Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Article in English | MEDLINE | ID: mdl-38589640

ABSTRACT

The term 'fibroblast' often serves as a catch-all for a diverse array of mesenchymal cells, including perivascular cells, stromal progenitor cells and bona fide fibroblasts. Although phenotypically similar, these subpopulations are functionally distinct, maintaining tissue integrity and serving as local progenitor reservoirs. In response to tissue injury, these cells undergo a dynamic fibroblast-myofibroblast transition, marked by extracellular matrix secretion and contraction of actomyosin-based stress fibres. Importantly, whereas transient activation into myofibroblasts aids in tissue repair, persistent activation triggers pathological fibrosis. In this Review, we discuss the roles of mechanical cues, such as tissue stiffness and strain, alongside cell signalling pathways and extracellular matrix ligands in modulating myofibroblast activation and survival. We also highlight the role of epigenetic modifications and myofibroblast memory in physiological and pathological processes. Finally, we discuss potential strategies for therapeutically interfering with these factors and the associated signal transduction pathways to improve the outcome of dysregulated healing.

3.
Nat Immunol ; 17(7): 797-805, 2016 07.
Article in English | MEDLINE | ID: mdl-27135602

ABSTRACT

Perivascular, subdural meningeal and choroid plexus macrophages are non-parenchymal macrophages that mediate immune responses at brain boundaries. Although the origin of parenchymal microglia has recently been elucidated, much less is known about the precursors, the underlying transcriptional program and the dynamics of the other macrophages in the central nervous system (CNS). It was assumed that they have a high turnover from blood-borne monocytes. However, using parabiosis and fate-mapping approaches in mice, we found that CNS macrophages arose from hematopoietic precursors during embryonic development and established stable populations, with the notable exception of choroid plexus macrophages, which had dual origins and a shorter life span. The generation of CNS macrophages relied on the transcription factor PU.1, whereas the MYB, BATF3 and NR4A1 transcription factors were not required.


Subject(s)
Central Nervous System/immunology , Hematopoietic Stem Cells/physiology , Macrophages/physiology , Microglia/physiology , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Cell Differentiation , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Monocytes/immunology , Parabiosis , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics
4.
Proc Natl Acad Sci U S A ; 119(44): e2209976119, 2022 11.
Article in English | MEDLINE | ID: mdl-36279473

ABSTRACT

IFNγ is traditionally known as a proinflammatory cytokine with diverse roles in antimicrobial and antitumor immunity. Yet, findings regarding its sources and functions during the regeneration process following a sterile injury are conflicting. Here, we show that natural killer (NK) cells are the main source of IFNγ in regenerating muscle. Beyond this cell population, IFNγ production is limited to a small population of T cells. We further show that NK cells do not play a major role in muscle regeneration following an acute injury or in dystrophic mice. Surprisingly, the absence of IFNγ per se also has no effect on muscle regeneration following an acute injury. However, the role of IFNγ is partially unmasked when TNFα is also neutralized, suggesting a compensatory mechanism. Using transgenic mice, we showed that conditional inhibition of IFNGR1 signaling in muscle stem cells or fibro-adipogenic progenitors does not play a major role in muscle regeneration. In contrast to common belief, we found that IFNγ is not present in the early inflammatory phase of the regeneration process but rather peaks when macrophages are acquiring an anti-inflammatory phenotype. Further transcriptomic analysis suggests that IFNγ cooperates with TNFα to regulate the transition of macrophages from pro- to anti-inflammatory states. The absence of the cooperative effect of these cytokines on macrophages, however, does not result in significant regeneration impairment likely due to the presence of other compensatory mechanisms. Our findings support the arising view of IFNγ as a pleiotropic inflammatory regulator rather than an inducer of the inflammatory response.


Subject(s)
Macrophages , Tumor Necrosis Factor-alpha , Mice , Animals , Interferon-gamma , Cytokines , Regeneration , Anti-Inflammatory Agents , Muscles
5.
Exp Cell Res ; 410(1): 112947, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34822813

ABSTRACT

While the majority of healthy skeletal muscle consists of multinucleated syncytial repetitive contractile myofibers, repaired by skeletal muscle stem cells when damaged, the maintenance of muscle function also requires a range of tissue-resident stromal populations. In fact, the careful orchestration of damage response processes upon muscle injury relies heavily on stromal cell contribution for effective repair. The two main types of muscle-resident stromal cells are fibro/adipogenic progenitors and mural cells. The latter is comprised of pericytes and vascular smooth muscle cells. Recent publications identifying common markers for stromal cell populations have allowed investigating population dynamics throughout the regenerative process at a higher resolution. Mounting evidence now suggests that subpopulations with distinct roles may exist among stromal cells. In various degenerative muscle wasting conditions, critical cross-talk and spatial signalling amongst various cell populations become dysregulated. This can result in the failure to curb pathological fibro/adipogenic progenitor proliferation and propensity for laying down excessive extracellular matrix, which in turn leads to fibrotic infiltration, reduced contractile units and gradual decline in muscle function. Restoration of physiologically appropriate stromal cell function is therefore just as crucial for therapeutic targeting as the homeostatic maintenance of muscle function.


Subject(s)
Adipogenesis/genetics , Cell Differentiation/genetics , Muscle, Skeletal/metabolism , Stromal Cells/metabolism , Animals , Flow Cytometry , Gene Expression Regulation, Developmental/genetics , Humans , Muscle, Skeletal/growth & development , Pericytes/metabolism , Signal Transduction/genetics , Stem Cells/cytology , Stem Cells/metabolism
6.
J Cell Sci ; 133(12)2020 06 19.
Article in English | MEDLINE | ID: mdl-32434871

ABSTRACT

Mesenchymal stromal cells (MSCs) are multipotent progenitors essential for organogenesis, tissue homeostasis, regeneration and scar formation. Tissue injury upregulates transforming growth factor ß (TGF-ß) signaling, which modulates myofibroblast fate, extracellular matrix remodeling and fibrosis. However, the molecular determinants of MSC differentiation and survival remain poorly understood. During canonical Wnt signaling, T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription factors regulate development and stemness, but the mechanisms by which injury-induced cues modulate their expression remain underexplored. Here, we studied the cell type-specific gene expression of TCF/LEF transcription factors and, more specifically, we investigated whether damage-induced TGF-ß signaling impairs the expression and function of TCF7L2 (also known as TCF4), using several models of MSCs, including skeletal muscle fibro-adipogenic progenitors. We show that TCF/LEFs are differentially expressed and that TGF-ß reduces the expression of TCF7L2 in MSCs but not in myoblasts. We also found that the ubiquitin-proteasome system regulates TCF7L2 proteostasis and participates in TGF-ß-mediated TCF7L2 protein downregulation. Finally, we show that TGF-ß requires histone deacetylase activity to repress the expression of TCF7L2. Thus, our work reports a novel interplay between TGF-ß and canonical Wnt signaling cascades in PDGFRα+ fibroblasts and suggests that this mechanism could be targeted in tissue repair and regeneration.


Subject(s)
Transforming Growth Factor beta , Wnt Signaling Pathway , Down-Regulation , Fibroblasts/metabolism , Receptor, Platelet-Derived Growth Factor alpha , Transcription Factors , Transforming Growth Factor beta/metabolism , beta Catenin/genetics , beta Catenin/metabolism
7.
Muscle Nerve ; 66(4): 513-522, 2022 10.
Article in English | MEDLINE | ID: mdl-35859452

ABSTRACT

INTRODUCTION/AIMS: Most mouse models of muscular dystrophy (MD) show mild phenotypes, which limits the translatability of experimental therapies to patients. A growing body of evidence suggests that MD is accompanied by metabolic abnormalities that could potentially exacerbate the primary muscle wasting process. Since thermoneutral (TN) housing of mice (~30°C) has been shown to affect many metabolic parameters, particularly when combined with a Western diet (WD), our aim was to determine whether the combination of TN and WD exacerbates muscle wasting in dysferlin-deficient BLAJ mice, a common model of limb-girdle MD type 2b (LGMD2b). METHODS: The 2-mo-old wild-type (WT) and BLAJ mice were housed at TN or room temperature (RT) and fed a WD or regular chow for 9 mo. Ambulatory function, muscle histology, and protein immunoblots of skeletal muscle were assessed. RESULTS: BLAJ mice at RT and fed a chow diet showed normal ambulation function similar to WT mice, whereas 90% of BLAJ mice under WD and TN combination showed ambulatory dysfunction (p < 0.001), and an up to 4.1-fold increase in quadriceps and gastrocnemius fat infiltration. Western blotting revealed decreased autophagy marker microtubules-associated protein 1 light chain 3-B (LC3BII/LC3BI) ratio and up-regulation of protein kinase B/AKT and ribosomal protein S6 phosphorylation, suggesting inefficient cellular debris and protein clearance in TN BLAJ mice fed a WD. Male and female BLAJ mice under TN and WD combination showed heterogenous fibro-fatty infiltrate composition. DISCUSSION: TN and WD combination exacerbates rodent LGMD2b without affecting WT mice. This improves rodent modeling of human MD and helps elucidate how metabolic abnormalities may play a causal role in muscle wasting.


Subject(s)
Muscular Dystrophies, Limb-Girdle , Muscular Dystrophies , Animals , Diet, Western/adverse effects , Dysferlin/genetics , Dysferlin/metabolism , Female , Housing , Humans , Male , Mice , Muscle, Skeletal , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Dystrophies/pathology , Muscular Dystrophies, Limb-Girdle/pathology , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6/metabolism
8.
Mol Cell ; 43(4): 673-80, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21855805

ABSTRACT

Methylation of specific lysine residues in the C terminus of p53 is thought to govern p53-dependent transcription following genotoxic and oncogenic stress. In particular, Set7/9 (KMT7)-mediated monomethylation of human p53 at lysine 372 (p53K372me1) was suggested to be essential for p53 activation in human cell lines. This finding was confirmed in a Set7/9 knockout mouse model (Kurash et al., 2008). In an independent knockout mouse strain deficient in Set7/9, we have investigated its involvement in p53 regulation and find that cells from these mice are normal in their ability to induce p53-dependent transcription following genotoxic and oncogenic insults. Most importantly, we detect no impairment in canonical p53 functions in these mice, indicating that Set7/9-mediated methylation of p53 does not seem to represent a major regulatory event and does not appreciably control p53 activity in vivo.


Subject(s)
Protein Methyltransferases/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis/genetics , Cell Cycle , Cellular Senescence/genetics , Gene Expression Regulation , Histone-Lysine N-Methyltransferase , Mice , Mice, Inbred C57BL , Protein Methyltransferases/metabolism , Protein Methyltransferases/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Proc Natl Acad Sci U S A ; 111(35): 12853-8, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25136132

ABSTRACT

SET domain containing (lysine methyltransferase) 7 (SETD7) is implicated in multiple signaling and disease related pathways with a broad diversity of reported substrates. Here, we report the discovery of (R)-PFI-2-a first-in-class, potent (Ki (app) = 0.33 nM), selective, and cell-active inhibitor of the methyltransferase activity of human SETD7-and its 500-fold less active enantiomer, (S)-PFI-2. (R)-PFI-2 exhibits an unusual cofactor-dependent and substrate-competitive inhibitory mechanism by occupying the substrate peptide binding groove of SETD7, including the catalytic lysine-binding channel, and by making direct contact with the donor methyl group of the cofactor, S-adenosylmethionine. Chemoproteomics experiments using a biotinylated derivative of (R)-PFI-2 demonstrated dose-dependent competition for binding to endogenous SETD7 in MCF7 cells pretreated with (R)-PFI-2. In murine embryonic fibroblasts, (R)-PFI-2 treatment phenocopied the effects of Setd7 deficiency on Hippo pathway signaling, via modulation of the transcriptional coactivator Yes-associated protein (YAP) and regulation of YAP target genes. In confluent MCF7 cells, (R)-PFI-2 rapidly altered YAP localization, suggesting continuous and dynamic regulation of YAP by the methyltransferase activity of SETD7. These data establish (R)-PFI-2 and related compounds as a valuable tool-kit for the study of the diverse roles of SETD7 in cells and further validate protein methyltransferases as a druggable target class.


Subject(s)
Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Pyrrolidines/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology , Tetrahydroisoquinolines/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Fibroblasts/drug effects , Hippo Signaling Pathway , Histone-Lysine N-Methyltransferase/genetics , Humans , MCF-7 Cells , Methyltransferases/antagonists & inhibitors , Methyltransferases/metabolism , Mutation , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Pyrrolidines/chemistry , Structure-Activity Relationship , Sulfonamides/chemistry , Tetrahydroisoquinolines/chemistry , Transcription Factors , YAP-Signaling Proteins
10.
Biochem Biophys Res Commun ; 451(1): 148-51, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25073114

ABSTRACT

The cellular substrate underlying aberrant craniofacial connective tissue accumulation that occurs in disorders such as congenital infiltration of the face (CILF) remain elusive. Here we analyze the in vivo properties of a recently identified population of neural crest-derived CD31-:CD45-:alpha7-:Sca1+:PDGFRa+ fibro/adipogenic progenitors (NCFAPs). In serial transplantation experiments in which NCFAPs were prospectively purified and transplanted into wild type mice, NCFAPs were found to be capable of self-renewal while keeping their adipogenic potential. NCFAPs constitute the main responsive FAP fraction following acute masseter muscle damage, surpassing the number of mesoderm-derived FAPs (MFAPs) during the regenerative response. Lastly, NCFAPs differentiate into adipocytes during muscle regeneration in response to pro-adipogenic systemic cues. Altogether our data indicate that NCFAPs are a population of stem/primitive progenitor cells primarily involved in craniofacial muscle regeneration that can cause tissue degeneration when the damage co-occurs with an obesity inducing diet.


Subject(s)
Adipocytes/cytology , Craniofacial Abnormalities/pathology , Neural Crest/cytology , Stem Cells/cytology , Adipogenesis , Animals , Cell Differentiation/physiology , Mice , Mice, Transgenic , Muscle Development , Muscle, Skeletal/physiology , Regeneration , Stem Cell Transplantation
11.
Acta Neuropathol ; 128(3): 363-80, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25107477

ABSTRACT

Microglia have long been the focus of much attention due to their strong proliferative response (microgliosis) to essentially any kind of damage to the CNS. More recently, we reached the realization that these cells play specific roles in determining progression and outcomes of essentially all CNS disease. Thus, microglia has ceased to be viewed as an accessory to underlying pathologies and has now taken center stage as a therapeutic target. Here, we review how our understanding of microglia's involvement in promoting or limiting the pathogenesis of diseases such as amyotrophic lateral sclerosis, Alzheimer's disease, Huntington's disease, multiple sclerosis, X-linked adrenoleukodystrophy (X-ALD) and lysosomal storage diseases (LSD) has changed over time. While strategies to suppress the deleterious and promote the virtuous functions of microglia will undoubtedly be forthcoming, replacement of these cells has already proven its usefulness in a clinical setting. Over the past few years, we have reached the realization that microglia have a developmental origin that is distinct from that of bone marrow-derived myelomonocytic cells. Nevertheless, microglia can be replaced, in specific situations, by the progeny of hematopoietic stem cells (HSCs), pointing to a strategy to engineer the CNS environment through the transplantation of modified HSCs. Thus, microglia replacement has been successfully exploited to deliver therapeutics to the CNS in human diseases such as X-ALD and LSD. With this outlook in mind, we will discuss the evidence existing so far for microglial involvement in the pathogenesis and the therapy of specific CNS disease.


Subject(s)
Central Nervous System Diseases , Microglia/physiology , Central Nervous System Diseases/pathology , Central Nervous System Diseases/physiopathology , Central Nervous System Diseases/therapy , Humans
12.
Front Immunol ; 15: 1368142, 2024.
Article in English | MEDLINE | ID: mdl-38585275

ABSTRACT

Eosinophils are a type of granulocyte named after the presence of their eosin-stained granules. Traditionally, eosinophils have been best known to play prominent roles in anti-parasitic responses and mediating allergic reactions. Knowledge of their behaviour has expanded with time, and they are now recognized to play integral parts in the homeostasis of gastrointestinal, respiratory, skeletal muscle, adipose, and connective tissue systems. As such, they are implicated in a myriad of pathologies, and have been the target of several medical therapies. This review focuses on the lifespan of eosinophils, from their origins in the bone marrow, to their tissue-resident role. In particular, we wish to highlight the functions of eosinophils in non-mucosal tissues with skeletal muscle and the adipose tissues as examples, and to discuss the current understanding of their participation in diseased states in these tissues.


Subject(s)
Adiposity , Eosinophils , Humans , Eosinophils/pathology , Obesity/pathology
13.
Cell Stem Cell ; 31(5): 597-616, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38593798

ABSTRACT

Advances in modern medicine have enabled a rapid increase in lifespan and, consequently, have highlighted the immune system as a key driver of age-related disease. Immune regeneration therapies present exciting strategies to address age-related diseases by rebooting the host's primary lymphoid tissues or rebuilding the immune system directly via biomaterials or artificial tissue. Here, we identify important, unanswered questions regarding the safety and feasibility of these therapies. Further, we identify key design parameters that should be primary considerations guiding technology design, including timing of application, interaction with the host immune system, and functional characterization of the target patient population.


Subject(s)
Stem Cells , Humans , Stem Cells/immunology , Stem Cells/cytology , Animals , Stem Cell Transplantation , Immunity , Immune System
14.
Free Neuropathol ; 52024 Jan.
Article in English | MEDLINE | ID: mdl-38357523

ABSTRACT

Background: Fibro-adipogenic progenitors (FAP) are muscle resident mesenchymal stem cells pivotal for regulation of myofiber repair. Experimental results show in addition involvement in a range of other pathological conditions and potential for pharmacological intervention. FAP histopathology in human muscle biopsies is largely unknown, but has potential to inform translational research. Methods: CD10+ FAPs in 32 archival muscle biopsies from 8 groups (normal, dermatomyositis, inclusion body myositis (IBM), anti-synthetase syndrome, immune-mediated necrotizing myopathy (IMNM), denervation, type 2 atrophy, rhabdomyolysis) were visualized by CD10 immunohistochemistry and their histology compared. Groups are compared by semi-quantitative scoring. Results: Histological activation of endomysial CD10+ FAPs includes prominent expansion of a network of cell processes surrounding muscle fibers, as well as endomysial cell clusters evidencing proliferation. Prominence of periarteriolar processes is a notable feature in some pathologies. FAP activation is often associated with fiber degeneration/regeneration, foci of inflammation, and denervation in keeping with experimental results. Type 2 atrophy shows no evidence of FAP activation. Dermatomyositis and anti-synthetase syndrome associated myositis demonstrate diffuse activation. Conclusion: Assessment of CD10+ FAP activation is routinely possible using CD10 immunohistochemistry and demonstrates several patterns in keeping with preclinical results. Prominent expansion of FAP processes surrounding myofibers suggests enhanced interaction between myofiber/basement membranes and FAPs during activation. The presence of diffuse FAP activation in dermatomyositis biopsies unrelated to fiber repair raises the possibility of FAP activation as part of the autoimmune process. Future diagnostic applications, clinical significance and therapeutic potential remain to be elucidated.

15.
Stem Cells ; 30(6): 1152-62, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22415977

ABSTRACT

Pathologies characterized by lipomatous infiltration of craniofacial structures as well as certain forms of lipodystrophies suggest the existence of a distinct adipogenic program in the cephalic region of mammals. Using lineage tracing, we studied the origin of craniofacial adipocytes that accumulate both in cranial fat depots and during ectopic lipomatous infiltration of craniofacial muscles. We found that unlike their counterparts in limb muscle, a significant percentage of cranial adipocytes is derived from the neural crest (NC). In addition, we identified a population of NC-derived Lin(-)/α7(-)/CD34(+)/Sca-1(+) fibro/adipogenic progenitors (NC-FAPs) that resides exclusively in the mesenchyme of cephalic fat and muscle. Comparative analysis of the adipogenic potential, impact on metabolism, and contribution to the regenerative response of NC-FAPs and mesoderm-derived FAPs (M-FAPs) suggests that these cells are functionally indistinguishable. While both NC- and M-FAPs express mesenchymal markers and promyogenic cytokines upon damage-induced activation, NC-FAPs additionally express components of the NC developmental program. Furthermore, we show that craniofacial FAP composition changes with age, with young mice containing FAPs that are almost exclusively of NC origin, while NC-FAPs are progressively replaced by M-FAPs as mice age. Based on these results, we propose that in the adult, ontogenetically distinct FAPs form a diffused system reminiscent of the endothelium, which can originate from multiple developmental intermediates to seed all anatomical locations.


Subject(s)
Adipocytes/physiology , Regeneration/physiology , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cell Differentiation/physiology , Cell Lineage , Female , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
16.
STAR Protoc ; 4(4): 102638, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37831606

ABSTRACT

Efficient skeletal muscle regeneration necessitates fine-tuned coordination among multiple cell types through an intricate network of intercellular communication. We present a protocol for generation of a time-resolved cellular interactome during tissue remodeling. We describe steps for isolating distinct cell populations from skeletal muscle of adult mice after acute damage and extracting RNA from purified cells prior to the generation of RNA sequencing data. We then detail procedures for generating and deciphering a time- and lineage-resolved model of intercellular crosstalk. For complete details on the use and execution of this protocol, please refer to Groppa et al. (2023).1.


Subject(s)
Cell Communication , Muscle, Skeletal , Animals , Mice , RNA , Sequence Analysis, RNA
17.
Nat Commun ; 14(1): 8273, 2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38092736

ABSTRACT

Adult tissue-resident macrophages (RMs) are either maintained by blood monocytes or through self-renewal. While the presence of a nurturing niche is likely crucial to support the survival and function of self-renewing RMs, evidence regarding its nature is limited. Here, we identify fibro-adipogenic progenitors (FAPs) as the main source of colony-stimulating factor 1 (CSF1) in resting skeletal muscle. Using parabiosis in combination with FAP-deficient transgenic mice (PdgfrαCreERT2 × DTA) or mice lacking FAP-derived CSF1 (PdgfrαCreERT2 × Csf1flox/null), we show that local CSF1 from FAPs is required for the survival of both TIM4- monocyte-derived and TIM4+ self-renewing RMs in adult skeletal muscle. The spatial distribution and number of TIM4+ RMs coincide with those of dipeptidyl peptidase IV (DPPIV)+ FAPs, suggesting their role as CSF1-producing niche cells for self-renewing RMs. This finding identifies opportunities to precisely manipulate the function of self-renewing RMs in situ to further unravel their role in health and disease.


Subject(s)
Dipeptidyl Peptidase 4 , Receptor, Platelet-Derived Growth Factor alpha , Mice , Animals , Cell Differentiation/physiology , Dipeptidyl Peptidase 4/genetics , Adipogenesis , Muscle, Skeletal , Mice, Transgenic , Macrophages
18.
Dev Cell ; 58(6): 489-505.e7, 2023 03 27.
Article in English | MEDLINE | ID: mdl-36898377

ABSTRACT

Loss of muscle mass is a common manifestation of chronic disease. We find the canonical Wnt pathway to be activated in mesenchymal progenitors (MPs) from cancer-induced cachectic mouse muscle. Next, we induce ß-catenin transcriptional activity in murine MPs. As a result, we observe expansion of MPs in the absence of tissue damage, as well as rapid loss of muscle mass. Because MPs are present throughout the organism, we use spatially restricted CRE activation and show that the induction of tissue-resident MP activation is sufficient to induce muscle atrophy. We further identify increased expression of stromal NOGGIN and ACTIVIN-A as key drivers of atrophic processes in myofibers, and we verify their expression by MPs in cachectic muscle. Finally, we show that blocking ACTIVIN-A rescues the mass loss phenotype triggered by ß-catenin activation in MPs, confirming its key functional role and strengthening the rationale for targeting this pathway in chronic disease.


Subject(s)
Wnt Signaling Pathway , beta Catenin , Mice , Animals , beta Catenin/metabolism , Activins , Muscles/metabolism
19.
Cardiovasc Res ; 118(17): 3374-3385, 2023 01 18.
Article in English | MEDLINE | ID: mdl-35709329

ABSTRACT

AIMS: Methylation of non-histone proteins is emerging as a central regulatory mechanism in health and disease. The methyltransferase SETD7 has shown to methylate and alter the function of a variety of proteins in vitro; however, its function in the heart is poorly understood. The present study investigates the role of SETD7 in myocardial ischaemic injury. METHODS AND RESULTS: Experiments were performed in neonatal rat ventricular myocytes (NRVMs), SETD7 knockout mice (SETD7-/-) undergoing myocardial ischaemia/reperfusion (I/R) injury, left ventricular (LV) myocardial samples from patients with ischaemic cardiomyopathy (ICM), and peripheral blood mononuclear cells (PBMCs) from patients with ST-elevation MI (STEMI). We show that SETD7 is activated upon energy deprivation in cultured NRVMs and methylates the Hippo pathway effector YAP, leading to its cytosolic retention and impaired transcription of antioxidant genes manganese superoxide dismutase (MnSOD) and catalase (CAT). Such impairment of antioxidant defence was associated with mitochondrial reactive oxygen species (mtROS), organelle swelling, and apoptosis. Selective pharmacological inhibition of SETD7 by (R)-PFI-2 restored YAP nuclear localization, thus preventing mtROS, mitochondrial damage, and apoptosis in NRVMs. In mice, genetic deletion of SETD7 attenuated myocardial I/R injury, mtROS, and LV dysfunction by restoring YAP-dependent transcription of MnSOD and CAT. Moreover, in cardiomyocytes isolated from I/R mice and ICM patients, (R)-PFI-2 prevented mtROS accumulation, while improving Ca2+-activated tension. Finally, SETD7 was up-regulated in PBMCs from STEMI patients and negatively correlated with MnSOD and CAT. CONCLUSION: We show a methylation-dependent checkpoint regulating oxidative stress during myocardial ischaemia. SETD7 inhibition may represent a valid therapeutic strategy in this setting.


Subject(s)
Antioxidants , Histone-Lysine N-Methyltransferase , ST Elevation Myocardial Infarction , Animals , Mice , Rats , Apoptosis , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Leukocytes, Mononuclear/metabolism , Methylation , Myocytes, Cardiac/metabolism , ST Elevation Myocardial Infarction/metabolism , Mice, Knockout , Humans
20.
Cell Rep ; 42(2): 112051, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36729831

ABSTRACT

Efficient regeneration requires multiple cell types acting in coordination. To better understand the intercellular networks involved and how they change when regeneration fails, we profile the transcriptome of hematopoietic, stromal, myogenic, and endothelial cells over 14 days following acute muscle damage. We generate a time-resolved computational model of interactions and identify VEGFA-driven endothelial engagement as a key differentiating feature in models of successful and failed regeneration. In addition, the analysis highlights that the majority of secreted signals, including VEGFA, are simultaneously produced by multiple cell types. To test whether the cellular source of a factor determines its function, we delete VEGFA from two cell types residing in close proximity: stromal and myogenic progenitors. By comparing responses to different types of damage, we find that myogenic and stromal VEGFA have distinct functions in regeneration. This suggests that spatial compartmentalization of signaling plays a key role in intercellular communication networks.


Subject(s)
Endothelial Cells , Signal Transduction , Stem Cells/physiology , Cell Communication , Muscle, Skeletal/physiology , Cell Differentiation , Muscle Development
SELECTION OF CITATIONS
SEARCH DETAIL