotency of hPSCs after expansion were assessed using cell counting, size analysis, real-time quantitative polymerase chain reaction, flow cytometry analysis, immunofluorescence staining, embryoid body formation, teratoma formation and transcriptome sequencing. RESULTS Our results demonstrated that the addition of DS alone effectively prevented hPSC aggregation, while the addition of PVA significantly enhanced hPSC proliferation. The combination of PVA and DS not only promoted cell proliferation of hPSCs but also produced uniform and size-controlled cell aggregates. Moreover, hPSCs treated with PVA, or DS or a combination, maintained the pluripotency and were capable of differentiating into all three germ layers. mRNA-seq analysis demonstrated that the combination of PVA and DS significantly promoted hPSC proliferation and prevented cell aggregation through improving energy metabolism-related processes, regulating cell growth, cell proliferation and cell division, as well as reducing the adhesion among hPSC aggregates by affecting expression of genes related to cell adhesion. CONCLUSIONS Our results represent a significant step towards developing a simple and robust approach for the expansion of hPSCs in large scale.
Collapse
Affiliation(s)
- Xianglian Tang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China.,Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Haibin Wu
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Jinghe Xie
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China.,Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Ning Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China.,Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Qicong Chen
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China.,Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhiyong Zhong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China.,Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Yaqi Qiu
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Jue Wang
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiajing Li
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ping Situ
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Liangxue Lai
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Mark A Zern
- Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, USA
| | - Honglin Chen
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China.,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China.,Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, China.,Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, China.,Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| | - Yuyou Duan
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China.,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China.,Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, China.,Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, China.,Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| |
Collapse
|
103
|
Tortelote GG, Colón-Leyva M, Saifudeen Z. Metabolic programming of nephron progenitor cell fate. Pediatr Nephrol 2021; 36:2155-2164. [PMID: 33089379 PMCID: PMC10734399 DOI: 10.1007/s00467-020-04752-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/30/2020] [Accepted: 08/31/2020] [Indexed: 11/28/2022]
Abstract
Metabolic pathways are one of the first responses at the cellular level to maternal/fetal interface stressors. Studies have revealed the previously unrecognized contributions of intermediary metabolism to developmental programs. Here, we provide an overview of cellular metabolic pathways and the cues that modulate metabolic states. We discuss the developmental and physiological implications of metabolic reprogramming and the key role of metabolites in epigenetic and epiproteomic modifications during embryonic development and with respect to kidney development and nephrogenesis.
Collapse
Affiliation(s)
- Giovane G Tortelote
- Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue SL37, Room 5534, New Orleans, LA, 70112, USA
| | - Mariel Colón-Leyva
- Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue SL37, Room 5534, New Orleans, LA, 70112, USA
| | - Zubaida Saifudeen
- Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue SL37, Room 5534, New Orleans, LA, 70112, USA.
| |
Collapse
|
104
|
Xu Y, Zhang J, Hu Y, Li X, Sun L, Peng Y, Sun Y, Liu B, Bian Z, Rong Z. Single-cell transcriptome analysis reveals the dynamics of human immune cells during early fetal skin development. Cell Rep 2021; 36:109524. [PMID: 34380039 DOI: 10.1016/j.celrep.2021.109524] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 05/12/2021] [Accepted: 07/22/2021] [Indexed: 01/12/2023] Open
Abstract
The immune system of skin develops in stages in mice. However, the developmental dynamics of immune cells in human skin remains elusive. Here, we perform transcriptome profiling of CD45+ hematopoietic cells in human fetal skin at an estimated gestational age of 10-17 weeks by single-cell RNA sequencing. A total of 13 immune cell types are identified. Skin macrophages show dynamic heterogeneity over the course of skin development. A major shift in lymphoid cell developmental states occurs from the first to the second trimester that implies an in situ differentiation process. Gene expression analysis reveals a typical developmental program in immune cells in accordance with their functional maturation, possibly involving metabolic reprogramming. Finally, we identify transcription factors (TFs) that potentially regulate cellular transitions by comparing TFs and TF target gene networks. These findings provide detailed insight into how the immune system of the human skin is established during development.
Collapse
Affiliation(s)
- Yingping Xu
- Experimental Research Center, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China.
| | - Jun Zhang
- Experimental Research Center, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
| | - Yongfei Hu
- Experimental Research Center, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
| | - Xuefei Li
- Experimental Research Center, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
| | - Lihua Sun
- Development of Gynaecology and Obstetrics, Nanhai Hospital, Southern Medical University, Guangzhou 528200, China
| | - Yu Peng
- Experimental Research Center, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
| | - Yuzhe Sun
- Experimental Research Center, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
| | - Bing Liu
- State Key Laboratory of Experimental Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Zhilei Bian
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China.
| | - Zhili Rong
- Experimental Research Center, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China; Cancer Research Institute, School of Basic Medical Sciences, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Southern Medical University, Guangzhou 510515, China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China.
| |
Collapse
|
105
|
Voigt AL, Kondro DA, Powell D, Valli-Pulaski H, Ungrin M, Stukenborg JB, Klein C, Lewis IA, Orwig KE, Dobrinski I. Unique metabolic phenotype and its transition during maturation of juvenile male germ cells. FASEB J 2021; 35:e21513. [PMID: 33811704 DOI: 10.1096/fj.202002799r] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/06/2021] [Accepted: 02/23/2021] [Indexed: 12/22/2022]
Abstract
Human male reproductive development has a prolonged prepubertal period characterized by juvenile quiescence of germ cells with immature spermatogonial stem cell (SSC) precursors (gonocytes) present in the testis for an extended period of time. The metabolism of gonocytes is not defined. We demonstrate with mitochondrial ultrastructure studies via TEM and IHC and metabolic flux studies with UHPLC-MS that a distinct metabolic transition occurs during the maturation to SSCs. The mitochondrial ultrastructure of prepubertal human spermatogonia is shared with prepubertal pig spermatogonia. The metabolism of early prepubertal porcine spermatogonia (gonocytes) is characterized by the reliance on OXPHOS fuelled by oxidative decarboxylation of pyruvate. Interestingly, at the same time, a high amount of the consumed pyruvate is also reduced and excreted as lactate. With maturation, prepubertal spermatogonia show a metabolic shift with decreased OXHPOS and upregulation of the anaerobic metabolism-associated uncoupling protein 2 (UCP2). This shift is accompanied with stem cell specific promyelocytic leukemia zinc finger protein (PLZF) protein expression and glial cell-derived neurotropic factor (GDNF) pathway activation. Our results demonstrate that gonocytes differently from mature spermatogonia exhibit unique metabolic demands that must be attained to enable their maintenance and growth in vitro.
Collapse
Affiliation(s)
- Anna Laura Voigt
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Douglas Andrew Kondro
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Diana Powell
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Hanna Valli-Pulaski
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mark Ungrin
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Jan-Bernd Stukenborg
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet and Karolinska University Hospital, Solna, Sweden
| | - Claudia Klein
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Ian A Lewis
- Department of Biological Sciences, Faculty of Sciences, University of Calgary, Calgary, AB, Canada
| | - Kyle E Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ina Dobrinski
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
106
|
Sakae Y, Tanaka M. Metabolism and Sex Differentiation in Animals from a Starvation Perspective. Sex Dev 2021; 15:168-178. [PMID: 34284403 DOI: 10.1159/000515281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/12/2021] [Indexed: 11/19/2022] Open
Abstract
Animals determine their sex genetically (GSD: genetic sex determination) and/or environmentally (ESD: environmental sex determination). Medaka (Oryzias latipes) employ a XX/XY GSD system, however, they display female-to-male sex reversal in response to various environmental changes such as temperature, hypoxia, and green light. Interestingly, we found that 5 days of starvation during sex differentiation caused female-to-male sex reversal. In this situation, the metabolism of pantothenate and fatty acid synthesis plays an important role in sex reversal. Metabolism is associated with other biological factors such as germ cells, HPG axis, lipids, and epigenetics, and supplys substances and acts as signal transducers. In this review, we discuss the importance of metabolism during sex differentiation and how metabolism contributes to sex differentiation.
Collapse
Affiliation(s)
- Yuta Sakae
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Minoru Tanaka
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
107
|
Hebbar S, Knust E. Reactive oxygen species (ROS) constitute an additional player in regulating epithelial development. Bioessays 2021; 43:e2100096. [PMID: 34260754 DOI: 10.1002/bies.202100096] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 12/18/2022]
Abstract
Reactive oxygen species (ROS) are highly reactive molecules produced in cells. So far, they have mostly been connected to diseases and pathological conditions. More recent results revealed a somewhat unexpected role of ROS in control of developmental processes. In this review, we elaborate on ROS in development, focussing on their connection to epithelial tissue morphogenesis. After briefly summarising unique characteristics of epithelial cells, we present some characteristic features of ROS species, their production and targets, with a focus on proteins important for epithelial development and function. Finally, we provide examples of regulation of epithelial morphogenesis by ROS, and also of developmental genes that regulate the overall redox status. We conclude by discussing future avenues of research that will further elucidate ROS regulation in epithelial development.
Collapse
Affiliation(s)
- Sarita Hebbar
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Elisabeth Knust
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
108
|
Tejedor G, Contreras-Lopez R, Barthelaix A, Ruiz M, Noël D, De Ceuninck F, Pastoureau P, Luz-Crawford P, Jorgensen C, Djouad F. Pyrroline-5-Carboxylate Reductase 1 Directs the Cartilage Protective and Regenerative Potential of Murphy Roths Large Mouse Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:604756. [PMID: 34277596 PMCID: PMC8284254 DOI: 10.3389/fcell.2021.604756] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 06/03/2021] [Indexed: 11/16/2022] Open
Abstract
Murphy Roths Large (MRL) mice possess outstanding capacity to regenerate several tissues. In the present study, we investigated whether this regenerative potential could be associated with the intrinsic particularities possessed by their mesenchymal stem cells (MSCs). We demonstrated that MSCs derived from MRL mice (MRL MSCs) display a superior chondrogenic potential than do C57BL/6 MSC (BL6 MSCs). This higher chondrogenic potential of MRL MSCs was associated with a higher expression level of pyrroline-5-carboxylate reductase 1 (PYCR1), an enzyme that catalyzes the biosynthesis of proline, in MRL MSCs compared with BL6 MSCs. The knockdown of PYCR1 in MRL MSCs, using a specific small interfering RNA (siRNA), abolishes their chondrogenic potential. Moreover, we showed that PYCR1 silencing in MRL MSCs induced a metabolic switch from glycolysis to oxidative phosphorylation. In two in vitro chondrocyte models that reproduce the main features of osteoarthritis (OA) chondrocytes including a downregulation of chondrocyte markers, a significant decrease of PYCR1 was observed. A downregulation of chondrocyte markers was also observed by silencing PYCR1 in freshly isolated healthy chondrocytes. Regarding MSC chondroprotective properties on chondrocytes with OA features, we showed that MSCs silenced for PYCR1 failed to protect chondrocytes from a reduced expression of anabolic markers, while MSCs overexpressing PYCR1 exhibited an increased chondroprotective potential. Finally, using the ear punch model, we demonstrated that MRL MSCs induced a regenerative response in non-regenerating BL6 mice, while BL6 and MRL MSCs deficient for PYCR1 did not. In conclusion, our results provide evidence that MRL mouse regenerative potential is, in part, attributed to its MSCs that exhibit higher PYCR1-dependent glycolytic potential, differentiation capacities, chondroprotective abilities, and regenerative potential than BL6 MSCs.
Collapse
Affiliation(s)
| | | | | | - Maxime Ruiz
- IRMB, INSERM, University Montpellier, Montpellier, France
| | - Danièle Noël
- IRMB, INSERM, University Montpellier, Montpellier, France.,CHU Montpellier, Montpellier, France
| | - Frédéric De Ceuninck
- Center for Therapeutic Innovation, Immuno-Inflammatory Disease, Institut de Recherches Servier, Croissy-sur-Seine, France
| | - Philippe Pastoureau
- Center for Therapeutic Innovation, Immuno-Inflammatory Disease, Institut de Recherches Servier, Croissy-sur-Seine, France
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Christian Jorgensen
- IRMB, INSERM, University Montpellier, Montpellier, France.,CHU Montpellier, Montpellier, France
| | - Farida Djouad
- IRMB, INSERM, University Montpellier, Montpellier, France
| |
Collapse
|
109
|
Nair S, Rocha‐Ferreira E, Fleiss B, Nijboer CH, Gressens P, Mallard C, Hagberg H. Neuroprotection offered by mesenchymal stem cells in perinatal brain injury: Role of mitochondria, inflammation, and reactive oxygen species. J Neurochem 2021; 158:59-73. [PMID: 33314066 PMCID: PMC8359360 DOI: 10.1111/jnc.15267] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022]
Abstract
Preclinical studies have shown that mesenchymal stem cells have a positive effect in perinatal brain injury models. The mechanisms that cause these neurotherapeutic effects are not entirely intelligible. Mitochondrial damage, inflammation, and reactive oxygen species are considered to be critically involved in the development of injury. Mesenchymal stem cells have immunomodulatory action and exert mitoprotective effects which attenuate production of reactive oxygen species and promote restoration of tissue function and metabolism after perinatal insults. This review summarizes the present state, the underlying causes, challenges and possibilities for effective clinical translation of mesenchymal stem cell therapy.
Collapse
Affiliation(s)
- Syam Nair
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Neuroscience and PhysiologySahlgrenska Academy, University of GothenburgGothenburgSweden
- Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Eridan Rocha‐Ferreira
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Neuroscience and PhysiologySahlgrenska Academy, University of GothenburgGothenburgSweden
- Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Bobbi Fleiss
- School of Health and Biomedical SciencesRMIT UniversityBundooraVictoriaAustralia
- Université de Paris, NeuroDiderotParisFrance
| | - Cora H Nijboer
- Department for Developmental Origins of DiseaseUniversity Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht UniversityUtrechtNetherlands
| | | | - Carina Mallard
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Neuroscience and PhysiologySahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Henrik Hagberg
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| |
Collapse
|
110
|
Fry AL, Webster AK, Burnett J, Chitrakar R, Baugh LR, Hubbard EJA. DAF-18/PTEN inhibits germline zygotic gene activation during primordial germ cell quiescence. PLoS Genet 2021; 17:e1009650. [PMID: 34288923 PMCID: PMC8294487 DOI: 10.1371/journal.pgen.1009650] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
Quiescence, an actively-maintained reversible state of cell cycle arrest, is not well understood. PTEN is one of the most frequently lost tumor suppressors in human cancers and regulates quiescence of stem cells and cancer cells. The sole PTEN ortholog in Caenorhabditis elegans is daf-18. In a C. elegans loss-of-function mutant for daf-18, primordial germ cells (PGCs) divide inappropriately in L1 larvae hatched into starvation conditions, in a TOR-dependent manner. Here, we further investigated the role of daf-18 in maintaining PGC quiescence in L1 starvation. We found that maternal or zygotic daf-18 is sufficient to maintain cell cycle quiescence, that daf-18 acts in the germ line and soma, and that daf-18 affects timing of PGC divisions in fed animals. Importantly, our results also implicate daf-18 in repression of germline zygotic gene activation, though not in germline fate specification. However, TOR is less important to germline zygotic gene expression, suggesting that in the absence of food, daf-18/PTEN prevents inappropriate germline zygotic gene activation and cell division by distinct mechanisms.
Collapse
Affiliation(s)
- Amanda L. Fry
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, NYU Grossman School of Medicine, New York, New York, United States of America
| | - Amy K. Webster
- Department of Biology, Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, United States of America
| | - Julia Burnett
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, NYU Grossman School of Medicine, New York, New York, United States of America
| | - Rojin Chitrakar
- Department of Biology, Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, United States of America
| | - L. Ryan Baugh
- Department of Biology, Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, United States of America
| | - E. Jane Albert Hubbard
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, NYU Grossman School of Medicine, New York, New York, United States of America
| |
Collapse
|
111
|
Deficiency of Antioxidative Paraoxonase 2 (Pon2) Leads to Increased Number of Phenotypic LT-HSCs and Disturbed Erythropoiesis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3917028. [PMID: 34257800 PMCID: PMC8253644 DOI: 10.1155/2021/3917028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 04/26/2021] [Accepted: 05/27/2021] [Indexed: 01/09/2023]
Abstract
Background Long-term hematopoietic stem cells (LT-HSCs) reside in bone marrow niches with tightly controlled reactive oxygen species (ROS) levels. ROS increase results into LT-HSC differentiation and stem cell exhaustion. Paraoxonase 2 (PON2) has been shown to be important for ROS control. Objectives We investigate the effects of inactivation of the PON2 gene on hematopoietic cell differentiation and activity. Methods and Results In young mice with inactivated Pon2 gene (Pon2−/−, <3 months), we observed an increase of LT-HSCs and a reduced frequency of progenitor cells. In competitive transplantations, young Pon2−/− BM outcompeted WT BM at early time points. ROS levels were significantly increased in Pon2−/− whole BM, but not in Pon2−/− LT-HSCs. In more differentiated stages of hematopoiesis, Pon2 deficiency led to a misbalanced erythropoiesis both in physiologic and stress conditions. In older mice (>9 months), Pon2 depletion caused an increase in LT-HSCs as well as increased levels of granulocyte/macrophage progenitors (GMPs) and myeloid skewing, indicating a premature aging phenotype. No significant changes in ROS levels in old Pon2−/− LT- and short-term (ST-) HSCs were observed, but a significant reduction of spontaneous apoptotic cell death was measured. RNA-seq analysis in Pon2−/− LT-HSCs identified overrepresentation of genes involved in the C-X-C chemokine receptor type 4 (Cxcr4) signaling, suggesting compensatory mechanisms to overcome ROS-mediated accelerated aging in hematopoietic progenitor cells. Conclusions In summary, our current data indicate that PON2 is involved in the regulation of HSC functions.
Collapse
|
112
|
Categorizing the characteristics of human carcinogens: a need for specificity. Arch Toxicol 2021; 95:2883-2889. [PMID: 34148101 DOI: 10.1007/s00204-021-03109-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 06/17/2021] [Indexed: 02/07/2023]
Abstract
The International Agency for Research on Cancer (IARC) has recently proposed employing "ten key characteristics of human carcinogens" (TKCs) to determine the potential of agents for harmful effects. The TKCs seem likely to confuse the unsatisfactory correlation from testing regimes that have ignored the differences evident when cellular changes are compared in short and long-lived species, with their very different stem cell and somatic cell phylogenies. The proposed characteristics are so broad that their use will lead to an increase in the current unacceptably high rate of false positives. It could be an informative experiment to take well-established approved therapeutics with well-known human safety profiles and test them against this new TKC paradigm. Cancers are initiated and driven by heritable and transient changes in gene expression, expand clonally, and progress via additional associated acquired mutations and epigenetic alterations that provide cells with an evolutionary advantage. The genotoxicity testing protocols currently employed and required by regulation, emphasize testing for the mutational potential of the test agent. Two-year, chronic rodent cancer bioassays are intended to test for the entire spectrum of carcinogenic transformation. The use of cytotoxic doses causing increased, sustained cell proliferation that facilitates accumulated genetic damage leads to a high false-positive rate of tumor induction. Current cancer hazard assessment protocols and weight-of-the-evidence analysis of agent-specific cancer risk align poorly with the pathogenesis of human carcinoma and so need modernization and improvement in ways suggested here.
Collapse
|
113
|
Soh R, Hardy A, Zur Nieden NI. The FOXO signaling axis displays conjoined functions in redox homeostasis and stemness. Free Radic Biol Med 2021; 169:224-237. [PMID: 33878426 PMCID: PMC9910585 DOI: 10.1016/j.freeradbiomed.2021.04.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023]
Abstract
Previous views of reactive oxygen species (ROS) depicted them as harmful byproducts of metabolism as uncontrolled levels of ROS can lead to DNA damage and cell death. However, recent studies have shed light into the key role of ROS in the self-renewal or differentiation of the stem cell. The interplay between ROS levels, metabolism, and the downstream redox signaling pathways influence stem cell fate. In this review we will define ROS, explain how they are generated, and how ROS signaling can influence transcription factors, first and foremost forkhead box-O transcription factors, that shape not only the cellular redox state, but also stem cell fate. Now that studies have illustrated the importance of redox homeostasis and the role of redox signaling, understanding the mechanisms behind this interplay will further shed light into stem cell biology.
Collapse
Affiliation(s)
- Ruthia Soh
- Department of Molecular, Cell and Systems Biology, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, 92521, CA, USA
| | - Ariana Hardy
- Department of Molecular, Cell and Systems Biology, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, 92521, CA, USA
| | - Nicole I Zur Nieden
- Department of Molecular, Cell and Systems Biology, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, 92521, CA, USA; Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, 92521, CA, USA.
| |
Collapse
|
114
|
Fus-Kujawa A, Mendrek B, Trybus A, Bajdak-Rusinek K, Stepien KL, Sieron AL. Potential of Induced Pluripotent Stem Cells for Use in Gene Therapy: History, Molecular Bases, and Medical Perspectives. Biomolecules 2021; 11:biom11050699. [PMID: 34067183 PMCID: PMC8151405 DOI: 10.3390/biom11050699] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 01/14/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) are defined as reprogrammed somatic cells exhibiting embryonic stem cell characteristics. Since their discovery in 2006, efforts have been made to utilize iPSCs in clinical settings. One of the promising fields of medicine, in which genetically patient-specific stem cells may prove themselves useful, is gene therapy. iPSCs technology holds potential in both creating models of genetic diseases and delivering therapeutic agents into the organism via auto-transplants, which reduces the risk of rejection compared to allotransplants. However, in order to safely administer genetically corrected stem cells into patients’ tissues, efforts must be made to establish stably pluripotent stem cells and reduce the risk of insertional tumorigenesis. In order to achieve this, optimal reprogramming factors and vectors must be considered. Therefore, in this review, the molecular bases of reprogramming safe iPSCs for clinical applications and recent attempts to translate iPSCs technology into the clinical setting are discussed.
Collapse
Affiliation(s)
- Agnieszka Fus-Kujawa
- Department of Molecular Biology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland; (A.F.-K.); (A.T.); (K.L.S.)
| | - Barbara Mendrek
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, M. Curie-Sklodowskiej 34, 41-819 Zabrze, Poland;
| | - Anna Trybus
- Department of Molecular Biology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland; (A.F.-K.); (A.T.); (K.L.S.)
| | - Karolina Bajdak-Rusinek
- Department of Medical Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland;
| | - Karolina L. Stepien
- Department of Molecular Biology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland; (A.F.-K.); (A.T.); (K.L.S.)
| | - Aleksander L. Sieron
- Department of Molecular Biology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland; (A.F.-K.); (A.T.); (K.L.S.)
- Correspondence:
| |
Collapse
|
115
|
Tiwari SK, Mandal S. Mitochondrial Control of Stem Cell State and Fate: Lessons From Drosophila. Front Cell Dev Biol 2021; 9:606639. [PMID: 34012959 PMCID: PMC8128071 DOI: 10.3389/fcell.2021.606639] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 04/06/2021] [Indexed: 01/09/2023] Open
Abstract
Over the years, Drosophila has served as a wonderful genetically tractable model system to unravel various facets of tissue-resident stem cells in their microenvironment. Studies in different stem and progenitor cell types of Drosophila have led to the discovery of cell-intrinsic and extrinsic factors crucial for stem cell state and fate. Though initially touted as the ATP generating machines for carrying various cellular processes, it is now increasingly becoming clear that mitochondrial processes alone can override the cellular program of stem cells. The last few years have witnessed a surge in our understanding of mitochondria's contribution to governing different stem cell properties in their subtissular niches in Drosophila. Through this review, we intend to sum up and highlight the outcome of these in vivo studies that implicate mitochondria as a central regulator of stem cell fate decisions; to find the commonalities and uniqueness associated with these regulatory mechanisms.
Collapse
Affiliation(s)
- Satish Kumar Tiwari
- Developmental Genetics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, India
| | - Sudip Mandal
- Molecular Cell and Developmental Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, India
| |
Collapse
|
116
|
Yang FR, Zhao YF, Hu XW, Liu ZK, Yu XD, Li CY, Li XR, Li HJ. Nano-realgar suppresses lung cancer stem cell growth by repressing metabolic reprogramming. Gene 2021; 788:145666. [PMID: 33887368 DOI: 10.1016/j.gene.2021.145666] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/08/2021] [Accepted: 04/15/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Recent studies in cancer biology suggest that metabolic glucose reprogramming is a potential target for cancer treatment. However, little is known about drug intervention in the glucose metabolism of cancer stem cells (CSCs) and its related underlying mechanisms. METHODS The crude realgar powder was Nano-grinded to meets the requirements of Nano-pharmaceutical preparations, and Nano-realgar solution (NRS) was prepared for subsequent experiments. Isolation and characterization of lung cancer stem cells (LCSCs) was performed by magnetic cell sorting (MACS) and immunocytochemistry, respectively. Cell viability and intracellular glucose concentration were detected by MTT assay and glucose oxidase (GOD) kit. Protein expressions related to metabolic reprogramming was detected by ELISA assay. Determination of the expression of HIF-1α and PI3K/Akt/mTOR pathways was carried out by RT-PCR and western blotting analysis. A subcutaneous tumor model in BALB/c-nu mice was successfully established to evaluate the effects of Nano-realgar on tumor growth and histological structure, and the expression of HIF-1α in tumor tissues was measured by immunofluorescence. RESULTS Nano-realgar inhibits cell viability and induces glucose metabolism in LCSCs, and inhibits protein expression related to metabolic reprogramming in a time- and dose-dependent manner. Nano-realgar downregulated the expression of HIF-1α and PI3K/Akt/mTOR pathways in vitro and in vivo. Nano-realgar inhibits tumor growth and changes the histological structure of tumors through in vivo experiments and consequently inhibits the constitutive activation of HIF-1α signaling. CONCLUSIONS These results reveal that Nano-realgar inhibits tumor growth in vitro and in vivo by repressing metabolic reprogramming. This inhibitory effect potentially related to the downregulation HIF-1α expression via PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Fei-Ran Yang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| | - Yu-Feng Zhao
- Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| | - Xi-Wen Hu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| | - Zong-Kai Liu
- Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| | - Xiao-Dan Yu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| | - Chang-Yang Li
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| | - Xiu-Rong Li
- Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| | - Hui-Jie Li
- Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
117
|
Perottoni S, Neto NGB, Di Nitto C, Dmitriev RI, Raimondi MT, Monaghan MG. Intracellular label-free detection of mesenchymal stem cell metabolism within a perivascular niche-on-a-chip. LAB ON A CHIP 2021; 21:1395-1408. [PMID: 33605282 DOI: 10.1039/d0lc01034k] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The stem cell niche at the perivascular space in human tissue plays a pivotal role in dictating the overall fate of stem cells within it. Mesenchymal stem cells (MSCs) in particular, experience influential microenvironmental conditions, which induce specific metabolic profiles that affect processes of cell differentiation and dysregulation of the immunomodulatory function. Reports focusing specifically on the metabolic status of MSCs under the effect of pathophysiological stimuli - in terms of flow velocities, shear stresses or oxygen tension - do not model heterogeneous gradients, highlighting the need for more advanced models reproducing the metabolic niche. Organ-on-a-chip technology offers the most advanced tools for stem cell niche modelling thus allowing for controlled dynamic culture conditions while profiling tuneable oxygen tension gradients. However, current systems for live cell detection of metabolic activity inside microfluidic devices require the integration of microsensors. The presence of such microsensors poses the potential to alter microfluidics and their resolution does not enable intracellular measurements but rather a global representation concerning cellular metabolism. Here, we present a metabolic toolbox coupling a miniaturised in vitro system for human-MSCs dynamic culture, which mimics microenvironmental conditions of the perivascular niche, with high-resolution imaging of cell metabolism. Using fluorescence lifetime imaging microscopy (FLIM) we monitor the spatial metabolic machinery and correlate it with experimentally validated intracellular oxygen concentration after designing the oxygen tension decay along the fluidic chamber by in silico models prediction. Our platform allows the metabolic regulation of MSCs, mimicking the physiological niche in space and time, and its real-time monitoring representing a functional tool for modelling perivascular niches, relevant diseases and metabolic-related uptake of pharmaceuticals.
Collapse
Affiliation(s)
- Simone Perottoni
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza Leonardo da Vinci, 32 - 20133 Milan, Italy.
| | | | | | | | | | | |
Collapse
|
118
|
Rodrigues DC, Mufteev M, Weatheritt RJ, Djuric U, Ha KCH, Ross PJ, Wei W, Piekna A, Sartori MA, Byres L, Mok RSF, Zaslavsky K, Pasceri P, Diamandis P, Morris Q, Blencowe BJ, Ellis J. Shifts in Ribosome Engagement Impact Key Gene Sets in Neurodevelopment and Ubiquitination in Rett Syndrome. Cell Rep 2021; 30:4179-4196.e11. [PMID: 32209477 DOI: 10.1016/j.celrep.2020.02.107] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 12/30/2019] [Accepted: 02/27/2020] [Indexed: 12/21/2022] Open
Abstract
Regulation of translation during human development is poorly understood, and its dysregulation is associated with Rett syndrome (RTT). To discover shifts in mRNA ribosomal engagement (RE) during human neurodevelopment, we use parallel translating ribosome affinity purification sequencing (TRAP-seq) and RNA sequencing (RNA-seq) on control and RTT human induced pluripotent stem cells, neural progenitor cells, and cortical neurons. We find that 30% of transcribed genes are translationally regulated, including key gene sets (neurodevelopment, transcription and translation factors, and glycolysis). Approximately 35% of abundant intergenic long noncoding RNAs (lncRNAs) are ribosome engaged. Neurons translate mRNAs more efficiently and have longer 3' UTRs, and RE correlates with elements for RNA-binding proteins. RTT neurons have reduced global translation and compromised mTOR signaling, and >2,100 genes are translationally dysregulated. NEDD4L E3-ubiquitin ligase is translationally impaired, ubiquitinated protein levels are reduced, and protein targets accumulate in RTT neurons. Overall, the dynamic translatome in neurodevelopment is disturbed in RTT and provides insight into altered ubiquitination that may have therapeutic implications.
Collapse
Affiliation(s)
- Deivid C Rodrigues
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Marat Mufteev
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Robert J Weatheritt
- Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Ugljesa Djuric
- Laboratory Medicine and Pathology Program, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Kevin C H Ha
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada; Vector Institute, 661 University Avenue, Toronto, ON M5G 1M1, Canada
| | - P Joel Ross
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Wei Wei
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Alina Piekna
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Maria A Sartori
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Loryn Byres
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Rebecca S F Mok
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Kirill Zaslavsky
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Peter Pasceri
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Phedias Diamandis
- Laboratory Medicine and Pathology Program, University Health Network, Toronto, ON M5G 2C4, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada; Department of Pathology, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Quaid Morris
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada; Vector Institute, 661 University Avenue, Toronto, ON M5G 1M1, Canada
| | - Benjamin J Blencowe
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - James Ellis
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
119
|
Büeler H. Mitochondrial and Autophagic Regulation of Adult Neurogenesis in the Healthy and Diseased Brain. Int J Mol Sci 2021; 22:ijms22073342. [PMID: 33805219 PMCID: PMC8036818 DOI: 10.3390/ijms22073342] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 02/07/2023] Open
Abstract
Adult neurogenesis is a highly regulated process during which new neurons are generated from neural stem cells in two discrete regions of the adult brain: the subventricular zone of the lateral ventricle and the subgranular zone of the dentate gyrus in the hippocampus. Defects of adult hippocampal neurogenesis have been linked to cognitive decline and dysfunction during natural aging and in neurodegenerative diseases, as well as psychological stress-induced mood disorders. Understanding the mechanisms and pathways that regulate adult neurogenesis is crucial to improving preventative measures and therapies for these conditions. Accumulating evidence shows that mitochondria directly regulate various steps and phases of adult neurogenesis. This review summarizes recent findings on how mitochondrial metabolism, dynamics, and reactive oxygen species control several aspects of adult neural stem cell function and their differentiation to newborn neurons. It also discusses the importance of autophagy for adult neurogenesis, and how mitochondrial and autophagic dysfunction may contribute to cognitive defects and stress-induced mood disorders by compromising adult neurogenesis. Finally, I suggest possible ways to target mitochondrial function as a strategy for stem cell-based interventions and treatments for cognitive and mood disorders.
Collapse
Affiliation(s)
- Hansruedi Büeler
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin 150080, China
| |
Collapse
|
120
|
Tomecka E, Lech W, Zychowicz M, Sarnowska A, Murzyn M, Oldak T, Domanska-Janik K, Buzanska L, Rozwadowska N. Assessment of the Neuroprotective and Stemness Properties of Human Wharton's Jelly-Derived Mesenchymal Stem Cells under Variable (5% vs. 21%) Aerobic Conditions. Cells 2021; 10:717. [PMID: 33804841 PMCID: PMC8063843 DOI: 10.3390/cells10040717] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/20/2021] [Accepted: 03/21/2021] [Indexed: 12/20/2022] Open
Abstract
To optimise the culture conditions for human Wharton's jelly-derived mesenchymal stem cells (hWJ-MSCs) intended for clinical use, we investigated ten different properties of these cells cultured under 21% (atmospheric) and 5% (physiological normoxia) oxygen concentrations. The obtained results indicate that 5% O2 has beneficial effects on the proliferation rate, clonogenicity, and slowdown of senescence of hWJ-MSCs; however, the oxygen level did not have an influence on the cell morphology, immunophenotype, or neuroprotective effect of the hWJ-MSCs. Nonetheless, the potential to differentiate into adipocytes, osteocytes, and chondrocytes was comparable under both oxygen conditions. However, spontaneous differentiation of hWJ-MSCs into neuronal lineages was observed and enhanced under atmospheric oxygen conditions. The cells relied more on mitochondrial respiration than glycolysis, regardless of the oxygen conditions. Based on these results, we can conclude that hWJ-MSCs could be effectively cultured and prepared under both oxygen conditions for cell-based therapy. However, the 5% oxygen level seemed to create a more balanced and appropriate environment for hWJ-MSCs.
Collapse
Affiliation(s)
- Ewelina Tomecka
- Polish Stem Cell Bank, FamiCord Group, 00-867 Warsaw, Poland; (E.T.); (M.M.); (T.O.)
| | - Wioletta Lech
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (W.L.); (M.Z.); (A.S.); (K.D.-J.)
| | - Marzena Zychowicz
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (W.L.); (M.Z.); (A.S.); (K.D.-J.)
| | - Anna Sarnowska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (W.L.); (M.Z.); (A.S.); (K.D.-J.)
| | - Magdalena Murzyn
- Polish Stem Cell Bank, FamiCord Group, 00-867 Warsaw, Poland; (E.T.); (M.M.); (T.O.)
| | - Tomasz Oldak
- Polish Stem Cell Bank, FamiCord Group, 00-867 Warsaw, Poland; (E.T.); (M.M.); (T.O.)
| | - Krystyna Domanska-Janik
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (W.L.); (M.Z.); (A.S.); (K.D.-J.)
| | - Leonora Buzanska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (W.L.); (M.Z.); (A.S.); (K.D.-J.)
| | - Natalia Rozwadowska
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland;
| |
Collapse
|
121
|
Jeske R, Yuan X, Fu Q, Bunnell BA, Logan TM, Li Y. In Vitro Culture Expansion Shifts the Immune Phenotype of Human Adipose-Derived Mesenchymal Stem Cells. Front Immunol 2021; 12:621744. [PMID: 33777002 PMCID: PMC7988085 DOI: 10.3389/fimmu.2021.621744] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/05/2021] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stem or stromal cells (hMSCs) are known for their potential in regenerative medicine due to their differentiation abilities, secretion of trophic factors, and regulation of immune responses in damaged tissues. Due to the limited quantity of hMSCs typically isolated from bone marrow, other tissue sources, such as adipose tissue-derived mesenchymal stem cells (hASCs), are considered a promising alternative. However, differences have been observed for hASCs in the context of metabolic characteristics and response to in vitro culture stress compared to bone marrow derived hMSCs (BM-hMSCs). In particular, the relationship between metabolic homeostasis and stem cell functions, especially the immune phenotype and immunomodulation of hASCs, remains unknown. This study thoroughly assessed the changes in metabolism, redox cycles, and immune phenotype of hASCs during in vitro expansion. In contrast to BM-hMSCs, hASCs did not respond to culture stress significantly during expansion as limited cellular senescence was observed. Notably, hASCs exhibited the increased secretion of pro-inflammatory cytokines and the decreased secretion of anti-inflammatory cytokines after extended culture expansion. The NAD+/NADH redox cycle and other metabolic characteristics associated with aging were relatively stable, indicating that hASC functional decline may be regulated through an alternative mechanism rather than NAD+/Sirtuin aging pathways as observed in BM-hMSCs. Furthermore, transcriptome analysis by mRNA-sequencing revealed the upregulation of genes for pro-inflammatory cytokines/chemokines and the downregulation of genes for anti-inflammatory cytokines for hASCs at high passage. Proteomics analysis indicated key pathways (e.g., tRNA charging, EIF2 signaling, protein ubiquitination pathway) that may be associated with the immune phenotype shift of hASCs. Together, this study advances our understanding of the metabolism and senescence of hASCs and may offer vital insights for the biomanufacturing of hASCs for clinical use.
Collapse
Affiliation(s)
- Richard Jeske
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Xuegang Yuan
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States.,The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, United States
| | - Qin Fu
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Bruce A Bunnell
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Timothy M Logan
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL, United States
| | - Yan Li
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States.,Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
122
|
Xi W, Chen W, Sun W, Li X, Suo Z, Jiang G, Gao P, Li Q. Mitochondrial activity regulates the differentiation of skin-derived mesenchymal stem cells into brown adipocytes to contribute to hypertension. Stem Cell Res Ther 2021; 12:167. [PMID: 33691786 PMCID: PMC7945215 DOI: 10.1186/s13287-021-02169-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 01/14/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Brown adipocytes (BAs) are major components of brown adipose tissue (BAT), which is involved in blood pressure regulation. BAs are derived from multiple progenitors, including PDGFRα+ adipose-derived stem cells (ASCs). Skin-derived mesenchymal stem cells (S-MSCs) have the capacity to differentiate into adipocytes; however, their ability to differentiate into BAs remains unexplored. We aim to study the ability and regulatory mechanism of the differentiation of S-MSCs into BAs and the direct role of BAT in blood pressure regulation. METHODS Protein expression was measured by flow cytometry or Western blotting, and gene mRNA levels were quantified by real-time quantitative PCR (RT-PCR). To induce the differentiation of S-MSCs into BAs, S-MSCs were stimulated with a brown adipogenic cocktail comprising insulin, IBMX, dexamethasone, triiodothyronine (T3), and rosiglitazone for the indicated periods. The oxygen consumption rate (OCR) was measured with an XF24 Extracellular Flux Analyzer. Mitochondrial mass was determined by flow cytometry and fluorescence staining. Hypertension was induced in WT mice by infusion of angiotensin II (Ang II), and systolic blood pressure (SBP) was measured using a tail cuff. Interscapular brown adipose tissue (iBAT)-deficient mice were generated by surgical removal of the iBAT depot, after which the animals were allowed to recover for 6 days. Aortic, iBAT, and heart tissue sections were analyzed by hematoxylin and eosin (HE) staining. RESULTS We found that in vitro, S-MSCs isolated from the mouse dermis expressed the stem cell markers CD90/105 and PDGFRα and readily differentiated into BAs. Mitochondrial biogenesis and oxygen consumption were markedly increased during differentiation of S-MSCs into BAs. In vivo, iBAT was converted to white adipose tissue (WAT) in Ang II-induced hypertensive mice. We assessed the direct role of BAT in blood pressure (BP) regulation by using iBAT-deficient mice (generated by surgical removal of iBAT) and C57BL/6 (wild-type (WT)) mice and found that Ang II-induced BP elevation and vascular damage were markedly aggravated in iBAT-deficient mice compared with WT mice. CONCLUSIONS This study demonstrates that PDGFRα+ S-MSCs are able to differentiate into BAs and that this differentiation is regulated by mitochondrial activity. We also show that BAT plays a direct role in ameliorating Ang II-induced hypertension. The therapeutic potential of BAT for the prevention of hypertension-induced organ remodeling thus warrants further investigation.
Collapse
Affiliation(s)
- Wenda Xi
- The Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Wendong Chen
- The Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Weihong Sun
- Department of Digestion, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Xiangxiao Li
- The Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Zhimin Suo
- Department of Digestion, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Gonghao Jiang
- The Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Pingjin Gao
- The Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Qun Li
- The Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China.
| |
Collapse
|
123
|
Jorgensen C, Khoury M. Musculoskeletal Progenitor/Stromal Cell-Derived Mitochondria Modulate Cell Differentiation and Therapeutical Function. Front Immunol 2021; 12:606781. [PMID: 33763061 PMCID: PMC7982675 DOI: 10.3389/fimmu.2021.606781] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/20/2021] [Indexed: 12/24/2022] Open
Abstract
Musculoskeletal stromal cells’ (MSCs’) metabolism impacts cell differentiation as well as immune function. During osteogenic and adipogenic differentiation, BM-MSCs show a preference for glycolysis during proliferation but shift to an oxidative phosphorylation (OxPhos)-dependent metabolism. The MSC immunoregulatory fate is achieved with cell polarization, and the result is sustained production of immunoregulatory molecules (including PGE2, HGF, IL1RA, IL6, IL8, IDO activity) in response to inflammatory stimuli. MSCs adapt their energetic metabolism when acquiring immunomodulatory property and shift to aerobic glycolysis. This can be achieved via hypoxia, pretreatment with small molecule-metabolic mediators such as oligomycin, or AKT/mTOR pathway modulation. The immunoregulatory effect of MSC on macrophages polarization and Th17 switch is related to the glycolytic status of the MSC. Indeed, MSCs pretreated with oligomycin decreased the M1/M2 ratio, inhibited T-CD4 proliferation, and prevented Th17 switch. Mitochondrial activity also impacts MSC metabolism. In the bone marrow, MSCs are present in a quiescent, low proliferation, but they keep their multi-progenitor function. In this stage, they appear to be glycolytic with active mitochondria (MT) status. During MSC expansion, we observed a metabolic shift toward OXPhos, coupled with an increased MT activity. An increased production of ROS and dysfunctional mitochondria is associated with the metabolic shift to glycolysis. In contrast, when MSC underwent chondro or osteoblast differentiation, they showed a decreased glycolysis and inhibition of the pentose phosphate pathway (PPP). In parallel the mitochondrial enzymatic activities increased associated with oxidative phosphorylation enhancement. MSCs respond to damaged or inflamed tissue through the transfer of MT to injured and immune cells, conveying a type of signaling that contributes to the restoration of cell homeostasis and immune function. The delivery of MT into injured cells increased ATP levels which in turn maintained cellular bioenergetics and recovered cell functions. MSC-derived MT may be transferred via tunneling nanotubes to undifferentiated cardiomyocytes and leading to their maturation. In this review, we will decipher the pathways and the mechanisms responsible for mitochondria transfer and activity. The eventual reversal of the metabolic and pro-inflammatory profile induced by the MT transfer will open new avenues for the control of inflammatory diseases.
Collapse
Affiliation(s)
- Christian Jorgensen
- Inserm, U1183, Montpellier, France.,Université MONTPELLIER 1, UFR de Médecine, Montpellier, France.,Service d'immuno-Rhumatologie, Hôpital Lapeyronie, Montpellier, France
| | - Maroun Khoury
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,Cells for Cells, Santiago, Chile.,Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| |
Collapse
|
124
|
Mohrin M. Mito-managing ROS & redox to reboot the immune system: Tapping mitochondria & redox management to extend the reach of hematopoietic stem cell transplantation. Free Radic Biol Med 2021; 165:38-53. [PMID: 33486089 DOI: 10.1016/j.freeradbiomed.2021.01.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 12/31/2022]
Abstract
Hematopoietic stem cells (HSCs) are responsible for life-long production of blood and immune cells. HSC transplantation (HSCT) is the original cell therapy which can cure hematological disorders but also has the potential to treat other diseases if technical and safety barriers are overcome. To maintain homeostatic hematopoiesis or to restore hematopoiesis during transplantation HSCs must perform both self-renewal, replication of themselves, and differentiation, generation of mature blood and immune cells. These are just two of the cell fate choices HSCs have; the transitional phases where HSCs undergo these cell fate decisions are regulated by reduction-oxidation (redox) signaling, mitochondrial activity, and cellular metabolism. Recent studies revealed that mitochondria, a key source of redox signaling components, are central to HSC cell fate decisions. Here we highlight how mitochondria serve as hubs in HSCs to manage redox signaling and metabolism and thus guide HSC fate choices. We focus on how mitochondrial activity is modulated by their clearance, biogenesis, dynamics, distribution, and quality control in HSCs. We also note how modulating mitochondria in HSCs can help overcome technical barriers limiting further use of HSCT.
Collapse
Affiliation(s)
- Mary Mohrin
- Immunology Discovery, Genentech, Inc. 1 DNA Way, South San Francisco, CA, 94080, USA.
| |
Collapse
|
125
|
Voigt AL, Thiageswaran S, de Lima e Martins Lara N, Dobrinski I. Metabolic Requirements for Spermatogonial Stem Cell Establishment and Maintenance In Vivo and In Vitro. Int J Mol Sci 2021; 22:1998. [PMID: 33670439 PMCID: PMC7922219 DOI: 10.3390/ijms22041998] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/11/2022] Open
Abstract
The spermatogonial stem cell (SSC) is a unique adult stem cell that requires tight physiological regulation during development and adulthood. As the foundation of spermatogenesis, SSCs are a potential tool for the treatment of infertility. Understanding the factors that are necessary for lifelong maintenance of a SSC pool in vivo is essential for successful in vitro expansion and safe downstream clinical usage. This review focused on the current knowledge of prepubertal testicular development and germ cell metabolism in different species, and implications for translational medicine. The significance of metabolism for cell biology, stem cell integrity, and fate decisions is discussed in general and in the context of SSC in vivo maintenance, differentiation, and in vitro expansion.
Collapse
Affiliation(s)
| | | | | | - Ina Dobrinski
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (A.L.V.); (S.T.); (N.d.L.e.M.L.)
| |
Collapse
|
126
|
Crespo D, Assis LHC, Zhang YT, Safian D, Furmanek T, Skaftnesmo KO, Norberg B, Ge W, Choi YC, den Broeder MJ, Legler J, Bogerd J, Schulz RW. Insulin-like 3 affects zebrafish spermatogenic cells directly and via Sertoli cells. Commun Biol 2021; 4:204. [PMID: 33589679 PMCID: PMC7884674 DOI: 10.1038/s42003-021-01708-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/13/2021] [Indexed: 01/31/2023] Open
Abstract
Pituitary hormones can use local signaling molecules to regulate target tissue functions. In adult zebrafish testes, follicle-stimulating hormone (Fsh) strongly increases the production of insulin-like 3 (Insl3), a Leydig cell-derived growth factor found in all vertebrates. Little information is available regarding Insl3 function in adult spermatogenesis. The Insl3 receptors Rxfp2a and 2b were expressed by type A spermatogonia and Sertoli and myoid cells, respectively, in zebrafish testis tissue. Loss of insl3 increased germ cell apoptosis in males starting at 9 months of age, but spermatogenesis appeared normal in fully fertile, younger adults. Insl3 changed the expression of 409 testicular genes. Among others, retinoic acid (RA) signaling was up- and peroxisome proliferator-activated receptor gamma (Pparg) signaling was down-regulated. Follow-up studies showed that RA and Pparg signaling mediated Insl3 effects, resulting in the increased production of differentiating spermatogonia. This suggests that Insl3 recruits two locally active nuclear receptor pathways to implement pituitary (Fsh) stimulation of spermatogenesis.
Collapse
Affiliation(s)
- Diego Crespo
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands ,grid.10917.3e0000 0004 0427 3161Present Address: Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Luiz H. C. Assis
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Yu Ting Zhang
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Fujian, PR China ,grid.449133.80000 0004 1764 3555Present Address: Institute of Oceanography, Minjiang University, Fuzhou, PR China
| | - Diego Safian
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands ,grid.4818.50000 0001 0791 5666Present Address: Experimental Zoology Group and Aquaculture and Fisheries Group, Department of Animal Science, Wageningen University, Wageningen, The Netherlands
| | - Tomasz Furmanek
- grid.10917.3e0000 0004 0427 3161Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Kai Ove Skaftnesmo
- grid.10917.3e0000 0004 0427 3161Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Birgitta Norberg
- grid.10917.3e0000 0004 0427 3161Institute of Marine Research, Austevoll Research Station, Storebø, Norway
| | - Wei Ge
- grid.437123.00000 0004 1794 8068Center of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau China
| | - Yung-Ching Choi
- grid.437123.00000 0004 1794 8068Center of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau China
| | - Marjo J. den Broeder
- grid.5477.10000000120346234Division of Toxicology, Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Juliette Legler
- grid.5477.10000000120346234Division of Toxicology, Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jan Bogerd
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Rüdiger W. Schulz
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands ,grid.10917.3e0000 0004 0427 3161Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| |
Collapse
|
127
|
Altundag Ö, Çelebi-Saltik B. From Embryo to Adult: One Carbon Metabolism in Stem Cells. Curr Stem Cell Res Ther 2021; 16:175-188. [PMID: 32652922 DOI: 10.2174/1574888x15666200712191308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/16/2020] [Accepted: 04/23/2020] [Indexed: 11/22/2022]
Abstract
Stem cells are undifferentiated cells with self-renewal property and varying differentiation potential that allow the regeneration of tissue cells of an organism throughout adult life beginning from embryonic development. Through the asymmetric cell divisions, each stem cell replicates itself and produces an offspring identical with the mother cell, and a daughter cell that possesses the characteristics of a progenitor cell and commits to a specific lineage to differentiate into tissue cells to maintain homeostasis. To maintain a pool of stem cells to ensure tissue regeneration and homeostasis, it is important to regulate the metabolic functioning of stem cells, progenitor cells and adult tissue stem cells that will meet their internal and external needs. Upon fertilization, the zygote transforms metabolic reprogramming while implantation, embryonic development, organogenesis processes and after birth through adult life. Metabolism in stem cells is a concept that is relatively new to be enlightened. There are no adequate and comprehensive in vitro studies on the comparative analysis of the effects of one-carbon (1-C) metabolism on fetal and adult stem cells compared to embryonic and cancer stem cells' studies that have been reported recently. Since 1-C metabolism is linking parental environmental/ dietary factors and fetal development, investigating the epigenetic, genetic, metabolic and developmental effects on adult period is necessary. Several mutations and abnormalities in 1-C metabolism have been noted in disease changing from diabetes, cancer, pregnancy-related outcomes such as pre-eclampsia, spontaneous abortion, placental abruption, premature delivery, and cardiovascular diseases. In this review, the effects of 1-C metabolism, mainly the methionine and folate metabolism, in stem cells that exist in different developmental stages will be discussed.
Collapse
Affiliation(s)
- Özlem Altundag
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, 06100, Sihhiye, Ankara, Turkey
| | - Betül Çelebi-Saltik
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, 06100, Sihhiye, Ankara, Turkey
| |
Collapse
|
128
|
Wang Q, Xiong Y, Zhang S, Sui Y, Yu C, Liu P, Li H, Guo W, Gao Y, Przepiorski A, Davidson AJ, Guo M, Zhang X. The Dynamics of Metabolic Characterization in iPSC-Derived Kidney Organoid Differentiation via a Comparative Omics Approach. Front Genet 2021; 12:632810. [PMID: 33643392 PMCID: PMC7902935 DOI: 10.3389/fgene.2021.632810] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/20/2021] [Indexed: 12/22/2022] Open
Abstract
The use of differentiating human induced pluripotent stem cells (hiPSCs) in mini-tissue organoids provides an invaluable resource for regenerative medicine applications, particularly in the field of disease modeling. However, most studies using a kidney organoid model, focused solely on the transcriptomics and did not explore mechanisms of regulating kidney organoids related to metabolic effects and maturational phenotype. Here, we applied metabolomics coupled with transcriptomics to investigate the metabolic dynamics and function during kidney organoid differentiation. Not only did we validate the dominant metabolic alteration from glycolysis to oxidative phosphorylation in the iPSC differentiation process but we also showed that glycine, serine, and threonine metabolism had a regulatory role during kidney organoid formation and lineage maturation. Notably, serine had a role in regulating S-adenosylmethionine (SAM) to facilitate kidney organoid formation by altering DNA methylation. Our data revealed that analysis of metabolic characterization broadens our ability to understand phenotype regulation. The utilization of this comparative omics approach, in studying kidney organoid formation, can aid in deciphering unique knowledge about the biological and physiological processes involved in organoid-based disease modeling or drug screening.
Collapse
Affiliation(s)
- Qizheng Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yucui Xiong
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Sheng Zhang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Yufei Sui
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
| | - Cunlai Yu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Peng Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
| | - Heying Li
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
| | - Wenjing Guo
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yubo Gao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Aneta Przepiorski
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Alan J. Davidson
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Meijin Guo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Xiao Zhang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| |
Collapse
|
129
|
Abstract
Human lifespan is now longer than ever and, as a result, modern society is getting older. Despite that, the detailed mechanisms behind the ageing process and its impact on various tissues and organs remain obscure. In general, changes in DNA, RNA and protein structure throughout life impair their function. Haematopoietic ageing refers to the age-related changes affecting a haematopoietic system. Aged blood cells display different functional aberrations depending on their cell type, which might lead to the development of haematologic disorders, including leukaemias, anaemia or declining immunity. In contrast to traditional bulk assays, which are not suitable to dissect cell-to-cell variation, single-cell-level analysis provides unprecedented insight into the dynamics of age-associated changes in blood. In this Review, we summarise recent studies that dissect haematopoietic ageing at the single-cell level. We discuss what cellular changes occur during haematopoietic ageing at the genomic, transcriptomic, epigenomic and metabolomic level, and provide an overview of the benefits of investigating those changes with single-cell precision. We conclude by considering the potential clinical applications of single-cell techniques in geriatric haematology, focusing on the impact on haematopoietic stem cell transplantation in the elderly and infection studies, including recent COVID-19 research.
Collapse
Affiliation(s)
- Paulina M Strzelecka
- Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
- German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
| | - Frederik Damm
- Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
- German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
- Berlin Institute of Health, 10178 Berlin, Germany
| |
Collapse
|
130
|
Osawa S, Kato H, Maeda Y, Takakura H, Ogasawara J, Izawa T. Metabolomic Profiles in Adipocytes Differentiated from Adipose-Derived Stem Cells Following Exercise Training or High-Fat Diet. Int J Mol Sci 2021; 22:ijms22020966. [PMID: 33478060 PMCID: PMC7835847 DOI: 10.3390/ijms22020966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 01/10/2021] [Indexed: 11/16/2022] Open
Abstract
Controlling the differentiation potential of adipose-derived stem cells (ADSCs) is attracting attention as a new strategy for the prevention and treatment of obesity. Here, we aimed to observe the effect of exercise training (TR) and high-fat diet (HFD) on the metabolic profiles of ADSCs-derived adipocytes. The rats were divided into four groups: normal diet (ND)-fed control (ND-SED), ND-fed TR (ND-TR), HFD-fed control (HFD-SED), and HFD-fed TR (HFD-TR). After 9 weeks of intervention, ADSCs of epididymal and inguinal adipose tissues were differentiated into adipocytes. In the metabolome analysis of adipocytes after isoproterenol stimulation, 116 metabolites were detected. The principal component analysis demonstrated that ADSCs-derived adipocytes segregated into four clusters in each fat pad. Amino acid accumulation was greater in epididymal ADSCs-derived adipocytes of ND-TR and HFD-TR, but lower in inguinal ADSCs-derived adipocytes of ND-TR, than in the respective controls. HFD accumulated several metabolites including amino acids in inguinal ADSCs-derived adipocytes and more other metabolites in epididymal ones. Kyoto Encyclopedia of Genes and Genomes enrichment analysis revealed that TR mainly affected the pathways related to amino acid metabolism, except in inguinal ADSCs-derived adipocytes of HFD-TR rats. These findings provide a new way to understand the mechanisms underlying possible changes in the differentiation of ADSCs due to TR or HFD.
Collapse
Affiliation(s)
- Seita Osawa
- Graduate School of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyoto 610-0394, Japan
| | - Hisashi Kato
- Graduate School of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyoto 610-0394, Japan
- Organisation for Research Initiatives and Development, Doshisha University, 1-3 Tatara-Miyakodani, Kyoto 610-0394, Japan
| | - Yuki Maeda
- Graduate School of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyoto 610-0394, Japan
| | - Hisashi Takakura
- Graduate School of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyoto 610-0394, Japan
| | - Junetsu Ogasawara
- Division of Health Science, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Hokkaido 078-8510, Japan
| | - Tetsuya Izawa
- Graduate School of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyoto 610-0394, Japan
| |
Collapse
|
131
|
Weidinger A, Poženel L, Wolbank S, Banerjee A. Sub-Regional Differences of the Human Amniotic Membrane and Their Potential Impact on Tissue Regeneration Application. Front Bioeng Biotechnol 2021; 8:613804. [PMID: 33520964 PMCID: PMC7839410 DOI: 10.3389/fbioe.2020.613804] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/07/2020] [Indexed: 01/08/2023] Open
Abstract
For more than 100 years, the human amniotic membrane (hAM) has been used in multiple tissue regeneration applications. The hAM consists of cells with stem cell characteristics and a rich layer of extracellular matrix. Undoubtedly, the hAM with viable cells has remarkable properties such as the differentiation potential into all three germ layers, immuno-modulatory, and anti-fibrotic properties. At first sight, the hAM seems to be one structural entity. However, by integrating its anatomical location, the hAM can be divided into placental, reflected, and umbilical amniotic membrane. Recent studies show that cells of these amniotic sub-regions differ considerably in their properties such as morphology, structure, and content/release of certain bioactive factors. The aim of this review is to summarize these findings and discuss the relevance of these different properties for tissue regeneration. In summary, reflected amnion seems to be more immuno-modulatory and could have a higher reprogramming efficiency, whereas placental amnion seems to be pro-inflammatory, pro-angiogenic, with higher proliferation and differentiation capacity (e.g., chondrogenic and osteogenic), and could be more suitable for certain graft constructions. Therefore, we suggest that the respective hAM sub-region should be selected in consideration of its desired outcome. This will help to optimize and fine-tune the clinical application of the hAM.
Collapse
Affiliation(s)
- Adelheid Weidinger
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Austria
| | - Laura Poženel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Austria
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Austria
| | - Asmita Banerjee
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Austria
| |
Collapse
|
132
|
Effect of Jakyakgamcho-Tang Extracts on H 2O 2-Induced C2C12 Myoblasts. Molecules 2021; 26:molecules26010215. [PMID: 33406609 PMCID: PMC7795328 DOI: 10.3390/molecules26010215] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress is a major contributor to muscle aging and loss of muscle tissue. Jakyakgamcho-tang (JGT) has been used in traditional Eastern medicine to treat muscle pain. Here, we compared the total phenolic and flavonoid contents in 30% ethanol and water extracts of JGT and tested the preventive effects against oxidative stress (hydrogen peroxide)-induced cell death in murine C2C12 skeletal muscle cells. The total phenolic content and total flavonoid content in 30% ethanol extracts of JGT were higher than those of water extracts of JGT. Ethanol extracts of JGT (JGT-E) had stronger antioxidant activities of 2,2′-azino-bis-3-ethylbenzothiazoline-6-sulfonic acid (ABTS) and 2,2′-diphenyl-1-picrylhydrazyl-scavenging activity (DPPH) than water extracts of JGT (JGT-W). JGT-E contained 19–53% (1.8 to 4.9-fold) more active compounds (i.e., albiflorin, liquiritin, pentagalloylglucose, isoliquiritin apioside, isoliquiritin, liquiritigenin, and glycyrrhizin) than JGT-W. The ethanol extracts of JGT inhibited hydrogen peroxide-induced cell death and intracellular reactive oxygen species generation more effectively than the water extract of JGT in a dose-dependent manner. For the first time, these results suggest that ethanol extract of JGT is relatively more efficacious at protecting against oxidative stress-induced muscle cell death.
Collapse
|
133
|
Dunn AF, Catterton MA, Dixon DD, Pompano RR. Spatially resolved measurement of dynamic glucose uptake in live ex vivo tissues. Anal Chim Acta 2021; 1141:47-56. [PMID: 33248661 PMCID: PMC7701360 DOI: 10.1016/j.aca.2020.10.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 12/14/2022]
Abstract
Highly proliferative cells depend heavily on glycolysis as a source of energy and biological precursor molecules, and glucose uptake is a useful readout of this aspect of metabolic activity. Glucose uptake is commonly quantified by using flow cytometry for cell cultures and positron emission tomography for organs in vivo. However, methods to detect spatiotemporally resolved glucose uptake in intact tissues are far more limited, particularly those that can quantify changes in uptake over time in specific tissue regions and cell types. Using lymph node metabolism as a case study, we developed an optimized method to detect dynamic and spatially resolved glucose uptake in living tissue by combining ex vivo tissue slice culture with a fluorescent glucose analogue. Live slices of murine lymph node were treated with the glucose analogue 2-[N-(7-nitrobenz-2-oxa-1,3-dia-xol-4-yl)amino]-2-deoxyglucose (2-NBDG). Incubation parameters were optimized to differentiate glucose uptake in activated versus naïve lymphocytes. Regional glucose uptake could be imaged at both the tissue level, by widefield microscopy, and at the cellular level, by confocal microscopy. Furthermore, the glucose assay was readily multiplexed with live immunofluorescence labelling to generate maps of 2-NBDG uptake across tissue regions, revealing highest uptake in T cell-dense regions. The signal was predominantly intracellular and localized to lymphocytes rather than stromal cells. Finally, we demonstrated that the assay was repeatable in the same slices, and imaged the dynamic distribution of glucose uptake in response to ex vivo T cell stimulation for the first time. We anticipate that this method will serve as a broadly applicable, user-friendly platform to quantify dynamic metabolic activities in complex tissue microenvironments.
Collapse
Affiliation(s)
- Austin F Dunn
- Department of Chemistry, University of Virginia, PO BOX 400319, Charlottesville, VA, 22904, USA
| | - Megan A Catterton
- Department of Chemistry, University of Virginia, PO BOX 400319, Charlottesville, VA, 22904, USA
| | - Drake D Dixon
- Department of Chemistry, University of Virginia, PO BOX 400319, Charlottesville, VA, 22904, USA
| | - Rebecca R Pompano
- Department of Chemistry, Carter Immunology Center, University of Virginia, PO BOX 400319, Charlottesville, VA, 22904, USA.
| |
Collapse
|
134
|
Sheikh MA, Emerald BS, Ansari SA. Stem cell fate determination through protein O-GlcNAcylation. J Biol Chem 2021; 296:100035. [PMID: 33154167 PMCID: PMC7948975 DOI: 10.1074/jbc.rev120.014915] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 11/05/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
Embryonic and adult stem cells possess the capability of self-renewal and lineage-specific differentiation. The intricate balance between self-renewal and differentiation is governed by developmental signals and cell-type-specific gene regulatory mechanisms. A perturbed intra/extracellular environment during lineage specification could affect stem cell fate decisions resulting in pathology. Growing evidence demonstrates that metabolic pathways govern epigenetic regulation of gene expression during stem cell fate commitment through the utilization of metabolic intermediates or end products of metabolic pathways as substrates for enzymatic histone/DNA modifications. UDP-GlcNAc is one such metabolite that acts as a substrate for enzymatic mono-glycosylation of various nuclear, cytosolic, and mitochondrial proteins on serine/threonine amino acid residues, a process termed protein O-GlcNAcylation. The levels of GlcNAc inside the cells depend on the nutrient availability, especially glucose. Thus, this metabolic sensor could modulate gene expression through O-GlcNAc modification of histones or other proteins in response to metabolic fluctuations. Herein, we review evidence demonstrating how stem cells couple metabolic inputs to gene regulatory pathways through O-GlcNAc-mediated epigenetic/transcriptional regulatory mechanisms to govern self-renewal and lineage-specific differentiation programs. This review will serve as a primer for researchers seeking to better understand how O-GlcNAc influences stemness and may catalyze the discovery of new stem-cell-based therapeutic approaches.
Collapse
Affiliation(s)
- Muhammad Abid Sheikh
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE; Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
| | - Suraiya Anjum Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE; Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE.
| |
Collapse
|
135
|
Biological characteristics of aging in human acute myeloid leukemia cells: the possible importance of aldehyde dehydrogenase, the cytoskeleton and altered transcriptional regulation. Aging (Albany NY) 2020; 12:24734-24777. [PMID: 33349623 PMCID: PMC7803495 DOI: 10.18632/aging.202361] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/20/2020] [Indexed: 12/19/2022]
Abstract
Patients with acute myeloid leukemia (AML) have a median age of 65-70 years at diagnosis. Elderly patients have more chemoresistant disease, and this is partly due to decreased frequencies of favorable and increased frequencies of adverse genetic abnormalities. However, aging-dependent differences may also contribute. We therefore compared AML cell proteomic and phosphoproteomic profiles for (i) elderly low-risk and younger low-risk patients with favorable genetic abnormalities; and (ii) high-risk patients with adverse genetic abnormalities and a higher median age against all low-risk patients with lower median age. Elderly low-risk and younger low-risk patients showed mainly phosphoproteomic differences especially involving transcriptional regulators and cytoskeleton. When comparing high-risk and low-risk patients both proteomic and phosphoproteomic studies showed differences involving cytoskeleton and immunoregulation but also transcriptional regulation and cell division. The age-associated prognostic impact of cyclin-dependent kinases was dependent on the cellular context. The protein level of the adverse prognostic biomarker mitochondrial aldehyde dehydrogenase (ALDH2) showed a similar significant upregulation both in elderly low-risk and elderly high-risk patients. Our results suggest that molecular mechanisms associated with cellular aging influence chemoresistance of AML cells, and especially the cytoskeleton function may then influence cellular hallmarks of aging, e.g. mitosis, polarity, intracellular transport and adhesion.
Collapse
|
136
|
Spannl S, Buhl T, Nellas I, Zeidan SA, Iyer KV, Khaliullina H, Schultz C, Nadler A, Dye NA, Eaton S. Glycolysis regulates Hedgehog signalling via the plasma membrane potential. EMBO J 2020; 39:e101767. [PMID: 33021744 PMCID: PMC7604625 DOI: 10.15252/embj.2019101767] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 08/19/2020] [Accepted: 08/25/2020] [Indexed: 01/04/2023] Open
Abstract
Changes in cell metabolism and plasma membrane potential have been linked to shifts between tissue growth and differentiation, and to developmental patterning. How such changes mediate these effects is poorly understood. Here, we use the developing wing of Drosophila to investigate the interplay between cell metabolism and a key developmental regulator-the Hedgehog (Hh) signalling pathway. We show that reducing glycolysis both lowers steady-state levels of ATP and stabilizes Smoothened (Smo), the 7-pass transmembrane protein that transduces the Hh signal. As a result, the transcription factor Cubitus interruptus accumulates in its full-length, transcription activating form. We show that glycolysis is required to maintain the plasma membrane potential and that plasma membrane depolarization blocks cellular uptake of N-acylethanolamides-lipoprotein-borne Hh pathway inhibitors required for Smo destabilization. Similarly, pharmacological inhibition of glycolysis in mammalian cells induces ciliary translocation of Smo-a key step in pathway activation-in the absence of Hh. Thus, changes in cell metabolism alter Hh signalling through their effects on plasma membrane potential.
Collapse
Affiliation(s)
- Stephanie Spannl
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Present address:
Department of BiochemistryFaculty of MedicineUniversity of TorontoTorontoONCanada
| | - Tomasz Buhl
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Biotechnologisches ZentrumTechnische Universität DresdenDresdenGermany
| | - Ioannis Nellas
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Biotechnologisches ZentrumTechnische Universität DresdenDresdenGermany
| | - Salma A Zeidan
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Biotechnologisches ZentrumTechnische Universität DresdenDresdenGermany
| | - K Venkatesan Iyer
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Max Planck Institute for the Physics of Complex SystemsDresdenGermany
| | - Helena Khaliullina
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Present address:
Department of PhysiologyDevelopment and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Carsten Schultz
- Cell Biology and Biophysics UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
- Department of Chemical Physiology and BiochemistryOregon Health and Science UniversityPortlandORUSA
| | - André Nadler
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Natalie A Dye
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Suzanne Eaton
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Biotechnologisches ZentrumTechnische Universität DresdenDresdenGermany
- Center for Systems Biology DresdenDresdenGermany
| |
Collapse
|
137
|
Kim CS, Ding X, Allmeroth K, Biggs LC, Kolenc OI, L'Hoest N, Chacón-Martínez CA, Edlich-Muth C, Giavalisco P, Quinn KP, Denzel MS, Eming SA, Wickström SA. Glutamine Metabolism Controls Stem Cell Fate Reversibility and Long-Term Maintenance in the Hair Follicle. Cell Metab 2020; 32:629-642.e8. [PMID: 32905798 DOI: 10.1016/j.cmet.2020.08.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 06/30/2020] [Accepted: 08/13/2020] [Indexed: 12/12/2022]
Abstract
Stem cells reside in specialized niches that are critical for their function. Upon activation, hair follicle stem cells (HFSCs) exit their niche to generate the outer root sheath (ORS), but a subset of ORS progeny returns to the niche to resume an SC state. Mechanisms of this fate reversibility are unclear. We show that the ability of ORS cells to return to the SC state requires suppression of a metabolic switch from glycolysis to oxidative phosphorylation and glutamine metabolism that occurs during early HFSC lineage progression. HFSC fate reversibility and glutamine metabolism are regulated by the mammalian target of rapamycin complex 2 (mTORC2)-Akt signaling axis within the niche. Deletion of mTORC2 results in a failure to re-establish the HFSC niche, defective hair follicle regeneration, and compromised long-term maintenance of HFSCs. These findings highlight the importance of spatiotemporal control of SC metabolic states in organ homeostasis.
Collapse
Affiliation(s)
- Christine S Kim
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Xiaolei Ding
- Department of Dermatology, University of Cologne, Cologne, Germany
| | - Kira Allmeroth
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Leah C Biggs
- Helsinki Institute of Life Science, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland; Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland; Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olivia I Kolenc
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Nina L'Hoest
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Carlos Andrés Chacón-Martínez
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | | | | | - Kyle P Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Martin S Denzel
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Sabine A Eming
- Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany; Department of Dermatology, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany.
| | - Sara A Wickström
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany; Helsinki Institute of Life Science, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland; Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland; Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
138
|
Sharaireh AM, Fitzpatrick LM, Ward CM, McKay TR, Unwin RD. Epithelial cadherin regulates transition between the naïve and primed pluripotent states in mouse embryonic stem cells. Stem Cells 2020; 38:1292-1306. [PMID: 32621788 DOI: 10.1002/stem.3249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022]
Abstract
Inhibition of E-cad in mouse embryonic stem cells (mESCs) leads to a switch from LIF-BMP to Activin/Nodal-dependent pluripotency, consistent with transition from a naïve to primed pluripotent phenotype. We have used both genetic ablation and steric inhibition of E-cad function in mESCs to assess alterations to phenotype using quantitative mass spectrometry analysis, network models, and functional assays. Proteomic analyses revealed that one third of detected proteins were altered in E-cad null mESCs (Ecad-/- mESCs) compared to wild type (624 proteins were downregulated and 705 were proteins upregulated). Network pathway analysis and subsequent cellular flux assays confirmed a metabolic shift from oxidative phosphorylation (OXPHOS) to aerobic glycolysis, specifically through mitochondrial complex III downregulation and hypoxia inducible factor 1a target upregulation. Central to this was the transcriptional coactivator EP300. E-cad is a well-known tumor suppressor, its downregulation during cancer initiation and metastasis can be linked to the metabolic switch known as Warburg effect. This study highlights a phenomena found in both primed pluripotent state and cancer stemness and links it to loss of E-cad. Data are available via ProteomeXchange with identifier PXD012679.
Collapse
Affiliation(s)
- Aseel M Sharaireh
- Division of Dentistry, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Stem Cell Group, Centre for Bioscience, Manchester Metropolitan University, Manchester, UK
| | - Lorna M Fitzpatrick
- Stem Cell Group, Centre for Bioscience, Manchester Metropolitan University, Manchester, UK
| | - Chris M Ward
- Division of Dentistry, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Tristan R McKay
- Stem Cell Group, Centre for Bioscience, Manchester Metropolitan University, Manchester, UK
| | - Richard D Unwin
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Stoller Biomarker Discovery Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
139
|
Affiliation(s)
- Ryohei Iwata
- Vlaams Instituut voor Biotechnologie-Center for Brain & Disease Research, 3000 Leuven, Belgium.,Department of Neurosciences, Katholieke Universiteit (KU) Leuven, 3000 Leuven, Belgium.,Leuven Brain Institute, 3000 Leuven, Belgium.,Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium.,Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), 1070 Brussels, Belgium.,ULB Neuroscience Institute, 1070 Brussels, Belgium
| | - Pierre Vanderhaeghen
- Vlaams Instituut voor Biotechnologie-Center for Brain & Disease Research, 3000 Leuven, Belgium. .,Department of Neurosciences, Katholieke Universiteit (KU) Leuven, 3000 Leuven, Belgium.,Leuven Brain Institute, 3000 Leuven, Belgium.,Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium.,Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), 1070 Brussels, Belgium.,ULB Neuroscience Institute, 1070 Brussels, Belgium
| |
Collapse
|
140
|
Epigenetic Regulation in Mesenchymal Stem Cell Aging and Differentiation and Osteoporosis. Stem Cells Int 2020; 2020:8836258. [PMID: 32963550 PMCID: PMC7501554 DOI: 10.1155/2020/8836258] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/17/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a reliable source for cell-based regenerative medicine owing to their multipotency and biological functions. However, aging-induced systemic homeostasis disorders in vivo and cell culture passaging in vitro induce a functional decline of MSCs, switching MSCs to a senescent status with impaired self-renewal capacity and biased differentiation tendency. MSC functional decline accounts for the pathogenesis of many diseases and, more importantly, limits the large-scale applications of MSCs in regenerative medicine. Growing evidence implies that epigenetic mechanisms are a critical regulator of the differentiation programs for cell fate and are subject to changes during aging. Thus, we here review epigenetic dysregulations that contribute to MSC aging and osteoporosis. Comprehending detailed epigenetic mechanisms could provide us with a novel horizon for dissecting MSC-related pathogenesis and further optimizing MSC-mediated regenerative therapies.
Collapse
|
141
|
Gaggi G, Di Credico A, Izzicupo P, Alviano F, Di Mauro M, Di Baldassarre A, Ghinassi B. Human Mesenchymal Stromal Cells Unveil an Unexpected Differentiation Potential toward the Dopaminergic Neuronal Lineage. Int J Mol Sci 2020; 21:ijms21186589. [PMID: 32916865 PMCID: PMC7555006 DOI: 10.3390/ijms21186589] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 08/29/2020] [Accepted: 09/08/2020] [Indexed: 12/12/2022] Open
Abstract
Degeneration of dopaminergic neurons represents the cause of many neurodegenerative diseases, with increasing incidence worldwide. The replacement of dead cells with new healthy ones may represent an appealing therapeutic approach to these pathologies, but currently, only pluripotent stem cells can generate dopaminergic neurons with high efficiency. However, with the use of these cells arises safety and/or ethical issues. Human mesenchymal stromal cells (hFM-MSCs) are perinatal stem cells that can be easily isolated from the amniochorionic membrane after delivery. Generally considered multipotent, their real differentiative potential is not completely elucidated. The aim of this study was to analyze their stemness characteristics and to evaluate whether they may overcome their mesenchymal fate, generating dopaminergic neurons. We demonstrated that hFM-MSCs expressed embryonal genes OCT4, NANOG, SOX2, KLF4, OVOL1, and ESG1, suggesting they have some features of pluripotency. Moreover, hFM-MSCs that underwent a dopaminergic differentiation protocol gradually increased the transcription of dopaminergic markers LMX1b, NURR1, PITX3, and DAT. We finally obtained a homogeneous population of cells resembling the morphology of primary midbrain dopaminergic neurons that expressed the functional dopaminergic markers TH, DAT, and Nurr1. In conclusion, our results suggested that hFM-MSCs retain the expression of pluripotency genes and are able to differentiate not only into mesodermal cells, but also into neuroectodermal dopaminergic neuron-like cells.
Collapse
Affiliation(s)
- Giulia Gaggi
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (B.G.)
| | - Andrea Di Credico
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (B.G.)
| | - Pascal Izzicupo
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (B.G.)
| | - Francesco Alviano
- Department of Experimental Diagnostic and Speciality Medicine, Unit of Histology, Embriology and Applied Biology, University of Bologna, 40126 Bologna, Italy;
| | - Michele Di Mauro
- Cardio-Thoracic Surgery Unit, Heart and Vascular Centre, Maastricht University Medical Centre (MUMC), Cardiovascular Research Institute Maastricht (CARIM), 6202 Maastricht, The Netherlands;
| | - Angela Di Baldassarre
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (B.G.)
- Correspondence:
| | - Barbara Ghinassi
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (B.G.)
| |
Collapse
|
142
|
Carvalho-Santos Z, Cardoso-Figueiredo R, Elias AP, Tastekin I, Baltazar C, Ribeiro C. Cellular metabolic reprogramming controls sugar appetite in Drosophila. Nat Metab 2020; 2:958-973. [PMID: 32868922 DOI: 10.1038/s42255-020-0266-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 07/17/2020] [Indexed: 12/13/2022]
Abstract
Cellular metabolic reprogramming is an important mechanism by which cells rewire their metabolism to promote proliferation and cell growth. This process has been mostly studied in the context of tumorigenesis, but less is known about its relevance for nonpathological processes and how it affects whole-animal physiology. Here, we show that metabolic reprogramming in Drosophila female germline cells affects nutrient preferences of animals. Egg production depends on the upregulation of the activity of the pentose phosphate pathway in the germline, which also specifically increases the animal's appetite for sugar, the key nutrient fuelling this metabolic pathway. We provide functional evidence that the germline alters sugar appetite by regulating the expression of the fat-body-secreted satiety factor Fit. Our findings demonstrate that the cellular metabolic program of a small set of cells is able to increase the animal's preference for specific nutrients through inter-organ communication to promote specific metabolic and cellular outcomes.
Collapse
Affiliation(s)
- Zita Carvalho-Santos
- Behavior and Metabolism Laboratory, Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| | - Rita Cardoso-Figueiredo
- Behavior and Metabolism Laboratory, Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Ana Paula Elias
- Behavior and Metabolism Laboratory, Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Ibrahim Tastekin
- Behavior and Metabolism Laboratory, Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Célia Baltazar
- Behavior and Metabolism Laboratory, Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Carlos Ribeiro
- Behavior and Metabolism Laboratory, Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| |
Collapse
|
143
|
Riveiro AR, Brickman JM. From pluripotency to totipotency: an experimentalist's guide to cellular potency. Development 2020; 147:147/16/dev189845. [PMID: 32847824 DOI: 10.1242/dev.189845] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/16/2020] [Indexed: 12/12/2022]
Abstract
Embryonic stem cells (ESCs) are derived from the pre-implantation mammalian blastocyst. At this point in time, the newly formed embryo is concerned with the generation and expansion of both the embryonic lineages required to build the embryo and the extra-embryonic lineages that support development. When used in grafting experiments, embryonic cells from early developmental stages can contribute to both embryonic and extra-embryonic lineages, but it is generally accepted that ESCs can give rise to only embryonic lineages. As a result, they are referred to as pluripotent, rather than totipotent. Here, we consider the experimental potential of various ESC populations and a number of recently identified in vitro culture systems producing states beyond pluripotency and reminiscent of those observed during pre-implantation development. We also consider the nature of totipotency and the extent to which cell populations in these culture systems exhibit this property.
Collapse
Affiliation(s)
- Alba Redó Riveiro
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen N, Denmark
| | - Joshua Mark Brickman
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen N, Denmark
| |
Collapse
|
144
|
Chen W, Zhang Z, Chang C, Yang Z, Wang P, Fu H, Wei X, Chen E, Tan S, Huang W, Sun L, Ni T, Yang Y, Wang Y. A bioenergetic shift is required for spermatogonial differentiation. Cell Discov 2020; 6:56. [PMID: 32864161 PMCID: PMC7431567 DOI: 10.1038/s41421-020-0183-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/22/2020] [Indexed: 12/22/2022] Open
Abstract
A bioenergetic balance between glycolysis and mitochondrial respiration is particularly important for stem cell fate specification. It however remains to be determined whether undifferentiated spermatogonia switch their preference for bioenergy production during differentiation. In this study, we found that ATP generation in spermatogonia was gradually increased upon retinoic acid (RA)-induced differentiation. To accommodate this elevated energy demand, RA signaling concomitantly switched ATP production in spermatogonia from glycolysis to mitochondrial respiration, accompanied by increased levels of reactive oxygen species. Disrupting mitochondrial respiration significantly blocked spermatogonial differentiation. Inhibition of glucose conversion to glucose-6-phosphate or pentose phosphate pathway also repressed the formation of c-Kit+ differentiating germ cells, suggesting that metabolites produced from glycolysis are required for spermatogonial differentiation. We further demonstrated that the expression levels of several metabolic regulators and enzymes were significantly altered upon RA-induced differentiation, with both RNA-seq and quantitative proteomic analyses. Taken together, our data unveil a critically regulated bioenergetic balance between glycolysis and mitochondrial respiration that is required for spermatogonial proliferation and differentiation.
Collapse
Affiliation(s)
- Wei Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241 China
| | - Zhaoran Zhang
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824 USA
| | - Chingwen Chang
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824 USA
| | - Zhichang Yang
- Department of Chemistry, College of Natural Science, Michigan State University, East Lansing, MI 48824 USA
| | - Pengxiang Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241 China
| | - Haihui Fu
- State Key Laboratory of Genetic Engineering & MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Xiao Wei
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241 China
| | - Eric Chen
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824 USA
| | - Suxu Tan
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824 USA
| | - Wen Huang
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824 USA
| | - Liangliang Sun
- Department of Chemistry, College of Natural Science, Michigan State University, East Lansing, MI 48824 USA
| | - Ting Ni
- State Key Laboratory of Genetic Engineering & MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Yi Yang
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237 China
| | - Yuan Wang
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824 USA
| |
Collapse
|
145
|
Mendes BB, Gómez-Florit M, Araújo AC, Prada J, Babo PS, Domingues RMA, Reis RL, Gomes ME. Intrinsically Bioactive Cryogels Based on Platelet Lysate Nanocomposites for Hemostasis Applications. Biomacromolecules 2020; 21:3678-3692. [PMID: 32786530 DOI: 10.1021/acs.biomac.0c00787] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The currently used hemostatic agents are highly effective in stopping hemorrhages but have a limited role in the modulation of the wound-healing environment. Herein, we propose an intrinsically bioactive hemostatic cryogel based on platelet lysate (PL) and aldehyde-functionalized cellulose nanocrystals (a-CNCs). PL has attracted great attention as an inexpensive milieu of therapeutically relevant proteins; however, its application as a hemostatic agent exhibits serious constraints (e.g., structural integrity and short shelf-life). The incorporation of a-CNCs reinforced the low-strength PL matrix by covalent cross-linking its amine groups that exhibit an elastic interconnected porous network after full cryogelation. Upon blood immersion, the PL-CNC cryogels absorbed higher volumes of blood at a faster rate than commercial hemostatic porcine gelatin sponges. Simultaneously, the cryogels released biomolecules that increased stem cell proliferation, metabolic activity, and migration as well as downregulated the expression of markers of the fibrinolytic process. In an in vivo liver defect model, PL-CNC cryogels showed similar hemostatic performance in comparison with gelatin sponges and normal material-induced tissue response upon subcutaneous implantation. Overall, owing to their structure and bioactive composition, the proposed PL-CNC cryogels provide an alternative off-the-shelf hemostatic and antibacterial biomaterial with the potential to deliver therapeutically relevant proteins in situ.
Collapse
Affiliation(s)
- Bárbara B Mendes
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Avepark, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| | - Manuel Gómez-Florit
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Avepark, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| | - Ana C Araújo
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Avepark, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| | - Justina Prada
- UTAD, CECAV and Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, 5001-801 Vila Real, Portugal
| | - Pedro S Babo
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Avepark, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| | - Rui M A Domingues
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Avepark, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Avepark, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| | - Manuela E Gomes
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Avepark, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| |
Collapse
|
146
|
Lai X, Li Q, Wu F, Lin J, Chen J, Zheng H, Guo L. Epithelial-Mesenchymal Transition and Metabolic Switching in Cancer: Lessons From Somatic Cell Reprogramming. Front Cell Dev Biol 2020; 8:760. [PMID: 32850862 PMCID: PMC7423833 DOI: 10.3389/fcell.2020.00760] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) and its critical roles during cancer progression have long been recognized and extensively reviewed. Recent studies on the generation of induced pluripotent stem cells (iPSCs) have established the connections among EMT, energy metabolism, DNA methylation, and histone modification. Since energy metabolism, DNA methylation, and histone modification are important for cancer development and there are common characteristics between cancer cells and stem cells, it is reasonable to identify mechanisms that have been established during both reprogramming and cancer progression. In the current review, we start from a brief review on EMT and related processes during cancer progression, and then switch to the EMT during somatic cell reprogramming. We summarize the connection between EMT and metabolic switch during reprogramming, and further review the involvements of DNA methylation and cell proliferation. The connections between EMT and mesenchymal-epithelial transition (MET) and cellular aspects including DNA methylation, histone modification and energy metabolism may provide potential new targets for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Xiaowei Lai
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Qian Li
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Fang Wu
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Jiechun Lin
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Jiekai Chen
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Hui Zheng
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Lin Guo
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| |
Collapse
|
147
|
Chan CH, Wu CY, Dubey NK, Wei HJ, Lu JH, Mao S, Liang J, Liang YH, Cheng HC, Deng WP. Modulating redox homeostasis and cellular reprogramming through inhibited methylenetetrahydrofolate dehydrogenase 2 enzymatic activities in lung cancer. Aging (Albany NY) 2020; 12:17930-17947. [PMID: 32759461 PMCID: PMC7585109 DOI: 10.18632/aging.103471] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 05/27/2020] [Indexed: 01/24/2023]
Abstract
Recent reports have indicated the role of highly expressed methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) enzyme in cancers, showing poor survival; however, detailed mechanistic insight of metabolic functions of MTHFD2 have not been well-defined. Therefore, we aimed to examine the metabolic functions and cellular reprograming potential of MTHFD2 in lung cancer (LCa). In this study, we initially confirmed the expression levels of MTHFD2 in LCa not only in tissue and OncomineTM database, but also at molecular levels. Further, we reprogrammed metabolic activities in these cells through MTHFD2 gene knockdown via lentiviral transduction, and assessed their viability, transformation and self-renewal ability. In vivo tumorigenicity was also evaluated in NOD/SCID mice. Results showed that MTHFD2 was highly expressed in stage-dependent LCa tissues as well in cell lines, A549, H1299 and H441. Cellular viability, transformation and self-renewal abilities were significantly inhibited in MTHFD2-knockdown LCa cell lines. These cells also showed suppressed tumor-initiating ability and reduced tumor size compared to vector controls. Under low oxygen tension, MTHFD2-knockdown groups showed no significant increase in sphere formation, and hence the stemness. Conclusively, the suppressed levels of MTHFD2 is essential for cellular metabolic reprogramming leading to inhibited LCa growth and tumor aggressiveness.
Collapse
Affiliation(s)
- Chun-Hao Chan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan,Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chia-Yu Wu
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Taipei Medical University Hospital, Taipei 11031, Taiwan,School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Navneet Kumar Dubey
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan,Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Hong-Jian Wei
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Jui-Hua Lu
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan,Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Samantha Mao
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Joy Liang
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Hsuan Liang
- Department of Materials Science and Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Hsin-Chung Cheng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan,Department of Dentistry, Taipei Medical University Hospital, Taipei 110131, Taiwan
| | - Win-Ping Deng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan,Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan,Graduate Institute of Basic Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan
| |
Collapse
|
148
|
Miao S, Zhao D, Wang X, Ni X, Fang X, Yu M, Ye L, Yang J, Wu H, Han X, Qu L, Li L, Lan F, Shen Z, Lei W, Zhao ZA, Hu S. Retinoic acid promotes metabolic maturation of human Embryonic Stem Cell-derived Cardiomyocytes. Theranostics 2020; 10:9686-9701. [PMID: 32863954 PMCID: PMC7449904 DOI: 10.7150/thno.44146] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 07/20/2020] [Indexed: 12/15/2022] Open
Abstract
Cardiomyocytes differentiated from human embryonic stem cells (hESCs) represent a promising cell source for heart repair, disease modeling and drug testing. However, improving the differentiation efficiency and maturation of hESC-derived cardiomyocytes (hESC-CMs) is still a major concern. Retinoic acid (RA) signaling plays multiple roles in heart development. However, the effects of RA on cardiomyocyte differentiation efficiency and maturation are still unknown. Methods: RA was added at different time intervals to identify the best treatment windows for cardiomyocyte differentiation and maturation. The efficiency of cardiomyocyte differentiation was detected by quantitative real-time PCR and flow cytometry. Cardiomyocytes maturation was detected by immunofluorescence staining, metabolic assays and patch clamp to verify structural, metabolic and electrophysiological maturation, respectively. RNA sequencing was used for splicing analysis. Results: We found that RA treatment at the lateral mesoderm stage (days 2-4) significantly improved cardiomyocyte differentiation, as evidenced by the upregulation of TNNT2, NKX2.5 and MYH6 on day 10 of differentiation. In addition, flow cytometry showed that the proportion of differentiated cardiomyocytes in the RA-treated group was significantly higher than that in control group. RA treatment on days 15-20 increased cardiomyocyte area, sarcomere length, multinucleation and mitochondrial copy number. RNA sequencing revealed RA promoted RNA isoform switch to the maturation-related form. Meanwhile, RA promoted electrophysiological maturation and calcium handling of hESC-CMs. Importantly, RA-treated cardiomyocytes showed decreased glycolysis and enhanced mitochondrial oxidative phosphorylation, with the increased utilization of fatty acid and exogenous pyruvate but not glutamine. Conclusion: Our data indicated that RA treatment at an early time window (days 2-4) promotes the efficiency of cardiomyocyte differentiation and that RA treatment post beating (days 15-20) promotes cardiomyocyte maturation. The biphasic effects of RA provide new insights for improving cardiomyocyte differentiation and quality.
Collapse
|
149
|
Xu Y, Zhang Y, García-Cañaveras JC, Guo L, Kan M, Yu S, Blair IA, Rabinowitz JD, Yang X. Chaperone-mediated autophagy regulates the pluripotency of embryonic stem cells. Science 2020; 369:397-403. [PMID: 32703873 PMCID: PMC7939502 DOI: 10.1126/science.abb4467] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 02/26/2020] [Accepted: 05/27/2020] [Indexed: 12/14/2022]
Abstract
Embryonic stem cells can propagate indefinitely in a pluripotent state, able to differentiate into all types of specialized cells when restored to the embryo. What sustains their pluripotency during propagation remains unclear. Here, we show that core pluripotency factors OCT4 and SOX2 suppress chaperone-mediated autophagy (CMA), a selective form of autophagy, until the initiation of differentiation. Low CMA activity promotes embryonic stem cell self-renewal, whereas its up-regulation enhances differentiation. CMA degrades isocitrate dehydrogenases IDH1 and IDH2 and reduces levels of intracellular α-ketoglutarate, an obligatory cofactor for various histone and DNA demethylases involved in pluripotency. These findings suggest that CMA mediates the effect of core pluripotency factors on metabolism, shaping the epigenetic landscape of stem cells and governing the balance between self-renewal and differentiation.
Collapse
Affiliation(s)
- Yi Xu
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yang Zhang
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Juan C García-Cañaveras
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ 08540, USA
| | - Lili Guo
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mengyuan Kan
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sixiang Yu
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ian A Blair
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ 08540, USA
| | - Xiaolu Yang
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
150
|
Tencerova M, Frost M, Figeac F, Nielsen TK, Ali D, Lauterlein JJL, Andersen TL, Haakonsson AK, Rauch A, Madsen JS, Ejersted C, Højlund K, Kassem M. Obesity-Associated Hypermetabolism and Accelerated Senescence of Bone Marrow Stromal Stem Cells Suggest a Potential Mechanism for Bone Fragility. Cell Rep 2020; 27:2050-2062.e6. [PMID: 31091445 DOI: 10.1016/j.celrep.2019.04.066] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/06/2019] [Accepted: 04/12/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity is associated with increased risk for fragility fractures. However, the cellular mechanisms are unknown. Using a translational approach combining RNA sequencing and cellular analyses, we investigated bone marrow stromal stem cells (BM-MSCs) of 54 men divided into lean, overweight, and obese groups on the basis of BMI. Compared with BM-MSCs obtained from lean, obese BM-MSCs exhibited a shift of molecular phenotype toward committed adipocytic progenitors and increased expression of metabolic genes involved in glycolytic and oxidoreductase activity. Interestingly, compared with paired samples of peripheral adipose tissue-derived stromal cells (AT-MSCs), insulin signaling of obese BM-MSCs was enhanced and accompanied by increased abundance of insulin receptor positive (IR+) and leptin receptor positive (LEPR+) cells in BM-MSC cultures. Their hyper-activated metabolic state was accompanied by an accelerated senescence phenotype. Our data provide a plausible explanation for the bone fragility in obesity caused by enhanced insulin signaling leading to accelerated metabolic senescence of BM-MSCs.
Collapse
Affiliation(s)
- Michaela Tencerova
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; OPEN, Odense Patient Data Explorative Network, Odense University Hospital, Odense, Denmark.
| | - Morten Frost
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; Steno Diabetes Center Odense, Odense University Hospital, 5000 Odense C, Denmark
| | - Florence Figeac
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Tina Kamilla Nielsen
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Dalia Ali
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Jens-Jacob Lindegaard Lauterlein
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Thomas Levin Andersen
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark; Department of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| | - Anders Kristian Haakonsson
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; OPEN, Odense Patient Data Explorative Network, Odense University Hospital, Odense, Denmark
| | - Alexander Rauch
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Jonna Skov Madsen
- Institute of Regional Health Science, University of Southern Denmark, 5000 Odense C, Denmark; Department of Biochemistry and Immunology, Lillebaelt Hospital, 7100 Vejle, Denmark
| | - Charlotte Ejersted
- Department of Endocrinology, Odense University Hospital, 5000 Odense C, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, 5000 Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Moustapha Kassem
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; Department of Cellular and Molecular Medicine, DanStem (Danish Stem Cell Center), Panum Institute, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|