Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 332
Filtrer
1.
Sci Rep ; 14(1): 23410, 2024 10 08.
Article de Anglais | MEDLINE | ID: mdl-39379408

RÉSUMÉ

Muscle stem cells (MuSCs) are effective in treating inflammatory diseases driven by overactive innate immune responses, such as colitis and acute lung injury, due to their immunomodulatory properties. However, their potential in treating diseases driven by adaptive immune responses is still uncertain. When primed with inflammatory cytokines, MuSCs strongly suppressed T cell activation and proliferation in vitro in co-culture with activated splenocytes or peripheral blood mononuclear cells. Systemic administration of MuSCs from both mice and humans alleviated pathologies in mice with concanavalin A-induced acute liver injury, characterized by hyperactivated T lymphocytes. Importantly, MuSCs showed significant species-specific differences in their immunoregulatory functions. In mouse MuSCs (mMuSCs), deletion or inhibition of inducible nitric oxide synthase (iNOS) reduced their immunosuppressive activity, and absence of iNOS negated their therapeutic effects in liver injury. Conversely, in human MuSCs (hMuSCs), knockdown or inhibition of indoleamine 2,3-dioxygenase (IDO) eliminated their immunosuppressive effects, and loss of IDO function rendered hMuSCs ineffective in treating liver injury in mice. These results reveal significant species-specific differences in the mechanisms by which MuSCs mediate T cell immunosuppression. Mouse MuSCs rely on iNOS, while human MuSCs depend on IDO expression. This highlights the need to consider species-specific responses when evaluating MuSCs' therapeutic potential in immune-related disorders.


Sujet(s)
Nitric oxide synthase type II , Lymphocytes T , Animaux , Humains , Lymphocytes T/immunologie , Souris , Nitric oxide synthase type II/métabolisme , Cellules souches/métabolisme , Cellules souches/immunologie , Spécificité d'espèce , Activation des lymphocytes/immunologie , Indoleamine-pyrrole 2,3,-dioxygenase/métabolisme , Souris de lignée C57BL , Immunosuppression thérapeutique/méthodes , Tolérance immunitaire , Prolifération cellulaire , Concanavaline A/pharmacologie , Lésions hépatiques dues aux substances/immunologie
2.
Cell ; 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39305903

RÉSUMÉ

Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disease caused by mutations in the DMD gene. Muscle fibers rely on the coordination of multiple cell types for repair and regenerative capacity. To elucidate the cellular and molecular changes in these cell types under pathologic conditions, we generated a rhesus monkey model for DMD that displays progressive muscle deterioration and impaired motor function, mirroring human conditions. By leveraging these DMD monkeys, we analyzed freshly isolated muscle tissues using single-cell RNA sequencing (scRNA-seq). Our analysis revealed changes in immune cell landscape, a reversion of lineage progressing directions in fibrotic fibro-adipogenic progenitors (FAPs), and TGF-ß resistance in FAPs and muscle stem cells (MuSCs). Furthermore, MuSCs displayed cell-intrinsic defects, leading to differentiation deficiencies. Our study provides important insights into the pathogenesis of DMD, offering a valuable model and dataset for further exploration of the underlying mechanisms, and serves as a suitable platform for developing and evaluating therapeutic interventions.

3.
Proc Natl Acad Sci U S A ; 121(35): e2406787121, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39163337

RÉSUMÉ

Muscle stem cells (MuSCs) are specialized cells that reside in adult skeletal muscle poised to repair muscle tissue. The ability of MuSCs to regenerate damaged tissues declines markedly with aging and in diseases such as Duchenne muscular dystrophy, but the underlying causes of MuSC dysfunction remain poorly understood. Both aging and disease result in dramatic increases in the stiffness of the muscle tissue microenvironment from fibrosis. MuSCs are known to lose their regenerative potential if cultured on stiff plastic substrates. We sought to determine whether MuSCs harbor a memory of their past microenvironment and if it can be overcome. We tested MuSCs in situ using dynamic hydrogel biomaterials that soften or stiffen on demand in response to light and found that freshly isolated MuSCs develop a persistent memory of substrate stiffness characterized by loss of proliferative progenitors within the first three days of culture on stiff substrates. MuSCs cultured on soft hydrogels had altered cytoskeletal organization and activity of Rho and Rac guanosine triphosphate hydrolase (GTPase) and Yes-associated protein mechanotransduction pathways compared to those on stiff hydrogels. Pharmacologic inhibition identified RhoA activation as responsible for the mechanical memory phenotype, and single-cell RNA sequencing revealed a molecular signature of the mechanical memory. These studies highlight that microenvironmental stiffness regulates MuSC fate and leads to MuSC dysfunction that is not readily reversed by changing stiffness. Our results suggest that stiffness can be circumvented by targeting downstream signaling pathways to overcome stem cell dysfunction in aged and disease states with aberrant fibrotic tissue mechanics.


Sujet(s)
Matériaux biocompatibles , Hydrogels , Muscles squelettiques , Animaux , Hydrogels/composition chimique , Matériaux biocompatibles/composition chimique , Muscles squelettiques/métabolisme , Souris , Mécanotransduction cellulaire , Cellules souches/métabolisme , Cellules souches/cytologie , Protéine G RhoA/métabolisme , Cellules cultivées
4.
J Biochem ; 176(4): 277-283, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39194026

RÉSUMÉ

The skeletal muscle is a contractile tissue distributed throughout the body with various anatomical sizes, shapes and functions. In pathological conditions, such as muscular dystrophy, age-related sarcopenia and cancer cachexia, skeletal muscles are not uniformly affected throughout the body. This region-specific vulnerability cannot be fully explained by known physiological classifications, including muscle fiber types. Accumulating evidence indicates that the expression patterns of topographic homeobox (Hox) genes provide a molecular signature of positional memory, reflecting the anatomical locations and embryonic history of muscles and their associated muscle stem cells in adult mice and humans. Hox-based positional memory is not merely a remnant of embryonic development but is expected to be an intrinsic determinant controlling muscle function because recent studies have shown that aberrant Hox genes affect muscle stem cells. In this review, we discuss the concept of Hox-based positional memory, which may offer a new perspective on the region-specific pathophysiology of muscle disorders.


Sujet(s)
Muscles squelettiques , Animaux , Humains , Muscles squelettiques/métabolisme , Protéines à homéodomaine/métabolisme , Protéines à homéodomaine/génétique , Cellules souches/métabolisme , Cellules souches/cytologie , Gènes homéotiques , Souris
5.
Dev Cell ; 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39121856

RÉSUMÉ

Muscle stem cells (MuSCs) enable muscle growth and regeneration after exercise or injury, but how metabolism controls their regenerative potential is poorly understood. We describe that primary metabolic changes can determine murine MuSC fate decisions. We found that glutamine anaplerosis into the tricarboxylic acid (TCA) cycle decreases during MuSC differentiation and coincides with decreased expression of the mitochondrial glutamate deaminase GLUD1. Deletion of Glud1 in proliferating MuSCs resulted in precocious differentiation and fusion, combined with loss of self-renewal in vitro and in vivo. Mechanistically, deleting Glud1 caused mitochondrial glutamate accumulation and inhibited the malate-aspartate shuttle (MAS). The resulting defect in transporting NADH-reducing equivalents into the mitochondria induced compartment-specific NAD+/NADH ratio shifts. MAS activity restoration or directly altering NAD+/NADH ratios normalized myogenesis. In conclusion, GLUD1 prevents deleterious mitochondrial glutamate accumulation and inactivation of the MAS in proliferating MuSCs. It thereby acts as a compartment-specific metabolic brake on MuSC differentiation.

6.
Biochem Soc Trans ; 52(4): 1921-1926, 2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39136196

RÉSUMÉ

Skeletal muscle stem cells (MuSCs) display distinct behavior crucial for tissue maintenance and repair. Upon activation, MuSCs exhibit distinct modes of division: symmetric division, facilitating either self-renewal or differentiation, and asymmetric division, which dictates divergent cellular fates. This review explores the nuanced dynamics of MuSC division and the molecular mechanisms governing this behavior. Furthermore, it introduces a novel phenomenon observed in a subset of MuSCs under hypertrophic stimuli termed division-independent differentiation. Insights into the underlying mechanisms driving this process are discussed, alongside its broader implications for muscle physiology.


Sujet(s)
Différenciation cellulaire , Hypertrophie , Muscles squelettiques , Cellules satellites du muscle squelettique , Cellules satellites du muscle squelettique/métabolisme , Cellules satellites du muscle squelettique/physiologie , Humains , Animaux , Division cellulaire
7.
J Cell Sci ; 137(15)2024 08 01.
Article de Anglais | MEDLINE | ID: mdl-39037211

RÉSUMÉ

Muscle stem cells (MuSCs) play an indispensable role in postnatal muscle growth and hypertrophy in adults. MuSCs also retain a highly regenerative capacity and are therefore considered a promising stem cell source for regenerative therapy for muscle diseases. In this study, we identify tumor-suppressor protein Tob1 as a Pax7 target protein that negatively controls the population expansion of MuSCs. Tob1 protein is undetectable in the quiescent state but is upregulated during activation in MuSCs. Tob1 ablation in mice accelerates MuSC population expansion and boosts muscle regeneration. Moreover, inactivation of Tob1 in MuSCs ameliorates the efficiency of MuSC transplantation in a murine muscular dystrophy model. Collectively, selective targeting of Tob1 might be a therapeutic option for the treatment of muscular diseases, including muscular dystrophy and age-related sarcopenia.


Sujet(s)
Muscles squelettiques , Facteur de transcription PAX7 , Régénération , Cellules souches , Animaux , Souris , Muscles squelettiques/métabolisme , Facteur de transcription PAX7/métabolisme , Facteur de transcription PAX7/génétique , Cellules souches/métabolisme , Cellules souches/cytologie , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Prolifération cellulaire , Souris de lignée C57BL
8.
Dev Cell ; 59(17): 2375-2392.e8, 2024 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-38848717

RÉSUMÉ

The histone H3 lysine 9 methyltransferase SETDB1 controls transcriptional repression to direct stem cell fate. Here, we show that Setdb1 expression by adult muscle stem cells (MuSCs) is required for skeletal muscle regeneration. We find that SETDB1 represses the expression of endogenous retroviruses (ERVs) in MuSCs. ERV de-repression in Setdb1-null MuSCs prevents their amplification following exit from quiescence and promotes cell death. Multi-omics profiling shows that chromatin decompaction at ERV loci activates the DNA-sensing cGAS-STING pathway, entailing cytokine expression by Setdb1-null MuSCs. This is followed by aberrant infiltration of inflammatory cells, including pathological macrophages. The ensuing histiocytosis is accompanied by myofiber necrosis, which, in addition to progressive MuSCs depletion, completely abolishes tissue repair. In contrast, loss of Setdb1 in fibro-adipogenic progenitors (FAPs) does not impact immune cells. In conclusion, genome maintenance by SETDB1 in an adult somatic stem cell is necessary for both its regenerative potential and adequate reparative inflammation.


Sujet(s)
Histone-lysine N-methyltransferase , Inflammation , Développement musculaire , Régénération , Animaux , Histone-lysine N-methyltransferase/métabolisme , Histone-lysine N-methyltransferase/génétique , Souris , Inflammation/anatomopathologie , Inflammation/métabolisme , Inflammation/génétique , Régénération/génétique , Développement musculaire/génétique , Souris de lignée C57BL , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Rétrovirus endogènes/génétique , Cellules souches/métabolisme , Cellules souches/cytologie , Génome , Différenciation cellulaire/génétique
9.
Stem Cell Reports ; 19(7): 1024-1040, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38876109

RÉSUMÉ

Increasing evidence suggests that the muscle stem cell (MuSC) pool is heterogeneous. In particular, a rare subset of PAX7-positive MuSCs that has never expressed the myogenic regulatory factor MYF5 displays unique self-renewal and engraftment characteristics. However, the scarcity and limited availability of protein markers make the characterization of these cells challenging. Here, we describe the generation of StemRep reporter mice enabling the monitoring of PAX7 and MYF5 proteins based on equimolar levels of dual nuclear fluorescence. High levels of PAX7 protein and low levels of MYF5 delineate a deeply quiescent MuSC subpopulation with an increased capacity for asymmetric division and distinct dynamics of activation, proliferation, and commitment. Aging primarily reduces the MYF5Low MuSCs and skews the stem cell pool toward MYF5High cells with lower quiescence and self-renewal potential. Altogether, we establish the StemRep model as a versatile tool to study MuSC heterogeneity and broaden our understanding of mechanisms regulating MuSC quiescence and self-renewal in homeostatic, regenerating, and aged muscles.


Sujet(s)
Vieillissement , Gènes rapporteurs , Facteur-5 de régulation myogène , Facteur de transcription PAX7 , Régénération , Animaux , Facteur de transcription PAX7/métabolisme , Facteur de transcription PAX7/génétique , Facteur-5 de régulation myogène/métabolisme , Facteur-5 de régulation myogène/génétique , Souris , Vieillissement/métabolisme , Cellules souches/métabolisme , Cellules souches/cytologie , Prolifération cellulaire , Muscles squelettiques/métabolisme , Muscles squelettiques/cytologie , Différenciation cellulaire , Souris transgéniques , Auto-renouvellement cellulaire
10.
Front Cell Dev Biol ; 12: 1385399, 2024.
Article de Anglais | MEDLINE | ID: mdl-38840849

RÉSUMÉ

Skeletal muscle regeneration relies on the intricate interplay of various cell populations within the muscle niche-an environment crucial for regulating the behavior of muscle stem cells (MuSCs) and ensuring postnatal tissue maintenance and regeneration. This review delves into the dynamic interactions among key players of this process, including MuSCs, macrophages (MPs), fibro-adipogenic progenitors (FAPs), endothelial cells (ECs), and pericytes (PCs), each assuming pivotal roles in orchestrating homeostasis and regeneration. Dysfunctions in these interactions can lead not only to pathological conditions but also exacerbate muscular dystrophies. The exploration of cellular and molecular crosstalk among these populations in both physiological and dystrophic conditions provides insights into the multifaceted communication networks governing muscle regeneration. Furthermore, this review discusses emerging strategies to modulate the muscle-regenerating niche, presenting a comprehensive overview of current understanding and innovative approaches.

11.
Stem Cell Res Ther ; 15(1): 179, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38902774

RÉSUMÉ

BACKGROUND: Adult skeletal muscle contains resident muscle stem cells (MuSC) with high myogenic and engraftment potentials, making them suitable for cell therapy and regenerative medicine approaches. However, purification process of MuSC remains a major hurdle to their use in the clinic. Indeed, muscle tissue enzymatic dissociation triggers a massive activation of stress signaling pathways, among which P38 and JNK MAPK, associated with a premature loss of MuSC quiescence. While the role of these pathways in the myogenic progression of MuSC is well established, the extent to which their dissociation-induced activation affects the functionality of these cells remains unexplored. METHODS: We assessed the effect of P38 and JNK MAPK induction on stemness marker expression and MuSC activation state during isolation by pharmacological approaches. MuSC functionality was evaluated by in vitro assays and in vivo transplantation experiments. We performed a comparative analysis of the transcriptome of human MuSC purified with pharmacological inhibitors of P38 and JNK MAPK (SB202190 and SP600125, respectively) versus available RNAseq resources. RESULTS: We monitored PAX7 protein levels in murine MuSC during muscle dissociation and demonstrated a two-step decline partly dependent on P38 and JNK MAPK activities. We showed that simultaneous inhibition of these pathways throughout the MuSC isolation process preserves the expression of stemness markers and limits their premature activation, leading to improved survival and amplification in vitro as well as increased engraftment in vivo. Through a comparative RNAseq analysis of freshly isolated human MuSC, we provide evidence that our findings in murine MuSC could be relevant to human MuSC. Based on these findings, we implemented a purification strategy, significantly improving the recovery yields of human MuSC. CONCLUSION: Our study highlights the pharmacological limitation of P38 and JNK MAPK activities as a suitable strategy to qualitatively and quantitatively ameliorate human MuSC purification process, which could be of great interest for cell-based therapies.


Sujet(s)
p38 Mitogen-Activated Protein Kinases , p38 Mitogen-Activated Protein Kinases/métabolisme , Animaux , Humains , Souris , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Muscles squelettiques/métabolisme , Muscles squelettiques/cytologie , Cellules souches/métabolisme , Cellules souches/cytologie , Cellules souches/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Mâle , Anthracènes/pharmacologie , JNK Mitogen-Activated Protein Kinases/métabolisme
12.
Int J Biol Macromol ; 271(Pt 1): 131980, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38821790

RÉSUMÉ

The mass proliferation of seed cells and imitation of meat structures remain challenging for cell-cultured meat production. With excellent biocompatibility, high water content and porosity, hydrogels are frequently-studied materials for anchorage-dependent cell scaffolds in biotechnology applications. Herein, a scaffold based on gelatin/alginate/ε-Poly-l-lysine (GAL) hydrogel is developed for skeletal muscle cells, which has a great prospect in cell-cultured meat production. In this work, the hydrogel GAL-4:1, composed of gelatin (5 %, w/v), alginate (5 %, w/v) and ε-Poly-l-lysine (molar ratio vs. alginate: 4:1) is selected as cell scaffold based on Young's modulus of 11.29 ± 1.94 kPa, satisfactory shear-thinning property and suitable porous organized structure. The commercially available C2C12 mouse skeletal myoblasts and porcine muscle stem cells (PMuSCs), are cultured in the 3D-printed scaffold. The cells show strong ability of attachment, proliferation and differentiation after induction, showing high biocompatibility. Furthermore, the cellular bioprinting is performed with GAL-4:1 hydrogel and freshly extracted PMuSCs. The extracted PMuSCs exhibit high viability and display early myogenesis (desmin) on the 3D scaffold, suggesting the great potential of GAL hydrogel as 3D cellular constructs scaffolds. Overall, we develop a novel GAL hydrogel as a 3D-printed bioactive platform for cultured meat research.


Sujet(s)
Alginates , Différenciation cellulaire , Prolifération cellulaire , Gélatine , Hydrogels , Polylysine , Impression tridimensionnelle , Structures d'échafaudage tissulaires , Animaux , Alginates/composition chimique , Gélatine/composition chimique , Polylysine/composition chimique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Structures d'échafaudage tissulaires/composition chimique , Suidae , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris , Hydrogels/composition chimique , Cellules souches/cytologie , Viande , Développement musculaire , Ingénierie tissulaire/méthodes , Lignée cellulaire , Bio-impression/méthodes , In Vitro Meat
13.
Food Sci Anim Resour ; 44(3): 710-722, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38765289

RÉSUMÉ

Extracellular matrix (ECM) proteins play a crucial role in culturing muscle stem cells (MuSCs). However, there is a lack of extensive research on how each of these proteins influences proliferation and differentiation of MuSCs from livestock animals. Therefore, we investigated the effects of various ECM coatings-collagen, fibronectin, gelatin, and laminin-on the proliferation, differentiation, and maturation of porcine MuSCs. Porcine MuSCs, isolated from 14-day-old Berkshire piglets, were cultured on ECM-coated plates, undergoing three days of proliferation followed by three days of differentiation. MuSCs on laminin showed higher proliferation rate than others (p<0.05). There was no significant difference in the mRNA expression levels of PAX7, MYF5, and MYOD among MuSCs on laminin, collagen, and fibronectin (p>0.05). During the differentiation period, MuSCs cultured on laminin exhibited a significantly higher differentiation rate, resulting in thicker myotubes compared to those on other ECMs (p<0.05). Also, MuSCs on laminin showed higher expression of mRNA related with maturated muscle fiber such as MYH1 and MYH4 corresponding to muscle fiber type IIx and muscle fiber type IIb, respectively, compared with MuSCs on other ECM coatings (p<0.05). In summary, our comparison of ECMs revealed that laminin significantly enhances MuSC proliferation and differentiation, outperforming other ECMs. Specifically, muscle fibers cultured on laminin exhibited a more mature phenotype. These findings underscore laminin's potential to advance in vitro muscle research and cultured meat production, highlighting its role in supporting rapid cell proliferation, higher differentiation rates, and the development of mature muscle fibers.

14.
Am J Physiol Cell Physiol ; 327(1): C213-C219, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38586876

RÉSUMÉ

Muscle isometric torque fluctuates according to time-of-day with such variation owed to the influence of circadian molecular clock genes. Satellite cells (SCs), the muscle stem cell population, also express molecular clock genes with several contractile-related genes oscillating in a diurnal pattern. Currently, limited evidence exists regarding the relationship between SCs and contractility, although long-term SC ablation alters muscle contractile function. Whether there are acute alterations in contractility following SC ablation and with respect to the time-of-day is unknown. We investigated whether short-term SC ablation affected contractile function at two times of day and whether any such alterations led to different extents of eccentric contraction-induced injury. Using an established mouse model to deplete SCs, we characterized muscle clock gene expression and ex vivo contractility at two times-of-day (morning: 0700 and afternoon: 1500). Morning-SC+ animals demonstrated ∼25%-30% reductions in tetanic/eccentric specific forces and, after eccentric injury, exhibited ∼30% less force-loss and ∼50% less dystrophinnegative fibers versus SC- counterparts; no differences were noted between Afternoon groups (Morning-SC+: -5.63 ± 0.61, Morning-SC-: -7.93 ± 0.61; N/cm2; P < 0.05) (Morning-SC+: 32 ± 2.1, Morning-SC-: 64 ± 10.2; dystrophinnegative fibers; P < 0.05). As Ca++ kinetics underpin force generation, we also evaluated caffeine-induced contracture force as an indirect marker of Ca++ availability and found similar force reductions in Morning-SC+ vs. SC- mice. We conclude that force production is reduced in the presence of SCs in the morning but not in the afternoon, suggesting that SCs may have a time-of-day influence over contractile function.NEW & NOTEWORTHY Muscle isometric torque fluctuates according to time-of-day with such variation owed to molecular clock regulation. Satellite cells (SCs) have recently demonstrated diurnal characteristics related to muscle physiology. In our work, force production was reduced in the presence versus absence of SCs in the morning but, not in the afternoon. Morning-SC+ animals, producing lower force, sustained lesser degrees of injury versus SC- counterparts. One potential mechanism underpinning lower forces produced appears to be lower calcium availability.


Sujet(s)
Rythme circadien , Contraction musculaire , Cellules satellites du muscle squelettique , Animaux , Cellules satellites du muscle squelettique/métabolisme , Souris , Rythme circadien/physiologie , Mâle , Souris de lignée C57BL , Muscles squelettiques/métabolisme , Muscles squelettiques/physiologie , Facteurs temps
15.
Stem Cell Reports ; 19(5): 673-688, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38579709

RÉSUMÉ

Maintenance of mitochondrial function plays a crucial role in the regulation of muscle stem cell (MuSC), but the underlying mechanisms remain ill defined. In this study, we monitored mitophagy in MuSCS under various myogenic states and examined the role of PINK1 in maintaining regenerative capacity. Results indicate that quiescent MuSCs actively express mitophagy genes and exhibit a measurable mitophagy flux and prominent mitochondrial localization to autophagolysosomes, which become rapidly decreased during activation. Genetic disruption of Pink1 in mice reduces PARKIN recruitment to mitochondria and mitophagy in quiescent MuSCs, which is accompanied by premature activation/commitment at the expense of self-renewal and progressive loss of muscle regeneration, but unhindered proliferation and differentiation capacity. Results also show that impaired fate decisions in PINK1-deficient MuSCs can be restored by scavenging excess mitochondrial ROS. These data shed light on the regulation of mitophagy in MuSCs and position PINK1 as an important regulator of their mitochondrial properties and fate decisions.


Sujet(s)
Différenciation cellulaire , Mitophagie , Protein kinases , Régénération , Cellules souches , Animaux , Mitophagie/génétique , Protein kinases/métabolisme , Protein kinases/génétique , Protein kinases/déficit , Souris , Différenciation cellulaire/génétique , Cellules souches/métabolisme , Cellules souches/cytologie , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/déficit , Mitochondries/métabolisme , Muscles squelettiques/métabolisme , Muscles squelettiques/cytologie , Espèces réactives de l'oxygène/métabolisme , Développement musculaire/génétique , Prolifération cellulaire
16.
Curr Top Dev Biol ; 158: 15-51, 2024.
Article de Anglais | MEDLINE | ID: mdl-38670703

RÉSUMÉ

Skeletal muscle is a highly represented tissue in mammals and is composed of fibers that are extremely adaptable and capable of regeneration. This characteristic of muscle fibers is made possible by a cell type called satellite cells. Adjacent to the fibers, satellite cells are found in a quiescent state and located between the muscle fibers membrane and the basal lamina. These cells are required for the growth and regeneration of skeletal muscle through myogenesis. This process is known to be tightly sequenced from the activation to the differentiation/fusion of myofibers. However, for the past fifteen years, researchers have been interested in examining satellite cell heterogeneity and have identified different subpopulations displaying distinct characteristics based on localization, quiescence state, stemness capacity, cell-cycle progression or gene expression. A small subset of satellite cells appears to represent multipotent long-term self-renewing muscle stem cells (MuSC). All these distinctions led us to the hypothesis that the characteristics of myogenesis might not be linear and therefore may be more permissive based on the evidence that satellite cells are a heterogeneous population. In this review, we discuss the different subpopulations that exist within the satellite cell pool to highlight the heterogeneity and to gain further understanding of the myogenesis progress. Finally, we discuss the long term self-renewing MuSC subpopulation that is capable of dividing asymmetrically and discuss the molecular mechanisms regulating MuSC polarization during health and disease.


Sujet(s)
Développement musculaire , Muscles squelettiques , Cellules satellites du muscle squelettique , Cellules satellites du muscle squelettique/cytologie , Cellules satellites du muscle squelettique/physiologie , Cellules satellites du muscle squelettique/métabolisme , Animaux , Humains , Muscles squelettiques/cytologie , Muscles squelettiques/physiologie , Différenciation cellulaire , Régénération/physiologie
17.
Dev Cell ; 59(11): 1457-1474.e5, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38569550

RÉSUMÉ

The function of many organs, including skeletal muscle, depends on their three-dimensional structure. Muscle regeneration therefore requires not only reestablishment of myofibers but also restoration of tissue architecture. Resident muscle stem cells (SCs) are essential for regeneration, but how SCs regenerate muscle architecture is largely unknown. We address this problem using genetic labeling of mouse SCs and whole-mount imaging to reconstruct, in three dimensions, muscle regeneration. Unexpectedly, we found that myofibers form via two distinct phases of fusion and the residual basement membrane of necrotic myofibers is critical for promoting fusion and orienting regenerated myofibers. Furthermore, the centralized myonuclei characteristic of regenerated myofibers are associated with myofibrillogenesis and endure months post injury. Finally, we elucidate two cellular mechanisms for the formation of branched myofibers, a pathology characteristic of diseased muscle. We provide a synthesis of the cellular events of regeneration and show that these differ from those used during development.


Sujet(s)
Imagerie tridimensionnelle , Muscles squelettiques , Régénération , Animaux , Régénération/physiologie , Souris , Muscles squelettiques/physiologie , Imagerie tridimensionnelle/méthodes , Fibres musculaires squelettiques/métabolisme , Fibres musculaires squelettiques/cytologie , Développement musculaire/physiologie , Cellules souches/cytologie , Cellules souches/métabolisme , Membrane basale/métabolisme
18.
Methods Mol Biol ; 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38647863

RÉSUMÉ

Adult skeletal muscle stem cells (MuSC) are the regenerative precursors of myofibers and also have an important role in myofiber growth, adaptation, and maintenance by fusing to the myofibers-a process referred to as "myonuclear accretion." Due to a focus on MuSC function during regeneration, myofibers remain a largely overlooked component of the MuSC niche influencing MuSC fate. Here, we describe a method to directly measure the rate of myonuclear accretion in vitro and in vivo using ethynyl-2'-deoxyuridine (EdU)-based tracing of MuSC progeny. This method supports the dissection of MuSC intrinsic and myofiber-derived factors influencing myonuclear accretion as an alternative fate of MuSCs supporting myofiber homeostasis and plasticity.

19.
Curr Top Dev Biol ; 158: 203-220, 2024.
Article de Anglais | MEDLINE | ID: mdl-38670706

RÉSUMÉ

Skeletal muscle is composed of a variety of tissue and non-tissue resident cells that participate in homeostasis. In particular, the muscle stem cell niche is a dynamic system, requiring direct and indirect communications between cells, involving local and remote cues. Interactions within the niche must happen in a timely manner for the maintenance or recovery of the homeostatic niche. For instance, after an injury, pro-myogenic cues delivered too early will impact on muscle stem cell proliferation, delaying the repair process. Within the niche, myofibers, endothelial cells, perivascular cells (pericytes, smooth muscle cells), fibro-adipogenic progenitors, fibroblasts, and immune cells are in close proximity with each other. Each cell behavior, membrane profile, and secretome can interfere with muscle stem cell fate and skeletal muscle regeneration. On top of that, the muscle stem cell niche can also be modified by extra-muscle (remote) cues, as other tissues may act on muscle regeneration via the production of circulating factors or the delivery of cells. In this review, we highlight recent publications evidencing both local and remote effectors of the muscle stem cell niche.


Sujet(s)
Communication cellulaire , Muscles squelettiques , Niche de cellules souches , Animaux , Muscles squelettiques/cytologie , Muscles squelettiques/physiologie , Humains , Niche de cellules souches/physiologie , Régénération/physiologie , Développement musculaire , Différenciation cellulaire
20.
Curr Top Dev Biol ; 158: 151-177, 2024.
Article de Anglais | MEDLINE | ID: mdl-38670704

RÉSUMÉ

The process of skeletal muscle regeneration involves a coordinated interplay of specific cellular and molecular interactions within the injury site. This review provides an overview of the cellular and molecular components in regenerating skeletal muscle, focusing on how these cells or molecules in the niche regulate muscle stem cell functions. Dysfunctions of muscle stem cell-to-niche cell communications during aging and disease will also be discussed. A better understanding of how niche cells coordinate with muscle stem cells for muscle repair will greatly aid the development of therapeutic strategies for treating muscle-related disorders.


Sujet(s)
Homéostasie , Muscles squelettiques , Régénération , Niche de cellules souches , Régénération/physiologie , Humains , Muscles squelettiques/physiologie , Muscles squelettiques/cytologie , Animaux , Niche de cellules souches/physiologie , Cellules souches/cytologie , Cellules souches/physiologie , Cellules souches/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE