47 results on '"Perdiguero E"'
Search Results
2. Stem Cells and Aging
- Author
-
Perdiguero, E., primary, García-Prat, L., additional, Sousa-Victor, P., additional, and Muñoz-Cánoves, P., additional
- Published
- 2016
- Full Text
- View/download PDF
3. Musculoskeletal senescence: a moving target ready to be eliminated
- Author
-
Baar, M.P. (Marjolein), Perdiguero, E. (Eusebio), Muñoz-Cánoves, P. (Pura), Keizer, P.L.J. (Peter) de, Baar, M.P. (Marjolein), Perdiguero, E. (Eusebio), Muñoz-Cánoves, P. (Pura), and Keizer, P.L.J. (Peter) de
- Abstract
Aging is the prime risk factor for the broad-based development of diseases. Frailty is a phenotypical hallmark of aging and is often used to assess whether the predicted benefits of a therapy outweigh the risks for older patients. Senescent cells form as a consequence of unresolved molecular damage and persistently secrete molecules that can impair tissue function. Recent evidence shows senescent cells can chronically interfere with stem cell function and drive aging of the musculoskeletal system. In addition, targeted apoptosis of senescent cells can restore tissue homeostasis in aged animals. Thus, targeting cellular senescence provides new therapeutic opportunities for the intervention of frailty-associated pathologies and could have pleiotropic health benefits.
- Published
- 2018
- Full Text
- View/download PDF
4. Musculoskeletal senescence: a moving target ready to be eliminated
- Author
-
Baar, Marjolein, Perdiguero, E, Munoz-Canoves, P, de Keizer, PLJ, Baar, Marjolein, Perdiguero, E, Munoz-Canoves, P, and de Keizer, PLJ
- Published
- 2018
5. Cripto shapes macrophage plasticity and restricts EndMT in injured and diseased skeletal muscle
- Author
-
Gennaro Andolfi, Ombretta Guardiola, Antonio L. Serrano, Francescopaolo Iavarone, Eusebio Perdiguero, Alessandra Scagliola, Pura Muñoz-Cánoves, Federica Esposito, Gabriella Minchiotti, Silvia Brunelli, Ministerio de Ciencia, Innovación y Universidades (España), European Research Council, European Commission, Fundación La Caixa, Iavarone, F, Guardiola, O, Scagliola, A, Andolfi, G, Esposito, F, Serrano, A, Perdiguero, E, Brunelli, S, Muñoz-Cánoves, P, and Minchiotti, G
- Subjects
Duchenne muscular dystrophy ,Immunology ,macrophage plasticity ,SMAD ,Biology ,Cripto ,Biochemistry ,Macrophage plasticity ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Skeletal muscle regeneration ,skeletal muscle regeneration ,Genetics ,medicine ,Macrophage ,Animals ,Molecular Biology of Disease ,Muscle, Skeletal ,Molecular Biology ,Endothelial‐to‐Mesenchymal Transition ,030304 developmental biology ,0303 health sciences ,Endothelial-to-Mesenchymal Transition ,Regeneration (biology) ,Macrophages ,Skeletal muscle ,Endothelial Cells ,Articles ,medicine.disease ,Phenotype ,cripto ,inflammation ,macrophages ,muscles ,regeneration ,Cell biology ,Muscular Dystrophy, Duchenne ,medicine.anatomical_structure ,Membrane protein ,Mice, Inbred mdx ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Macrophages are characterized by a high plasticity in response to changes in tissue microenvironment, which allows them to acquire different phenotypes and to exert essential functions in complex processes, such as tissue regeneration. Here, we report that the membrane protein Cripto plays a key role in shaping macrophage plasticity in skeletal muscle during regeneration and disease. Conditional deletion of Cripto in the myeloid lineage (CriptoMy‐LOF) perturbs MP plasticity in acutely injured muscle and in mouse models of Duchenne muscular dystrophy (mdx). Specifically, CriptoMy‐LOF macrophages infiltrate the muscle, but fail to properly expand as anti‐inflammatory CD206+ macrophages, which is due, at least in part, to aberrant activation of TGFβ/Smad signaling. This reduction in macrophage plasticity disturbs vascular remodeling by increasing Endothelial‐to‐Mesenchymal Transition (EndMT), reduces muscle regenerative potential, and leads to an exacerbation of the dystrophic phenotype. Thus, in muscle‐infiltrating macrophages, Cripto is required to promote the expansion of the CD206+ anti‐inflammatory macrophage type and to restrict the EndMT process, providing a direct functional link between this macrophage population and endothelial cells., The membrane protein Cripto is an extrinsic determinant of macrophage plasticity in skeletal muscle regeneration and disease. Cripto‐dependent modulation of macrophage phenotypes controls endothelial plasticity and contributes to proper muscle repair.
- Published
- 2020
- Full Text
- View/download PDF
6. Muscle aging and sarcopenia: The pathology, etiology, and most promising therapeutic targets.
- Author
-
Grima-Terrén M, Campanario S, Ramírez-Pardo I, Cisneros A, Hong X, Perdiguero E, Serrano AL, Isern J, and Muñoz-Cánoves P
- Subjects
- Humans, Animals, Signal Transduction, Mitochondria metabolism, Sarcopenia metabolism, Sarcopenia etiology, Aging, Muscle, Skeletal metabolism, Muscle, Skeletal pathology
- Abstract
Sarcopenia is a progressive muscle wasting disorder that severely impacts the quality of life of elderly individuals. Although the natural aging process primarily causes sarcopenia, it can develop in response to other conditions. Because muscle function is influenced by numerous changes that occur with age, the etiology of sarcopenia remains unclear. However, recent characterizations of the aging muscle transcriptional landscape, signaling pathway disruptions, fiber and extracellular matrix compositions, systemic metabolomic and inflammatory responses, mitochondrial function, and neurological inputs offer insights and hope for future treatments. This review will discuss age-related changes in healthy muscle and our current understanding of how this can deteriorate into sarcopenia. As our elderly population continues to grow, we must understand sarcopenia and find treatments that allow individuals to maintain independence and dignity throughout an extended lifespan., (Copyright © 2024. Published by Elsevier Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
7. Extraembryonic hematopoietic lineages-to macrophages and beyond.
- Author
-
Sommer A and Gomez Perdiguero E
- Subjects
- Humans, Animals, Hematopoietic Stem Cells cytology, Mice, Macrophages cytology, Macrophages metabolism, Yolk Sac cytology, Hematopoiesis, Cell Lineage
- Abstract
The first blood and immune cells in vertebrates emerge in the extraembryonic yolk sac. Throughout the last century, it has become evident that this extraembryonic tissue gives rise to transient primitive and definitive hematopoiesis but not hematopoietic stem cells. More recently, studies have elucidated that yolk sac-derived blood and immune cells are present far longer than originally expected. These cells take over essential roles for the survival and proper organogenesis of the developing fetus up until birth. In this review, we discuss the most recent findings and views on extraembryonic hematopoiesis in mice and humans., Competing Interests: Conflicts of Interest Disclosure The authors have nothing to disclose., (Copyright © 2024 International Society for Experimental Hematology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Resident and recruited macrophages differentially contribute to cardiac healing after myocardial ischemia.
- Author
-
Weinberger T, Denise M, Joppich M, Fischer M, Garcia Rodriguez C, Kumaraswami K, Wimmler V, Ablinger S, Räuber S, Fang J, Liu L, Liu WH, Winterhalter J, Lichti J, Thomas L, Esfandyari D, Percin G, Matin S, Hidalgo A, Waskow C, Engelhardt S, Todica A, Zimmer R, Pridans C, Gomez Perdiguero E, and Schulz C
- Subjects
- Animals, Mice, Myocardial Ischemia immunology, Myocardial Infarction pathology, Myocardial Infarction physiopathology, Myocardial Infarction immunology, Male, Myocardial Reperfusion Injury immunology, Myocardial Reperfusion Injury pathology, Mice, Inbred C57BL, Myocardium pathology, Myocardium immunology, Disease Models, Animal, Macrophages immunology, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor genetics
- Abstract
Cardiac macrophages are heterogenous in phenotype and functions, which has been associated with differences in their ontogeny. Despite extensive research, our understanding of the precise role of different subsets of macrophages in ischemia/reperfusion (I/R) injury remains incomplete. We here investigated macrophage lineages and ablated tissue macrophages in homeostasis and after I/R injury in a CSF1R-dependent manner. Genomic deletion of a fms-intronic regulatory element (FIRE) in the Csf1r locus resulted in specific absence of resident homeostatic and antigen-presenting macrophages, without affecting the recruitment of monocyte-derived macrophages to the infarcted heart. Specific absence of homeostatic, monocyte-independent macrophages altered the immune cell crosstalk in response to injury and induced proinflammatory neutrophil polarization, resulting in impaired cardiac remodeling without influencing infarct size. In contrast, continuous CSF1R inhibition led to depletion of both resident and recruited macrophage populations. This augmented adverse remodeling after I/R and led to an increased infarct size and deterioration of cardiac function. In summary, resident macrophages orchestrate inflammatory responses improving cardiac remodeling, while recruited macrophages determine infarct size after I/R injury. These findings attribute distinct beneficial effects to different macrophage populations in the context of myocardial infarction., Competing Interests: TW, MD, MJ, MF, CG, KK, VW, SA, SR, JF, LL, WL, JW, JL, LT, DE, GP, SM, AH, CW, SE, AT, RZ, CP, EG, CS No competing interests declared, (© 2023, Weinberger et al.)
- Published
- 2024
- Full Text
- View/download PDF
9. Early-wave macrophages control late hematopoiesis.
- Author
-
Monticelli S, Sommer A, AlHajj Hassan Z, Garcia Rodriguez C, Adé K, Cattenoz P, Delaporte C, Gomez Perdiguero E, and Giangrande A
- Subjects
- Animals, Mice, Phagocytosis physiology, Drosophila melanogaster, Extracellular Matrix metabolism, Drosophila, Cell Differentiation, Hematopoiesis physiology, Macrophages metabolism, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism
- Abstract
Macrophages constitute the first defense line against the non-self, but their ability to remodel their environment in organ development/homeostasis is starting to be appreciated. Early-wave macrophages (EMs), produced from hematopoietic stem cell (HSC)-independent progenitors, seed the mammalian fetal liver niche wherein HSCs expand and differentiate. The involvement of niche defects in myeloid malignancies led us to identify the cues controlling HSCs. In Drosophila, HSC-independent EMs also colonize the larva when late hematopoiesis occurs. The evolutionarily conserved immune system allowed us to investigate whether/how EMs modulate late hematopoiesis in two models. We show that loss of EMs in Drosophila and mice accelerates late hematopoiesis, which does not correlate with inflammation and does not rely on macrophage phagocytic ability. Rather, EM-derived extracellular matrix components underlie late hematopoiesis acceleration. This demonstrates a developmental role for EMs., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. Brain-muscle communication prevents muscle aging by maintaining daily physiology.
- Author
-
Kumar A, Vaca-Dempere M, Mortimer T, Deryagin O, Smith JG, Petrus P, Koronowski KB, Greco CM, Segalés J, Andrés E, Lukesova V, Zinna VM, Welz PS, Serrano AL, Perdiguero E, Sassone-Corsi P, Benitah SA, and Muñoz-Cánoves P
- Subjects
- Animals, Male, Mice, Circadian Clocks physiology, Homeostasis, Mice, Knockout, ARNTL Transcription Factors genetics, Aging genetics, Aging physiology, Aging, Premature genetics, Aging, Premature prevention & control, Brain physiology, Circadian Rhythm genetics, Circadian Rhythm physiology, Muscle, Skeletal physiology
- Abstract
A molecular clock network is crucial for daily physiology and maintaining organismal health. We examined the interactions and importance of intratissue clock networks in muscle tissue maintenance. In arrhythmic mice showing premature aging, we created a basic clock module involving a central and a peripheral (muscle) clock. Reconstituting the brain-muscle clock network is sufficient to preserve fundamental daily homeostatic functions and prevent premature muscle aging. However, achieving whole muscle physiology requires contributions from other peripheral clocks. Mechanistically, the muscle peripheral clock acts as a gatekeeper, selectively suppressing detrimental signals from the central clock while integrating important muscle homeostatic functions. Our research reveals the interplay between the central and peripheral clocks in daily muscle function and underscores the impact of eating patterns on these interactions.
- Published
- 2024
- Full Text
- View/download PDF
11. Multimodal cell atlas of the ageing human skeletal muscle.
- Author
-
Lai Y, Ramírez-Pardo I, Isern J, An J, Perdiguero E, Serrano AL, Li J, García-Domínguez E, Segalés J, Guo P, Lukesova V, Andrés E, Zuo J, Yuan Y, Liu C, Viña J, Doménech-Fernández J, Gómez-Cabrera MC, Song Y, Liu L, Xu X, Muñoz-Cánoves P, and Esteban MA
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Young Adult, Cell Nucleus metabolism, Chromatin metabolism, Chromatin genetics, Disease Susceptibility, Epigenesis, Genetic, Frailty genetics, Frailty pathology, Muscular Atrophy genetics, Muscular Atrophy pathology, Sarcopenia genetics, Sarcopenia pathology, Transcriptome, Aging genetics, Aging pathology, Aging physiology, Muscle, Skeletal cytology, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, Single-Cell Analysis
- Abstract
Muscle atrophy and functional decline (sarcopenia) are common manifestations of frailty and are critical contributors to morbidity and mortality in older people
1 . Deciphering the molecular mechanisms underlying sarcopenia has major implications for understanding human ageing2 . Yet, progress has been slow, partly due to the difficulties of characterizing skeletal muscle niche heterogeneity (whereby myofibres are the most abundant) and obtaining well-characterized human samples3,4 . Here we generate a single-cell/single-nucleus transcriptomic and chromatin accessibility map of human limb skeletal muscles encompassing over 387,000 cells/nuclei from individuals aged 15 to 99 years with distinct fitness and frailty levels. We describe how cell populations change during ageing, including the emergence of new populations in older people, and the cell-specific and multicellular network features (at the transcriptomic and epigenetic levels) associated with these changes. On the basis of cross-comparison with genetic data, we also identify key elements of chromatin architecture that mark susceptibility to sarcopenia. Our study provides a basis for identifying targets in the skeletal muscle that are amenable to medical, pharmacological and lifestyle interventions in late life., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
12. Context-dependent roles of cellular senescence in normal, aged, and disease states.
- Author
-
Moiseeva V, Cisneros A, Cobos AC, Tarrega AB, Oñate CS, Perdiguero E, Serrano AL, and Muñoz-Cánoves P
- Subjects
- Phenotype, Biological Transport, Cellular Senescence genetics, Longevity
- Abstract
Cellular senescence is a state of irreversible cell cycle arrest that often emerges after tissue damage and in age-related diseases. Through the production of a multicomponent secretory phenotype (SASP), senescent cells can impact the regeneration and function of tissues. However, the effects of senescent cells and their SASP are very heterogeneous and depend on the tissue environment and type as well as the duration of injury, the degree of persistence of senescent cells and the organism's age. While the transient presence of senescent cells is widely believed to be beneficial, recent data suggest that it is detrimental for tissue regeneration after acute damage. Furthermore, although senescent cell persistence is typically associated with the progression of age-related chronic degenerative diseases, it now appears to be also necessary for correct tissue function in the elderly. Here, we discuss what is currently known about the roles of senescent cells and their SASP in tissue regeneration in ageing and age-related diseases, highlighting their (negative and/or positive) contributions. We provide insight for future research, including the possibility of senolytic-based therapies and cellular reprogramming, with aims ranging from enhancing tissue repair to extending a healthy lifespan., (© 2022 Federation of European Biochemical Societies.)
- Published
- 2023
- Full Text
- View/download PDF
13. Author Correction: Senescence atlas reveals an aged-like inflamed niche that blunts muscle regeneration.
- Author
-
Moiseeva V, Cisneros A, Sica V, Deryagin O, Lai Y, Jung S, Andrés E, An J, Segalés J, Ortet L, Lukesova V, Volpe G, Benguria A, Dopazo A, Benitah SA, Urano Y, Del Sol A, Esteban MA, Ohkawa Y, Serrano AL, Perdiguero E, and Muñoz-Cánoves P
- Published
- 2023
- Full Text
- View/download PDF
14. Senescence atlas reveals an aged-like inflamed niche that blunts muscle regeneration.
- Author
-
Moiseeva V, Cisneros A, Sica V, Deryagin O, Lai Y, Jung S, Andrés E, An J, Segalés J, Ortet L, Lukesova V, Volpe G, Benguria A, Dopazo A, Benitah SA, Urano Y, Del Sol A, Esteban MA, Ohkawa Y, Serrano AL, Perdiguero E, and Muñoz-Cánoves P
- Subjects
- Aged, Animals, Humans, Mice, Stem Cells physiology, Fibrosis physiopathology, Transcriptome, Chromatin genetics, Geroscience, Aging metabolism, Aging physiology, Cellular Senescence physiology, Inflammation metabolism, Inflammation physiopathology, Muscle, Skeletal physiology, Muscle, Skeletal physiopathology, Regeneration, Stem Cell Niche physiology
- Abstract
Tissue regeneration requires coordination between resident stem cells and local niche cells
1,2 . Here we identify that senescent cells are integral components of the skeletal muscle regenerative niche that repress regeneration at all stages of life. The technical limitation of senescent-cell scarcity3 was overcome by combining single-cell transcriptomics and a senescent-cell enrichment sorting protocol. We identified and isolated different senescent cell types from damaged muscles of young and old mice. Deeper transcriptome, chromatin and pathway analyses revealed conservation of cell identity traits as well as two universal senescence hallmarks (inflammation and fibrosis) across cell type, regeneration time and ageing. Senescent cells create an aged-like inflamed niche that mirrors inflammation associated with ageing (inflammageing4 ) and arrests stem cell proliferation and regeneration. Reducing the burden of senescent cells, or reducing their inflammatory secretome through CD36 neutralization, accelerates regeneration in young and old mice. By contrast, transplantation of senescent cells delays regeneration. Our results provide a technique for isolating in vivo senescent cells, define a senescence blueprint for muscle, and uncover unproductive functional interactions between senescent cells and stem cells in regenerative niches that can be overcome. As senescent cells also accumulate in human muscles, our findings open potential paths for improving muscle repair throughout life., (© 2022. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
15. Mitochondrial dynamics maintain muscle stem cell regenerative competence throughout adult life by regulating metabolism and mitophagy.
- Author
-
Hong X, Isern J, Campanario S, Perdiguero E, Ramírez-Pardo I, Segalés J, Hernansanz-Agustín P, Curtabbi A, Deryagin O, Pollán A, González-Reyes JA, Villalba JM, Sandri M, Serrano AL, Enríquez JA, and Muñoz-Cánoves P
- Published
- 2022
- Full Text
- View/download PDF
16. Erythro-myeloid progenitor origin of Hofbauer cells in the early mouse placenta.
- Author
-
Freyer L, Lallemand Y, Dardenne P, Sommer A, Biton A, and Gomez Perdiguero E
- Subjects
- Animals, Female, Hematopoietic Stem Cells, Mice, Myeloid Progenitor Cells, Pregnancy, Yolk Sac, Macrophages, Placenta
- Abstract
Hofbauer cells (HBCs) are tissue macrophages of the placenta thought to be important for fetoplacental vascular development and innate immune protection. The developmental origins of HBCs remain unresolved and could implicate functional diversity of HBCs in placenta development and disease. In this study, we used flow cytometry and paternally inherited reporters to phenotype placenta macrophages and to identify fetal-derived HBCs and placenta-associated maternal macrophages in the mouse. In vivo pulse-labeling traced the ontogeny of HBCs from yolk sac-derived erythro-myeloid progenitors, with a minor contribution from fetal hematopoietic stem cells later on. Single-cell RNA-sequencing revealed transcriptional similarities between placenta macrophages and erythro-myeloid progenitor-derived fetal liver macrophages and microglia. As with other fetal tissue macrophages, HBCs were dependent on the transcription factor Pu.1, the loss-of-function of which in embryos disrupted fetoplacental labyrinth morphology, supporting a role for HBC in labyrinth angiogenesis and/or remodeling. HBC were also sensitive to Pu.1 (Spi1) haploinsufficiency, which caused an initial deficiency in the numbers of macrophages in the early mouse placenta. These results provide groundwork for future investigation into the relationship between HBC ontogeny and function in placenta pathophysiology., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2022. Published by The Company of Biologists Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
17. CHD4 ensures stem cell lineage fidelity during skeletal muscle regeneration.
- Author
-
Sreenivasan K, Rodríguez-delaRosa A, Kim J, Mesquita D, Segalés J, Arco PG, Espejo I, Ianni A, Di Croce L, Relaix F, Redondo JM, Braun T, Serrano AL, Perdiguero E, and Muñoz-Cánoves P
- Subjects
- Animals, Computational Biology, Gene Expression Profiling, Gene Expression Regulation, Developmental, Mi-2 Nucleosome Remodeling and Deacetylase Complex metabolism, Mice, Models, Biological, Satellite Cells, Skeletal Muscle cytology, Satellite Cells, Skeletal Muscle metabolism, Cell Differentiation genetics, Cell Lineage genetics, DNA Helicases genetics, Muscle, Skeletal physiology, Regeneration, Stem Cells cytology, Stem Cells metabolism
- Abstract
Regeneration of skeletal muscle requires resident stem cells called satellite cells. Here, we report that the chromatin remodeler CHD4, a member of the nucleosome remodeling and deacetylase (NuRD) repressive complex, is essential for the expansion and regenerative functions of satellite cells. We show that conditional deletion of the Chd4 gene in satellite cells results in failure to regenerate muscle after injury. This defect is principally associated with increased stem cell plasticity and lineage infidelity during the expansion of satellite cells, caused by de-repression of non-muscle-cell lineage genes in the absence of Chd4. Thus, CHD4 ensures that a transcriptional program that safeguards satellite cell identity during muscle regeneration is maintained. Given the therapeutic potential of muscle stem cells in diverse neuromuscular pathologies, CHD4 constitutes an attractive target for satellite cell-based therapies., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
18. Translational control by DHX36 binding to 5'UTR G-quadruplex is essential for muscle stem-cell regenerative functions.
- Author
-
Chen X, Yuan J, Xue G, Campanario S, Wang D, Wang W, Mou X, Liew SW, Umar MI, Isern J, Zhao Y, He L, Li Y, Mann CJ, Yu X, Wang L, Perdiguero E, Chen W, Xue Y, Nagamine Y, Kwok CK, Sun H, Muñoz-Cánoves P, and Wang H
- Subjects
- Animals, Animals, Genetically Modified, Cells, Cultured, Disease Models, Animal, GTP-Binding Protein alpha Subunit, Gi2 metabolism, Gene Expression Regulation, Humans, Mice, Muscles metabolism, Myoblasts metabolism, Polyribosomes metabolism, Protein Biosynthesis, RNA, Messenger genetics, Stem Cells metabolism, 5' Untranslated Regions, DEAD-box RNA Helicases metabolism, G-Quadruplexes, Muscles cytology, Regeneration physiology, Stem Cells cytology
- Abstract
Skeletal muscle has a remarkable ability to regenerate owing to its resident stem cells (also called satellite cells, SCs). SCs are normally quiescent; when stimulated by damage, they activate and expand to form new fibers. The mechanisms underlying SC proliferative progression remain poorly understood. Here we show that DHX36, a helicase that unwinds RNA G-quadruplex (rG4) structures, is essential for muscle regeneration by regulating SC expansion. DHX36 (initially named RHAU) is barely expressed at quiescence but is highly induced during SC activation and proliferation. Inducible deletion of Dhx36 in adult SCs causes defective proliferation and muscle regeneration after damage. System-wide mapping in proliferating SCs reveals DHX36 binding predominantly to rG4 structures at various regions of mRNAs, while integrated polysome profiling shows that DHX36 promotes mRNA translation via 5'-untranslated region (UTR) rG4 binding. Furthermore, we demonstrate that DHX36 specifically regulates the translation of Gnai2 mRNA by unwinding its 5' UTR rG4 structures and identify GNAI2 as a downstream effector of DHX36 for SC expansion. Altogether, our findings uncover DHX36 as an indispensable post-transcriptional regulator of SC function and muscle regeneration acting through binding and unwinding rG4 structures at 5' UTR of target mRNAs., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
19. Megakaryocyte production is sustained by direct differentiation from erythromyeloid progenitors in the yolk sac until midgestation.
- Author
-
Iturri L, Freyer L, Biton A, Dardenne P, Lallemand Y, and Gomez Perdiguero E
- Subjects
- Animals, Cell Lineage physiology, Cells, Cultured, Embryo, Mammalian cytology, Female, Granulocytes cytology, Hematopoiesis physiology, Hematopoietic Stem Cells cytology, Macrophages cytology, Male, Mice, Mice, Inbred C57BL, Monocytes cytology, Multipotent Stem Cells cytology, Pregnancy, Cell Differentiation physiology, Erythrocytes cytology, Megakaryocytes cytology, Myeloid Cells cytology, Stem Cells cytology, Yolk Sac cytology
- Abstract
The extra-embryonic yolk sac contains the first definitive multipotent hematopoietic cells, denominated erythromyeloid progenitors. They originate in situ prior to the emergence of hematopoietic stem cells and give rise to erythroid, monocytes, granulocytes, mast cells and macrophages, the latter in a Myb transcription factor-independent manner. We uncovered here the heterogeneity of yolk sac erythromyeloid progenitors, at the single cell level, and discriminated multipotent from committed progenitors, prior to fetal liver colonization. We identified two temporally distinct megakaryocyte differentiation pathways. The first occurs in the yolk sac, bypasses intermediate bipotent megakaryocyte-erythroid progenitors and, similar to the differentiation of macrophages, is Myb-independent. By contrast, the second originates later, from Myb-dependent bipotent progenitors expressing Csf2rb and colonize the fetal liver, where they give rise to megakaryocytes and to large numbers of erythrocytes. Understanding megakaryocyte development is crucial as they play key functions during vascular development, in particular in separating blood and lymphatic networks., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
20. Yolk sac, but not hematopoietic stem cell-derived progenitors, sustain erythropoiesis throughout murine embryonic life.
- Author
-
Soares-da-Silva F, Freyer L, Elsaid R, Burlen-Defranoux O, Iturri L, Sismeiro O, Pinto-do-Ó P, Gomez-Perdiguero E, and Cumano A
- Subjects
- Animals, Cell Lineage genetics, Erythropoietin metabolism, Female, Macrophages metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Pregnancy, Proto-Oncogene Proteins c-myb deficiency, Proto-Oncogene Proteins c-myb genetics, Embryonic Development genetics, Erythrocytes metabolism, Erythropoiesis, Megakaryocyte Progenitor Cells metabolism, Yolk Sac physiology
- Abstract
In the embryo, the first hematopoietic cells derive from the yolk sac and are thought to be rapidly replaced by the progeny of hematopoietic stem cells. We used three lineage-tracing mouse models to show that, contrary to what was previously assumed, hematopoietic stem cells do not contribute significantly to erythrocyte production up until birth. Lineage tracing of yolk sac erythromyeloid progenitors, which generate tissue resident macrophages, identified highly proliferative erythroid progenitors that rapidly differentiate after intra-embryonic injection, persisting as the major contributors to the embryonic erythroid compartment. We show that erythrocyte progenitors of yolk sac origin require 10-fold lower concentrations of erythropoietin than their hematopoietic stem cell-derived counterparts for efficient erythrocyte production. We propose that, in a low erythropoietin environment in the fetal liver, yolk sac-derived erythrocyte progenitors efficiently outcompete hematopoietic stem cell progeny, which fails to generate megakaryocyte and erythrocyte progenitors., Competing Interests: Disclosures: The authors declare no competing interests exist., (© 2021 Soares-da-Silva et al.)
- Published
- 2021
- Full Text
- View/download PDF
21. Functionally distinct resident macrophage subsets differentially shape responses to infection in the bladder.
- Author
-
Lacerda Mariano L, Rousseau M, Varet H, Legendre R, Gentek R, Saenz Coronilla J, Bajenoff M, Gomez Perdiguero E, and Ingersoll MA
- Subjects
- Animals, Gene Expression Profiling, Macrophages metabolism, Mice, Urinary Bladder, Urinary Tract Infections metabolism
- Abstract
Resident macrophages are abundant in the bladder, playing key roles in immunity to uropathogens. Yet, whether they are heterogeneous, where they come from, and how they respond to infection remain largely unknown. We identified two macrophage subsets in mouse bladders, MacM in muscle and MacL in the lamina propria, each with distinct protein expression and transcriptomes. Using a urinary tract infection model, we validated our transcriptomic analyses, finding that MacM macrophages phagocytosed more bacteria and polarized to an anti-inflammatory profile, whereas MacL macrophages died rapidly during infection. During resolution, monocyte-derived cells contributed to tissue-resident macrophage pools and both subsets acquired transcriptional profiles distinct from naïve macrophages. Macrophage depletion resulted in the induction of a type 1-biased immune response to a second urinary tract infection, improving bacterial clearance. Our study uncovers the biology of resident macrophages and their responses to an exceedingly common infection in a largely overlooked organ, the bladder., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)
- Published
- 2020
- Full Text
- View/download PDF
22. FoxO maintains a genuine muscle stem-cell quiescent state until geriatric age.
- Author
-
García-Prat L, Perdiguero E, Alonso-Martín S, Dell'Orso S, Ravichandran S, Brooks SR, Juan AH, Campanario S, Jiang K, Hong X, Ortet L, Ruiz-Bonilla V, Flández M, Moiseeva V, Rebollo E, Jardí M, Sun HW, Musarò A, Sandri M, Del Sol A, Sartorelli V, and Muñoz-Cánoves P
- Subjects
- Age Factors, Animals, Cardiotoxins toxicity, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cells, Cultured, Forkhead Box Protein O1 genetics, Forkhead Box Protein O1 metabolism, Forkhead Box Protein O3 genetics, Forkhead Box Protein O3 metabolism, Forkhead Transcription Factors genetics, Gene Expression Regulation, Male, Mice, Inbred C57BL, Mice, Knockout, Mice, SCID, Muscle, Skeletal drug effects, Muscle, Skeletal pathology, Muscle, Skeletal transplantation, Phenotype, Proto-Oncogene Proteins c-akt metabolism, Satellite Cells, Skeletal Muscle drug effects, Satellite Cells, Skeletal Muscle pathology, Satellite Cells, Skeletal Muscle transplantation, Signal Transduction, Stem Cell Niche, Antigens, CD34 metabolism, Cell Proliferation drug effects, Cell Proliferation genetics, Cell Self Renewal drug effects, Cell Self Renewal genetics, Cellular Senescence drug effects, Cellular Senescence genetics, Forkhead Transcription Factors metabolism, Muscle, Skeletal metabolism, Regeneration drug effects, Regeneration genetics, Satellite Cells, Skeletal Muscle metabolism
- Abstract
Tissue regeneration declines with ageing but little is known about whether this arises from changes in stem-cell heterogeneity. Here, in homeostatic skeletal muscle, we identify two quiescent stem-cell states distinguished by relative CD34 expression: CD34
High , with stemness properties (genuine state), and CD34Low , committed to myogenic differentiation (primed state). The genuine-quiescent state is unexpectedly preserved into later life, succumbing only in extreme old age due to the acquisition of primed-state traits. Niche-derived IGF1-dependent Akt activation debilitates the genuine stem-cell state by imposing primed-state features via FoxO inhibition. Interventions to neutralize Akt and promote FoxO activity drive a primed-to-genuine state conversion, whereas FoxO inactivation deteriorates the genuine state at a young age, causing regenerative failure of muscle, as occurs in geriatric mice. These findings reveal transcriptional determinants of stem-cell heterogeneity that resist ageing more than previously anticipated and are only lost in extreme old age, with implications for the repair of geriatric muscle.- Published
- 2020
- Full Text
- View/download PDF
23. Attenuated Epigenetic Suppression of Muscle Stem Cell Necroptosis Is Required for Efficient Regeneration of Dystrophic Muscles.
- Author
-
Sreenivasan K, Ianni A, Künne C, Strilic B, Günther S, Perdiguero E, Krüger M, Spuler S, Offermanns S, Gómez-Del Arco P, Redondo JM, Munoz-Canoves P, Kim J, and Braun T
- Subjects
- Humans, Epigenesis, Genetic genetics, Muscle, Skeletal metabolism, Necroptosis genetics
- Abstract
Somatic stem cells expand massively during tissue regeneration, which might require control of cell fitness, allowing elimination of non-competitive, potentially harmful cells. How or if such cells are removed to restore organ function is not fully understood. Here, we show that a substantial fraction of muscle stem cells (MuSCs) undergo necroptosis because of epigenetic rewiring during chronic skeletal muscle regeneration, which is required for efficient regeneration of dystrophic muscles. Inhibition of necroptosis strongly enhances suppression of MuSC expansion in a non-cell-autonomous manner. Prevention of necroptosis in MuSCs of healthy muscles is mediated by the chromatin remodeler CHD4, which directly represses the necroptotic effector Ripk3, while CHD4-dependent Ripk3 repression is dramatically attenuated in dystrophic muscles. Loss of Ripk3 repression by inactivation of Chd4 causes massive necroptosis of MuSCs, abolishing regeneration. Our study demonstrates how programmed cell death in MuSCs is tightly controlled to achieve optimal tissue regeneration., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
24. Sestrin prevents atrophy of disused and aging muscles by integrating anabolic and catabolic signals.
- Author
-
Segalés J, Perdiguero E, Serrano AL, Sousa-Victor P, Ortet L, Jardí M, Budanov AV, Garcia-Prat L, Sandri M, Thomson DM, Karin M, Hee Lee J, and Muñoz-Cánoves P
- Subjects
- Aging, Animals, Autophagy, Disease Models, Animal, Forkhead Box Protein O1 genetics, Forkhead Box Protein O1 metabolism, Forkhead Box Protein O3 genetics, Forkhead Box Protein O3 metabolism, Gene Expression, Heat-Shock Proteins genetics, Humans, Male, Mechanistic Target of Rapamycin Complex 1 genetics, Mechanistic Target of Rapamycin Complex 1 metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Muscle Proteins genetics, Muscle Proteins metabolism, Muscle, Skeletal metabolism, Muscular Atrophy genetics, Muscular Atrophy metabolism, Muscular Atrophy pathology, Nuclear Proteins genetics, Sarcopenia genetics, Sarcopenia metabolism, Sarcopenia pathology, Sarcopenia prevention & control, Heat-Shock Proteins metabolism, Muscle, Skeletal pathology, Muscular Atrophy prevention & control, Nuclear Proteins metabolism, Signal Transduction
- Abstract
A unique property of skeletal muscle is its ability to adapt its mass to changes in activity. Inactivity, as in disuse or aging, causes atrophy, the loss of muscle mass and strength, leading to physical incapacity and poor quality of life. Here, through a combination of transcriptomics and transgenesis, we identify sestrins, a family of stress-inducible metabolic regulators, as protective factors against muscle wasting. Sestrin expression decreases during inactivity and its genetic deficiency exacerbates muscle wasting; conversely, sestrin overexpression suffices to prevent atrophy. This protection occurs through mTORC1 inhibition, which upregulates autophagy, and AKT activation, which in turn inhibits FoxO-regulated ubiquitin-proteasome-mediated proteolysis. This study reveals sestrin as a central integrator of anabolic and degradative pathways preventing muscle wasting. Since sestrin also protected muscles against aging-induced atrophy, our findings have implications for sarcopenia.
- Published
- 2020
- Full Text
- View/download PDF
25. Simultaneous Isolation of Stem and Niche Cells of Skeletal Muscle: Applicability for Aging Studies.
- Author
-
Perdiguero E, Moiseeva V, and Muñoz-Cánoves P
- Subjects
- Adult Stem Cells cytology, Aging, Animals, Antibodies, Endothelial Cells cytology, Endothelial Cells metabolism, Macrophages cytology, Mesenchymal Stem Cells cytology, Mice, Muscle, Skeletal cytology, Real-Time Polymerase Chain Reaction, Regeneration, Satellite Cells, Skeletal Muscle cytology, Stem Cell Niche physiology, Workflow, Adult Stem Cells metabolism, Flow Cytometry methods, Macrophages metabolism, Mesenchymal Stem Cells metabolism, Muscle, Skeletal metabolism, Satellite Cells, Skeletal Muscle metabolism, Stem Cell Niche genetics
- Abstract
The maintenance of adult stem cells in their normal quiescent state depends on intrinsic factors and extrinsic signals originating from their microenvironment (also known as the stem cell niche). In skeletal muscle, its stem cells (satellite cells) lose their regenerative potential with aging, and this has been attributed, at least in part, to both age-associated changes in the satellite cells as in the niche cells, which include resident fibro-adipogenic progenitors (FAPs), macrophages, and endothelial cells, among others. To understand the regenerative decline of skeletal muscle with aging, there is a need for methods to specifically isolate stem and niche cells from resting muscle. Here we describe a fluorescence-activated cell sorting (FACS) protocol to simultaneously isolate discrete populations of satellite cells and niche cells from skeletal muscle of aging mice.
- Published
- 2019
- Full Text
- View/download PDF
26. Musculoskeletal senescence: a moving target ready to be eliminated.
- Author
-
Baar MP, Perdiguero E, Muñoz-Cánoves P, and de Keizer PL
- Subjects
- Age Factors, Animals, Apoptosis drug effects, Bone Remodeling drug effects, Bone and Bones drug effects, Bone and Bones metabolism, Bone and Bones pathology, Bone and Bones physiopathology, Cartilage drug effects, Cartilage metabolism, Cartilage pathology, Cartilage physiopathology, Chondrogenesis drug effects, Drug Design, Humans, Muscle Development drug effects, Muscle, Skeletal drug effects, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, Muscle, Skeletal physiopathology, Musculoskeletal Diseases metabolism, Musculoskeletal Diseases pathology, Musculoskeletal Diseases physiopathology, Musculoskeletal System metabolism, Musculoskeletal System pathology, Musculoskeletal System physiopathology, Signal Transduction drug effects, Cellular Senescence drug effects, Molecular Targeted Therapy methods, Musculoskeletal Diseases drug therapy, Musculoskeletal System drug effects, Regeneration drug effects
- Abstract
Aging is the prime risk factor for the broad-based development of diseases. Frailty is a phenotypical hallmark of aging and is often used to assess whether the predicted benefits of a therapy outweigh the risks for older patients. Senescent cells form as a consequence of unresolved molecular damage and persistently secrete molecules that can impair tissue function. Recent evidence shows senescent cells can chronically interfere with stem cell function and drive aging of the musculoskeletal system. In addition, targeted apoptosis of senescent cells can restore tissue homeostasis in aged animals. Thus, targeting cellular senescence provides new therapeutic opportunities for the intervention of frailty-associated pathologies and could have pleiotropic health benefits., (Copyright © 2018 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
27. Aged Stem Cells Reprogram Their Daily Rhythmic Functions to Adapt to Stress.
- Author
-
Solanas G, Peixoto FO, Perdiguero E, Jardí M, Ruiz-Bonilla V, Datta D, Symeonidi A, Castellanos A, Welz PS, Caballero JM, Sassone-Corsi P, Muñoz-Cánoves P, and Benitah SA
- Subjects
- Adult Stem Cells physiology, Animals, Autophagy, Caloric Restriction, Circadian Clocks, DNA Damage, Diet, High-Fat, Homeostasis, Mice, Stress, Physiological, Transcriptome, Adult Stem Cells pathology, Cellular Senescence, Circadian Rhythm, Epidermis pathology, Muscle, Skeletal pathology
- Abstract
Normal homeostatic functions of adult stem cells have rhythmic daily oscillations that are believed to become arrhythmic during aging. Unexpectedly, we find that aged mice remain behaviorally circadian and that their epidermal and muscle stem cells retain a robustly rhythmic core circadian machinery. However, the oscillating transcriptome is extensively reprogrammed in aged stem cells, switching from genes involved in homeostasis to those involved in tissue-specific stresses, such as DNA damage or inefficient autophagy. Importantly, deletion of circadian clock components did not reproduce the hallmarks of this reprogramming, underscoring that rewiring, rather than arrhythmia, is associated with physiological aging. While age-associated rewiring of the oscillatory diurnal transcriptome is not recapitulated by a high-fat diet in young adult mice, it is significantly prevented by long-term caloric restriction in aged mice. Thus, stem cells rewire their diurnal timed functions to adapt to metabolic cues and to tissue-specific age-related traits., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
28. Cilia Control Fat Deposition during Tissue Repair.
- Author
-
Perdiguero E, Serrano AL, and Muñoz-Cánoves P
- Subjects
- Adipogenesis, Cell Differentiation, Humans, Muscle, Skeletal, Adipocytes, Cilia
- Abstract
Fibro/adipogenic progenitors (FAPs) are emerging as crucial regulators of fibrous and fat deposits during skeletal muscle regeneration. In a recent issue of Cell, Kopinke et al. (2017) report that primary cilia induce the adipogenic fate of FAPs in injured and diseased muscle by restraining Hedgehog signaling., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
29. Identification Of Erythromyeloid Progenitors And Their Progeny In The Mouse Embryo By Flow Cytometry.
- Author
-
Iturri L, Saenz Coronilla J, Lallemand Y, and Gomez Perdiguero E
- Subjects
- Animals, Cell Differentiation, Cells, Cultured, Embryonic Development drug effects, Female, Leukocyte Common Antigens metabolism, Macrophages cytology, Macrophages metabolism, Mice, Proto-Oncogene Proteins c-kit metabolism, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor genetics, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Stem Cells metabolism, Tamoxifen pharmacology, Video Recording, Yolk Sac cytology, Embryo, Mammalian cytology, Flow Cytometry methods, Stem Cells cytology
- Abstract
Macrophages are professional phagocytes from the innate arm of the immune system. In steady-state, sessile macrophages are found in adult tissues where they act as front line sentinels of infection and tissue damage. While other immune cells are continuously renewed from hematopoietic stem and progenitor cells (HSPC) located in the bone marrow, a lineage of macrophages, known as resident macrophages, have been shown to be self-maintained in tissues without input from bone marrow HSPCs. This lineage is exemplified by microglia in the brain, Kupffer cells in the liver and Langerhans cells in the epidermis among others. The intestinal and colon lamina propria are the only adult tissues devoid of HSPC-independent resident macrophages. Recent investigations have identified that resident macrophages originate from the extra-embryonic yolk sac hematopoiesis from progenitor(s) distinct from fetal hematopoietic stem cells (HSC). Among yolk sac definitive hematopoiesis, erythromyeloid progenitors (EMP) give rise both to erythroid and myeloid cells, in particular resident macrophages. EMP are only generated within the yolk sac between E8.5 and E10.5 days of development and they migrate to the fetal liver as early as circulation is connected, where they expand and differentiate until at least E16.5. Their progeny includes erythrocytes, macrophages, neutrophils and mast cells but only EMP-derived macrophages persist until adulthood in tissues. The transient nature of EMP emergence and the temporal overlap with HSC generation renders the analysis of these progenitors difficult. We have established a tamoxifen-inducible fate mapping protocol based on expression of the macrophage cytokine receptor Csf1r promoter to characterize EMP and EMP-derived cells in vivo by flow cytometry.
- Published
- 2017
- Full Text
- View/download PDF
30. Genetic Rescue of Mitochondrial and Skeletal Muscle Impairment in an Induced Pluripotent Stem Cells Model of Coenzyme Q 10 Deficiency.
- Author
-
Romero-Moya D, Santos-Ocaña C, Castaño J, Garrabou G, Rodríguez-Gómez JA, Ruiz-Bonilla V, Bueno C, González-Rodríguez P, Giorgetti A, Perdiguero E, Prieto C, Moren-Nuñez C, Fernández-Ayala DJ, Victoria Cascajo M, Velasco I, Canals JM, Montero R, Yubero D, Jou C, López-Barneo J, Cardellach F, Muñoz-Cánoves P, Artuch R, Navas P, and Menendez P
- Subjects
- Ataxia enzymology, Ataxia pathology, CRISPR-Cas Systems, Cell Differentiation, Child, Preschool, Dopaminergic Neurons cytology, Dopaminergic Neurons metabolism, Electron Transport Chain Complex Proteins genetics, Electron Transport Chain Complex Proteins metabolism, Fatal Outcome, Female, Fibroblasts metabolism, Fibroblasts pathology, Gene Editing methods, Gene Expression, Genes, Lethal, Humans, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells pathology, Intellectual Disability enzymology, Intellectual Disability pathology, Mitochondria enzymology, Mitochondria pathology, Mitochondrial Diseases enzymology, Mitochondrial Diseases pathology, Mitochondrial Proteins deficiency, Motor Neurons cytology, Motor Neurons metabolism, Muscle Weakness enzymology, Muscle Weakness pathology, Primary Cell Culture, Rhabdomyolysis enzymology, Rhabdomyolysis pathology, Ubiquinone genetics, Ataxia genetics, Intellectual Disability genetics, Mitochondria genetics, Mitochondrial Diseases genetics, Mitochondrial Proteins genetics, Muscle Weakness genetics, Rhabdomyolysis genetics, Ubiquinone analogs & derivatives, Ubiquinone deficiency
- Abstract
Coenzyme Q
10 (CoQ10 ) plays a crucial role in mitochondria as an electron carrier within the mitochondrial respiratory chain (MRC) and is an essential antioxidant. Mutations in genes responsible for CoQ10 biosynthesis (COQ genes) cause primary CoQ10 deficiency, a rare and heterogeneous mitochondrial disorder with no clear genotype-phenotype association, mainly affecting tissues with high-energy demand including brain and skeletal muscle (SkM). Here, we report a four-year-old girl diagnosed with minor mental retardation and lethal rhabdomyolysis harboring a heterozygous mutation (c.483G > C (E161D)) in COQ4. The patient's fibroblasts showed a decrease in [CoQ10 ], CoQ10 biosynthesis, MRC activity affecting complexes I/II + III, and respiration defects. Bona fide induced pluripotent stem cell (iPSCs) lines carrying the COQ4 mutation (CQ4-iPSCs) were generated, characterized and genetically edited using the CRISPR-Cas9 system (CQ4ed -iPSCs). Extensive differentiation and metabolic assays of control-iPSCs, CQ4-iPSCs and CQ4ed -iPSCs demonstrated a genotype association, reproducing the disease phenotype. The COQ4 mutation in iPSC was associated with CoQ10 deficiency, metabolic dysfunction, and respiration defects. iPSC differentiation into SkM was compromised, and the resulting SkM also displayed respiration defects. Remarkably, iPSC differentiation in dopaminergic or motor neurons was unaffected. This study offers an unprecedented iPSC model recapitulating CoQ10 deficiency-associated functional and metabolic phenotypes caused by COQ4 mutation. Stem Cells 2017;35:1687-1703., (© 2017 AlphaMed Press.)- Published
- 2017
- Full Text
- View/download PDF
31. Erythro-myeloid progenitors can differentiate from endothelial cells and modulate embryonic vascular remodeling.
- Author
-
Kasaai B, Caolo V, Peacock HM, Lehoux S, Gomez-Perdiguero E, Luttun A, and Jones EA
- Subjects
- Animals, Endothelial Cells metabolism, Erythroid Precursor Cells metabolism, Female, Hematopoiesis, Lipoproteins, LDL metabolism, Male, Mice, Mice, Transgenic, Microscopy, Confocal, Mouse Embryonic Stem Cells metabolism, Myeloid Progenitor Cells metabolism, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor genetics, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Yolk Sac blood supply, Yolk Sac embryology, Endothelial Cells cytology, Erythroid Precursor Cells cytology, Mouse Embryonic Stem Cells cytology, Myeloid Progenitor Cells cytology, Vascular Remodeling, Yolk Sac cytology
- Abstract
Erythro-myeloid progenitors (EMPs) were recently described to arise from the yolk sac endothelium, just prior to vascular remodeling, and are the source of adult/post-natal tissue resident macrophages. Questions remain, however, concerning whether EMPs differentiate directly from the endothelium or merely pass through. We provide the first evidence in vivo that EMPs can emerge directly from endothelial cells (ECs) and demonstrate a role for these cells in vascular development. We find that EMPs express most EC markers but late EMPs and EMP-derived cells do not take up acetylated low-density lipoprotein (AcLDL), as ECs do. When the endothelium is labelled with AcLDL before EMPs differentiate, EMPs and EMP-derived cells arise that are AcLDL
+ . If AcLDL is injected after the onset of EMP differentiation, however, the majority of EMP-derived cells are not double labelled. We find that cell division precedes entry of EMPs into circulation, and that blood flow facilitates the transition of EMPs from the endothelium into circulation in a nitric oxide-dependent manner. In gain-of-function studies, we inject the CSF1-Fc ligand in embryos and found that this increases the number of CSF1R+ cells, which localize to the venous plexus and significantly disrupt venous remodeling. This is the first study to definitively establish that EMPs arise from the endothelium in vivo and show a role for early myeloid cells in vascular development.- Published
- 2017
- Full Text
- View/download PDF
32. Rejuvenating stem cells to restore muscle regeneration in aging.
- Author
-
Bengal E, Perdiguero E, Serrano AL, and Muñoz-Cánoves P
- Abstract
Adult muscle stem cells, originally called satellite cells, are essential for muscle repair and regeneration throughout life. Besides a gradual loss of mass and function, muscle aging is characterized by a decline in the repair capacity, which blunts muscle recovery after injury in elderly individuals. A major effort has been dedicated in recent years to deciphering the causes of satellite cell dysfunction in aging animals, with the ultimate goal of rejuvenating old satellite cells and improving muscle function in elderly people. This review focuses on the recently identified network of cell-intrinsic and -extrinsic factors and processes contributing to the decline of satellite cells in old animals. Some studies suggest that aging-related satellite-cell decay is mostly caused by age-associated extrinsic environmental changes that could be reversed by a "youthful environment". Others propose a central role for cell-intrinsic mechanisms, some of which are not reversed by environmental changes. We believe that these proposals, far from being antagonistic, are complementary and that both extrinsic and intrinsic factors contribute to muscle stem cell dysfunction during aging-related regenerative decline. The low regenerative potential of old satellite cells may reflect the accumulation of deleterious changes during the life of the cell; some of these changes may be inherent (intrinsic) while others result from the systemic and local environment (extrinsic). The present challenge is to rejuvenate aged satellite cells that have undergone reversible changes to provide a possible approach to improving muscle repair in the elderly., Competing Interests: The authors declare that they have no competing interests. No competing interests were disclosed. No competing interests were disclosed. No competing interests were disclosed.
- Published
- 2017
- Full Text
- View/download PDF
33. Muscle Stem Cells: A Model System for Adult Stem Cell Biology.
- Author
-
Cornelison D and Perdiguero E
- Subjects
- Adult Stem Cells cytology, Adult Stem Cells physiology, Animals, Biomarkers, Cell Differentiation, Cellular Senescence genetics, Humans, Muscular Dystrophies etiology, Muscular Dystrophies metabolism, Phenotype, Regeneration, Satellite Cells, Skeletal Muscle cytology, Satellite Cells, Skeletal Muscle physiology, Muscle, Skeletal cytology, Stem Cells cytology, Stem Cells physiology
- Abstract
Skeletal muscle stem cells, originally termed satellite cells for their position adjacent to differentiated muscle fibers, are absolutely required for the process of skeletal muscle repair and regeneration. In the last decade, satellite cells have become one of the most studied adult stem cell systems and have emerged as a standard model not only in the field of stem cell-driven tissue regeneration but also in stem cell dysfunction and aging. Here, we provide background in the field and discuss recent advances in our understanding of muscle stem cell function and dysfunction, particularly in the case of aging, and the potential involvement of muscle stem cells in genetic diseases such as the muscular dystrophies.
- Published
- 2017
- Full Text
- View/download PDF
34. Erratum to: Isolation, Culture, and Immunostaining of Skeletal Muscle Myofibers from Wildtype and Nestin-GFP Mice as a Means to Analyze Satellite Cells.
- Author
-
Perdiguero E and Cornelison D
- Published
- 2017
- Full Text
- View/download PDF
35. Muscle Interstitial Cells: A Brief Field Guide to Non-satellite Cell Populations in Skeletal Muscle.
- Author
-
Tedesco FS, Moyle LA, and Perdiguero E
- Subjects
- AC133 Antigen metabolism, Animals, Biomarkers, Cell Differentiation, Cell Lineage, Cell Proliferation, Humans, Kruppel-Like Transcription Factors metabolism, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells metabolism, Pericytes cytology, Pericytes metabolism, Phenotype, Muscle, Skeletal cytology, Stem Cells cytology, Stem Cells metabolism
- Abstract
Skeletal muscle regeneration is mainly enabled by a population of adult stem cells known as satellite cells. Satellite cells have been shown to be indispensable for adult skeletal muscle repair and regeneration. In the last two decades, other stem/progenitor cell populations resident in the skeletal muscle interstitium have been identified as "collaborators" of satellite cells during regeneration. They also appear to have a key role in replacing skeletal muscle with adipose, fibrous, or bone tissue in pathological conditions. Here, we review the role and known functions of these different interstitial skeletal muscle cell types and discuss their role in skeletal muscle tissue homeostasis, regeneration, and disease, including their therapeutic potential for cell transplantation protocols.
- Published
- 2017
- Full Text
- View/download PDF
36. The Heterogeneity of Ly6C hi Monocytes Controls Their Differentiation into iNOS + Macrophages or Monocyte-Derived Dendritic Cells.
- Author
-
Menezes S, Melandri D, Anselmi G, Perchet T, Loschko J, Dubrot J, Patel R, Gautier EL, Hugues S, Longhi MP, Henry JY, Quezada SA, Lauvau G, Lennon-Duménil AM, Gutiérrez-Martínez E, Bessis A, Gomez-Perdiguero E, Jacome-Galarza CE, Garner H, Geissmann F, Golub R, Nussenzweig MC, and Guermonprez P
- Subjects
- Adoptive Transfer, Animals, Antigens, Ly immunology, Cell Separation, Dendritic Cells cytology, Flow Cytometry, Macrophages cytology, Mice, Monocytes cytology, Nitric Oxide Synthase Type II immunology, Oligonucleotide Array Sequence Analysis, Polymerase Chain Reaction, Cell Differentiation immunology, Dendritic Cells immunology, Macrophages immunology, Monocytes immunology
- Abstract
Inflammation triggers the differentiation of Ly6C
hi monocytes into microbicidal macrophages or monocyte-derived dendritic cells (moDCs). Yet, it is unclear whether environmental inflammatory cues control the polarization of monocytes toward each of these fates or whether specialized monocyte progenitor subsets exist before inflammation. Here, we have shown that naive monocytes are phenotypically heterogeneous and contain an NR4A1- and Flt3L-independent, CCR2-dependent, Flt3+ CD11c- MHCII+ PU.1hi subset. This subset acted as a precursor for FcγRIII+ PD-L2+ CD209a+ , GM-CSF-dependent moDCs but was distal from the DC lineage, as shown by fate-mapping experiments using Zbtb46. By contrast, Flt3- CD11c- MHCII- PU.1lo monocytes differentiated into FcγRIII+ PD-L2- CD209a- iNOS+ macrophages upon microbial stimulation. Importantly, Sfpi1 haploinsufficiency genetically distinguished the precursor activities of monocytes toward moDCs or microbicidal macrophages. Indeed, Sfpi1+/- mice had reduced Flt3+ CD11c- MHCII+ monocytes and GM-CSF-dependent FcγRIII+ PD-L2+ CD209a+ moDCs but generated iNOS+ macrophages more efficiently. Therefore, intercellular disparities of PU.1 expression within naive monocytes segregate progenitor activity for inflammatory iNOS+ macrophages or moDCs., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2016
- Full Text
- View/download PDF
37. ANGPTL4-αvβ3 interaction counteracts hypoxia-induced vascular permeability by modulating Src signalling downstream of vascular endothelial growth factor receptor 2.
- Author
-
Gomez Perdiguero E, Liabotis-Fontugne A, Durand M, Faye C, Ricard-Blum S, Simonutti M, Augustin S, Robb BM, Paques M, Valenzuela DM, Murphy AJ, Yancopoulos GD, Thurston G, Galaup A, Monnot C, and Germain S
- Subjects
- Angiopoietin-Like Protein 4, Angiopoietins deficiency, Animals, Cell Hypoxia physiology, Choroidal Neovascularization metabolism, Choroidal Neovascularization physiopathology, Humans, Mice, Knockout, Phosphorylation physiology, Retina metabolism, Signal Transduction physiology, src-Family Kinases metabolism, Angiopoietins metabolism, Capillary Permeability physiology, Integrin alphaVbeta3 metabolism, Vascular Endothelial Growth Factor Receptor-2 metabolism
- Abstract
Dynamic control of endothelial cell junctions is essential for vascular homeostasis and angiogenesis. We recently provided genetic evidence that ANGPTL4 is a key regulator of vascular integrity both during developmental and in hypoxia-induced pathological conditions. The purpose of the present study was to decipher the molecular mechanisms through which ANGPTL4 regulates vascular integrity. Using surface plasmon resonance and proximity ligation assays, we show that ANGPTL4 binds integrin αvβ3. In vitro and in vivo functional assays with Angptl4-deficient mice demonstrate that ANGPTL4-αvβ3 interaction is necessary to mediate ANGPTL4 vasoprotective effects. Mechanistically, ANGPTL4-αvβ3 interaction enhances Src recruitment to integrin αvβ3 and inhibits Src signalling downstream of vascular endothelial growth factor receptor 2 (VEFGR2), thereby repressing hypoxia-induced breakdown of VEGFR2-VE-cadherin and VEGFR2-αvβ3 complexes. We further demonstrate that intravitreal injection of recombinant human ANGPTL4 limits vascular permeability and leads to increased adherens junction and tight junction integrity. These findings identify a novel mechanism by which ANGPTL4 counteracts hypoxia-driven vascular permeability through integrin αvβ3 binding, modulation of VEGFR2-Src kinase signalling, and endothelial junction stabilization. We further demonstrate that Angptl4-deficient mice show increased vascular leakage in vivo in a model of laser-induced choroidal neovascularization, indicating that this newly identified ANGPTL4-αvβ3 axis might be a target for pharmaceutical intervention in pathological conditions. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd., (Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
38. Specification of tissue-resident macrophages during organogenesis.
- Author
-
Mass E, Ballesteros I, Farlik M, Halbritter F, Günther P, Crozet L, Jacome-Galarza CE, Händler K, Klughammer J, Kobayashi Y, Gomez-Perdiguero E, Schultze JL, Beyer M, Bock C, and Geissmann F
- Subjects
- Animals, CX3C Chemokine Receptor 1, Embryonic Development, Embryonic Induction, Erythroid Precursor Cells cytology, Erythroid Precursor Cells metabolism, Female, Hematopoiesis genetics, Hematopoiesis physiology, Inhibitor of Differentiation Proteins metabolism, Kupffer Cells cytology, Kupffer Cells metabolism, Macrophages metabolism, Mice, Mice, Mutant Strains, Myeloid Progenitor Cells metabolism, Organ Specificity, Receptors, Chemokine genetics, Transcriptome, Cell Differentiation genetics, Embryo, Mammalian cytology, Gene Expression Regulation, Developmental, Macrophages cytology, Myeloid Progenitor Cells cytology, Organogenesis
- Abstract
Tissue-resident macrophages support embryonic development and tissue homeostasis and repair. The mechanisms that control their differentiation remain unclear. We report here that erythro-myeloid progenitors in mice generate premacrophages (pMacs) that simultaneously colonize the whole embryo from embryonic day 9.5 in a chemokine-receptor-dependent manner. The core macrophage program initiated in pMacs is rapidly diversified as expression of transcriptional regulators becomes tissue-specific in early macrophages. This process appears essential for macrophage specification and maintenance, as inactivation of Id3 impairs the development of liver macrophages and results in selective Kupffer cell deficiency in adults. We propose that macrophage differentiation is an integral part of organogenesis, as colonization of organ anlagen by pMacs is followed by their specification into tissue macrophages, hereby generating the macrophage diversity observed in postnatal tissues., (Copyright © 2016, American Association for the Advancement of Science.)
- Published
- 2016
- Full Text
- View/download PDF
39. Regulation of Muscle Stem Cell Functions: A Focus on the p38 MAPK Signaling Pathway.
- Author
-
Segalés J, Perdiguero E, and Muñoz-Cánoves P
- Abstract
Formation of skeletal muscle fibers (myogenesis) during development and after tissue injury in the adult constitutes an excellent paradigm to investigate the mechanisms whereby environmental cues control gene expression programs in muscle stem cells (satellite cells) by acting on transcriptional and epigenetic effectors. Here we will review the molecular mechanisms implicated in the transition of satellite cells throughout the distinct myogenic stages (i.e., activation from quiescence, proliferation, differentiation, and self-renewal). We will also discuss recent findings on the causes underlying satellite cell functional decline with aging. In particular, our review will focus on the epigenetic changes underlying fate decisions and on how the p38 MAPK signaling pathway integrates the environmental signals at the chromatin to build up satellite cell adaptive responses during the process of muscle regeneration, and how these responses are altered in aging. A better comprehension of the signaling pathways connecting external and intrinsic factors will illuminate the path for improving muscle regeneration in the aged.
- Published
- 2016
- Full Text
- View/download PDF
40. The Chromatin Remodeling Complex Chd4/NuRD Controls Striated Muscle Identity and Metabolic Homeostasis.
- Author
-
Gómez-Del Arco P, Perdiguero E, Yunes-Leites PS, Acín-Pérez R, Zeini M, Garcia-Gomez A, Sreenivasan K, Jiménez-Alcázar M, Segalés J, López-Maderuelo D, Ornés B, Jiménez-Borreguero LJ, D'Amato G, Enshell-Seijffers D, Morgan B, Georgopoulos K, Islam AB, Braun T, de la Pompa JL, Kim J, Enriquez JA, Ballestar E, Muñoz-Cánoves P, and Redondo JM
- Subjects
- Aging pathology, Animals, Cardiomyopathies metabolism, Cardiomyopathies pathology, Cell Differentiation genetics, CpG Islands genetics, Gene Expression Regulation, Developmental, Heart embryology, Mice, Transgenic, Mitochondria, Heart metabolism, Muscle, Striated embryology, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Promoter Regions, Genetic genetics, Protein Binding, Chromatin Assembly and Disassembly, DNA Helicases metabolism, Homeostasis, Mi-2 Nucleosome Remodeling and Deacetylase Complex metabolism, Muscle, Striated metabolism
- Abstract
Heart muscle maintains blood circulation, while skeletal muscle powers skeletal movement. Despite having similar myofibrilar sarcomeric structures, these striated muscles differentially express specific sarcomere components to meet their distinct contractile requirements. The mechanism responsible is still unclear. We show here that preservation of the identity of the two striated muscle types depends on epigenetic repression of the alternate lineage gene program by the chromatin remodeling complex Chd4/NuRD. Loss of Chd4 in the heart triggers aberrant expression of the skeletal muscle program, causing severe cardiomyopathy and sudden death. Conversely, genetic depletion of Chd4 in skeletal muscle causes inappropriate expression of cardiac genes and myopathy. In both striated tissues, mitochondrial function was also dependent on the Chd4/NuRD complex. We conclude that an epigenetic mechanism controls cardiac and skeletal muscle structural and metabolic identities and that loss of this regulation leads to hybrid striated muscle tissues incompatible with life., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
41. Chromatin-wide and transcriptome profiling integration uncovers p38α MAPK as a global regulator of skeletal muscle differentiation.
- Author
-
Segalés J, Islam AB, Kumar R, Liu QC, Sousa-Victor P, Dilworth FJ, Ballestar E, Perdiguero E, and Muñoz-Cánoves P
- Subjects
- Animals, Binding Sites, Cell Line, Cell Proliferation, Gene Expression Regulation, Genotype, Mice, Knockout, Mitogen-Activated Protein Kinase 14 antagonists & inhibitors, Mitogen-Activated Protein Kinase 14 deficiency, Mitogen-Activated Protein Kinase 14 genetics, Oligonucleotide Array Sequence Analysis, Phenotype, Promoter Regions, Genetic, Protein Kinase Inhibitors pharmacology, Satellite Cells, Skeletal Muscle drug effects, Signal Transduction, Transcription, Genetic, Cell Differentiation drug effects, Chromatin metabolism, Chromatin Immunoprecipitation, Gene Expression Profiling methods, Mitogen-Activated Protein Kinase 14 metabolism, Muscle Development drug effects, Satellite Cells, Skeletal Muscle enzymology
- Abstract
Background: Extracellular stimuli induce gene expression responses through intracellular signaling mediators. The p38 signaling pathway is a paradigm of the mitogen-activated protein kinase (MAPK) family that, although originally identified as stress-response mediator, contributes to establishing stem cell differentiation fates. p38α is central for induction of the differentiation fate of the skeletal muscle stem cells (satellite cells) through not fully characterized mechanisms., Methods: To investigate the global gene transcription program regulated by p38α during satellite cell differentiation (myogenesis), and to specifically address whether this regulation occurs through direct action of p38α on gene promoters, we performed a combination of microarray gene expression and genome-wide binding analyses. For experimental robustness, two myogenic cellular systems with genetic and chemical loss of p38α function were used: (1) satellite cells derived from mice with muscle-specific deletion of p38α, and (2) the C2C12 murine myoblast cell line cultured in the absence or presence of the p38α/β inhibitor SB203580. Analyses were performed at cell proliferation and early differentiation stages., Results: We show that p38α binds to a large set of active promoters during the transition of myoblasts from proliferation to differentiation stages. p38α-bound promoters are enriched with binding motifs for several transcription factors, with Sp1, Tcf3/E47, Lef1, FoxO4, MyoD, and NFATc standing out in all experimental conditions. p38α association with chromatin correlates very well with high levels of transcription, in agreement with its classical function as an activator of myogenic differentiation. Interestingly, p38α also associates with genes repressed at the onset of differentiation, thus highlighting the relevance of p38-dependent chromatin regulation for transcriptional activation and repression during myogenesis., Conclusions: These results uncover p38α association and function on chromatin at novel classes of target genes during skeletal muscle cell differentiation. This is consistent with this MAPK isoform being a transcriptional regulator.
- Published
- 2016
- Full Text
- View/download PDF
42. Autophagy maintains stemness by preventing senescence.
- Author
-
García-Prat L, Martínez-Vicente M, Perdiguero E, Ortet L, Rodríguez-Ubreva J, Rebollo E, Ruiz-Bonilla V, Gutarra S, Ballestar E, Serrano AL, Sandri M, and Muñoz-Cánoves P
- Subjects
- Aging pathology, Animals, Cell Count, Cyclin-Dependent Kinase Inhibitor p16 genetics, Epigenesis, Genetic, Homeostasis, Humans, Male, Mice, Mitochondria metabolism, Mitochondria pathology, Mitophagy, Muscle, Skeletal cytology, Muscle, Skeletal pathology, Organelles metabolism, Oxidative Stress, Proteins metabolism, Reactive Oxygen Species metabolism, Regeneration, Sarcopenia pathology, Sarcopenia prevention & control, Satellite Cells, Skeletal Muscle pathology, Autophagy physiology, Cellular Senescence, Satellite Cells, Skeletal Muscle cytology
- Abstract
During ageing, muscle stem-cell regenerative function declines. At advanced geriatric age, this decline is maximal owing to transition from a normal quiescence into an irreversible senescence state. How satellite cells maintain quiescence and avoid senescence until advanced age remains unknown. Here we report that basal autophagy is essential to maintain the stem-cell quiescent state in mice. Failure of autophagy in physiologically aged satellite cells or genetic impairment of autophagy in young cells causes entry into senescence by loss of proteostasis, increased mitochondrial dysfunction and oxidative stress, resulting in a decline in the function and number of satellite cells. Re-establishment of autophagy reverses senescence and restores regenerative functions in geriatric satellite cells. As autophagy also declines in human geriatric satellite cells, our findings reveal autophagy to be a decisive stem-cell-fate regulator, with implications for fostering muscle regeneration in sarcopenia.
- Published
- 2016
- Full Text
- View/download PDF
43. Development and function of tissue resident macrophages in mice.
- Author
-
Kierdorf K, Prinz M, Geissmann F, and Gomez Perdiguero E
- Subjects
- Animals, Mice, Models, Biological, Stem Cells cytology, Stem Cells immunology, Macrophages cytology, Macrophages immunology
- Abstract
Macrophages are important for tissue development, homeostasis as well as immune response upon injury or infection. For a long time they were only seen as one uniform group of phagocytes with a common origin and similar functions. However, this view has been challenged in the last decade and revealed a complex diversity of tissue resident macrophages. Here, we want to present the current view on macrophage development and tissue specification and we will discuss differences as well as common patterns between heterogeneous macrophage subpopulations., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
44. Fibrogenic Cell Plasticity Blunts Tissue Regeneration and Aggravates Muscular Dystrophy.
- Author
-
Pessina P, Kharraz Y, Jardí M, Fukada S, Serrano AL, Perdiguero E, and Muñoz-Cánoves P
- Subjects
- Animals, Antigens, CD genetics, Antigens, CD metabolism, Cells, Cultured, Endothelial Cells cytology, Endothelial Cells metabolism, Fibrosis, Integrin alpha Chains genetics, Integrin alpha Chains metabolism, Mice, Mice, Inbred C57BL, Mice, Inbred DBA, Mice, Transgenic, Muscle, Skeletal cytology, Muscle, Skeletal drug effects, Myoblasts cytology, Myoblasts drug effects, Myoblasts metabolism, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Real-Time Polymerase Chain Reaction, Receptor, Platelet-Derived Growth Factor alpha genetics, Receptor, Platelet-Derived Growth Factor alpha metabolism, Satellite Cells, Skeletal Muscle cytology, Satellite Cells, Skeletal Muscle metabolism, Signal Transduction, Smad2 Protein metabolism, Smad3 Protein metabolism, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism, Transforming Growth Factor beta pharmacology, Cell Plasticity, Muscle, Skeletal physiology, Muscular Dystrophy, Duchenne pathology, Regeneration physiology
- Abstract
Preservation of cell identity is necessary for homeostasis of most adult tissues. This process is challenged every time a tissue undergoes regeneration after stress or injury. In the lethal Duchenne muscular dystrophy (DMD), skeletal muscle regenerative capacity declines gradually as fibrosis increases. Using genetically engineered tracing mice, we demonstrate that, in dystrophic muscle, specialized cells of muscular, endothelial, and hematopoietic origins gain plasticity toward a fibrogenic fate via a TGFβ-mediated pathway. This results in loss of cellular identity and normal function, with deleterious consequences for regeneration. Furthermore, this fibrogenic process involves acquisition of a mesenchymal progenitor multipotent status, illustrating a link between fibrogenesis and gain of progenitor cell functions. As this plasticity also was observed in DMD patients, we propose that mesenchymal transitions impair regeneration and worsen diseases with a fibrotic component., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
45. Muscle stem cell aging: regulation and rejuvenation.
- Author
-
Sousa-Victor P, García-Prat L, Serrano AL, Perdiguero E, and Muñoz-Cánoves P
- Subjects
- Animals, Humans, Rejuvenation physiology, Cellular Senescence physiology, Muscle, Skeletal cytology, Stem Cells cytology
- Abstract
Aging is characterized by a progressive decline of physiological integrity leading to the loss of tissue function and vulnerability to disease, but its causes remain poorly understood. Skeletal muscle has an outstanding regenerative capacity that relies on its resident stem cells (satellite cells). This capacity declines with aging, and recent discoveries have redefined our view of why this occurs. Here, we discuss how an interconnection of extrinsic changes in the systemic and local environment and cell-intrinsic mechanisms might provoke failure of normal muscle stem cell functions with aging. We focus particularly on the emergent biology of rejuvenation of old satellite cells, including cells of geriatric age, by restoring traits of youthfulness, with the final goal of improving human health during aging., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
46. Epigenetic control of adult skeletal muscle stem cell functions.
- Author
-
Segalés J, Perdiguero E, and Muñoz-Cánoves P
- Subjects
- Adult, DNA Methylation, Gene Expression Regulation, Humans, Muscle Proteins genetics, Muscle Proteins physiology, Muscle, Skeletal metabolism, Transcription, Genetic, Epigenesis, Genetic, Muscle, Skeletal cytology, Stem Cells cytology
- Abstract
Skeletal muscle regeneration in the adult (de novo myogenesis) depends on a resident population of muscle stem cells (satellite cells) that are normally quiescent. In response to injury or stress, satellite cells are activated and expand as myoblast cells that differentiate and fuse to form new muscle fibers or return to quiescence to maintain the stem cell pool (self-renewal). Satellite cell-dependent myogenesis is a well-characterized multi-step process orchestrated by muscle-specific transcription factors, such as Pax3/Pax7 and members of the MyoD family of muscle regulatory factors, and epigenetically controlled by mechanisms such as DNA methylation, covalent modification of histones and non-coding RNAs. Recent results from next-generation genome-wide sequencing have increased our understanding about the highly intricate layers of epigenetic regulation involved in satellite cell maintenance, activation, differentiation and self-renewal, and their cross-talk with the muscle-specific transcriptional machinery., (© 2014 FEBS.)
- Published
- 2015
- Full Text
- View/download PDF
47. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors.
- Author
-
Gomez Perdiguero E, Klapproth K, Schulz C, Busch K, Azzoni E, Crozet L, Garner H, Trouillet C, de Bruijn MF, Geissmann F, and Rodewald HR
- Subjects
- Animals, Cell Proliferation, Cell Tracking, Female, Fetus cytology, Granulocytes cytology, Kupffer Cells cytology, Langerhans Cells cytology, Liver cytology, Liver embryology, Macrophages, Alveolar cytology, Male, Mice, Microglia cytology, Monocytes cytology, Receptor, Macrophage Colony-Stimulating Factor metabolism, Receptor, TIE-2 metabolism, fms-Like Tyrosine Kinase 3 metabolism, Cell Lineage, Erythrocytes cytology, Hematopoiesis, Macrophages cytology, Stem Cells cytology, Yolk Sac cytology
- Abstract
Most haematopoietic cells renew from adult haematopoietic stem cells (HSCs), however, macrophages in adult tissues can self-maintain independently of HSCs. Progenitors with macrophage potential in vitro have been described in the yolk sac before emergence of HSCs, and fetal macrophages can develop independently of Myb, a transcription factor required for HSC, and can persist in adult tissues. Nevertheless, the origin of adult macrophages and the qualitative and quantitative contributions of HSC and putative non-HSC-derived progenitors are still unclear. Here we show in mice that the vast majority of adult tissue-resident macrophages in liver (Kupffer cells), brain (microglia), epidermis (Langerhans cells) and lung (alveolar macrophages) originate from a Tie2(+) (also known as Tek) cellular pathway generating Csf1r(+) erythro-myeloid progenitors (EMPs) distinct from HSCs. EMPs develop in the yolk sac at embryonic day (E) 8.5, migrate and colonize the nascent fetal liver before E10.5, and give rise to fetal erythrocytes, macrophages, granulocytes and monocytes until at least E16.5. Subsequently, HSC-derived cells replace erythrocytes, granulocytes and monocytes. Kupffer cells, microglia and Langerhans cells are only marginally replaced in one-year-old mice, whereas alveolar macrophages may be progressively replaced in ageing mice. Our fate-mapping experiments identify, in the fetal liver, a sequence of yolk sac EMP-derived and HSC-derived haematopoiesis, and identify yolk sac EMPs as a common origin for tissue macrophages.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.