Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
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
2.
Nat Commun ; 14(1): 8498, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38129410

ABSTRACT

Therapeutic angiogenesis represents a promising avenue to revascularize the ischemic heart. Its limited success is partly due to our poor understanding of the cardiac stroma, specifically mural cells, and their response to ischemic injury. Here, we combine single-cell and positional transcriptomics to assess the behavior of mural cells within the healing heart. In response to myocardial infarction, mural cells adopt an altered state closely associated with the infarct and retain a distinct lineage from fibroblasts. This response is concurrent with vascular rarefaction and reduced vascular coverage by mural cells. Positional transcriptomics reveals that the infarcted heart is governed by regional-dependent and temporally regulated programs. While the remote zone acts as an important source of pro-angiogenic signals, the infarct zone is accentuated by chronic activation of anti-angiogenic, pro-fibrotic, and inflammatory cues. Together, our work unveils the spatiotemporal programs underlying cardiac repair and establishes an association between vascular deterioration and mural cell dysfunction.


Subject(s)
Microvascular Rarefaction , Myocardial Infarction , Humans , Myocardial Infarction/genetics , Myocardium , Myocytes, Cardiac , Signal Transduction
3.
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
4.
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
5.
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
6.
Development ; 149(8)2022 04 15.
Article in English | MEDLINE | ID: mdl-35333325

ABSTRACT

Eosinophils, best known for their role in anti-parasitic responses, have recently been shown to actively participate in tissue homeostasis and repair. Their regulation must be tightly controlled, as their absence or hyperplasia is associated with chronic disease (e.g. asthma or inflammatory bowel disease). In the context of skeletal muscle, eosinophils play a supportive role after acute damage. Indeed, their depletion leads to strong defects in skeletal muscle regeneration and, in the absence of eosinophil-secreted interleukin (IL) 4 and IL13, fibro-adipogenic progenitors fail to support muscle stem cell proliferation. However, the role of eosinophils in muscular dystrophy remains elusive. Although it has been shown that eosinophils are present in higher numbers in muscles from mdx mice (a mouse model for Duchenne muscular dystrophy), their depletion does not affect muscle histopathology at an early age. Here, we evaluated the impact of hyper-eosinophilia on the development of fibrofatty infiltration in aged mdx mice and found that muscle eosinophilia leads to defects in muscle homeostasis, regeneration and repair, and eventually hastens death.


Subject(s)
Muscular Dystrophy, Duchenne , Animals , Disease Models, Animal , Eosinophils/metabolism , Eosinophils/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology
7.
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
8.
Dev Cell ; 56(7): 1014-1029.e7, 2021 04 05.
Article in English | MEDLINE | ID: mdl-33735618

ABSTRACT

Negative elongation factor (NELF) is a critical transcriptional regulator that stabilizes paused RNA polymerase to permit rapid gene expression changes in response to environmental cues. Although NELF is essential for embryonic development, its role in adult stem cells remains unclear. In this study, through a muscle-stem-cell-specific deletion, we showed that NELF is required for efficient muscle regeneration and stem cell pool replenishment. In mechanistic studies using PRO-seq, single-cell trajectory analyses and myofiber cultures revealed that NELF works at a specific stage of regeneration whereby it modulates p53 signaling to permit massive expansion of muscle progenitors. Strikingly, transplantation experiments indicated that these progenitors are also necessary for stem cell pool repopulation, implying that they are able to return to quiescence. Thus, we identified a critical role for NELF in the expansion of muscle progenitors in response to injury and revealed that progenitors returning to quiescence are major contributors to the stem cell pool repopulation.


Subject(s)
Muscle, Skeletal/physiology , Satellite Cells, Skeletal Muscle/physiology , Transcription Factors/physiology , Animals , Cell Differentiation , Cells, Cultured , Eye Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Development , Nerve Growth Factors/metabolism , Regeneration/genetics , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/transplantation , Serpins/metabolism , Signal Transduction , Transcription Factors/genetics , Transcriptome , Tumor Suppressor Protein p53/metabolism
10.
PLoS One ; 14(9): e0222946, 2019.
Article in English | MEDLINE | ID: mdl-31560727

ABSTRACT

Human embryonic stem cell (hESC)-derived skeletal muscle progenitors (SMP)-defined as PAX7-expressing cells with myogenic potential-can provide an abundant source of donor material for muscle stem cell therapy. As in vitro myogenesis is decoupled from in vivo timing and 3D-embryo structure, it is important to characterize what stage or type of muscle is modeled in culture. Here, gene expression profiling is analyzed in hESCs over a 50 day skeletal myogenesis protocol and compared to datasets of other hESC-derived skeletal muscle and adult murine satellite cells. Furthermore, day 2 cultures differentiated with high or lower concentrations of CHIR99021, a GSK3A/GSK3B inhibitor, were contrasted. Expression profiling of the 50 day time course identified successively expressed gene subsets involved in mesoderm/paraxial mesoderm induction, somitogenesis, and skeletal muscle commitment/formation which could be regulated by a putative cascade of transcription factors. Initiating differentiation with higher CHIR99021 concentrations significantly increased expression of MSGN1 and TGFB-superfamily genes, notably NODAL, resulting in enhanced paraxial mesoderm and reduced ectoderm/neuronal gene expression. Comparison to adult satellite cells revealed that genes expressed in 50-day cultures correlated better with those expressed by quiescent or early activated satellite cells, which have the greatest therapeutic potential. Day 50 cultures were similar to other hESC-derived skeletal muscle and both expressed known and novel SMP surface proteins. Overall, a putative cascade of transcription factors has been identified which regulates four stages of myogenesis. Subsets of these factors were upregulated by high CHIR99021 or their binding sites were significantly over-represented during SMP activation, ranging from quiescent to late-activated stages. This analysis serves as a resource to further study the progression of in vitro skeletal myogenesis and could be mined to identify novel markers of pluripotent-derived SMPs or regulatory transcription/growth factors. Finally, 50-day hESC-derived SMPs appear similar to quiescent/early activated satellite cells, suggesting they possess therapeutic potential.


Subject(s)
Gene Expression Regulation, Developmental , Human Embryonic Stem Cells/metabolism , Muscle Development/genetics , Muscle, Skeletal/growth & development , Transcription Factors/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Gene Expression Profiling , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Muscle, Skeletal/cytology , Pyridines/pharmacology , Pyrimidines/pharmacology , Satellite Cells, Skeletal Muscle/metabolism
11.
Cell Stem Cell ; 23(5): 653-664, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30388423

ABSTRACT

Muscle stem cells, or satellite cells, are required for skeletal muscle maintenance, growth, and repair. Following satellite cell activation, several factors drive asymmetric cell division to generate a stem cell and a proliferative progenitor that forms new muscle. The balance between symmetric self-renewal and asymmetric division significantly impacts the efficiency of regeneration. In this Review, we discuss the relationship of satellite cell heterogeneity and the establishment of polarity to asymmetric division, as well as how these processes are impacted in homeostasis, aging, and disease. We also highlight therapeutic opportunities for targeting satellite cell polarity and self-renewal to stimulate muscle regeneration.


Subject(s)
Aging , Disease , Homeostasis , Muscle, Skeletal/cytology , Stem Cells/cytology , Animals , Humans , Satellite Cells, Skeletal Muscle/cytology
12.
Stem Cell Reports ; 9(5): 1573-1587, 2017 11 14.
Article in English | MEDLINE | ID: mdl-29033304

ABSTRACT

Human endothelial colony-forming cells (ECFCs) represent a promising source of adult stem cells for vascular repair, yet their regenerative capacity is limited. Here, we set out to understand the molecular mechanism restricting the repair function of ECFCs. We found that key pro-angiogenic pathways are repressed in ECFCs due to the presence of bivalent (H3K27me3/H3K4me3) epigenetic marks, which decreases the cells' regenerative potential. Importantly, ex vivo treatment with a combination of epigenetic drugs that resolves bivalent marks toward the transcriptionally active H3K4me3 state leads to the simultaneous activation of multiple pro-angiogenic signaling pathways (VEGFR, CXCR4, WNT, NOTCH, SHH). This in turn results in improved capacity of ECFCs to form capillary-like networks in vitro and in vivo. Furthermore, restoration of perfusion is accelerated upon transplantation of drug-treated ECFCs in a model of hindlimb ischemia. Thus, ex vivo treatment with epigenetic drugs increases the vascular repair properties of ECFCs through transient activation of pro-angiogenic signaling pathways.


Subject(s)
Endothelial Progenitor Cells/metabolism , Epigenesis, Genetic , Neovascularization, Physiologic , Signal Transduction , Animals , Cells, Cultured , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/transplantation , Female , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Hindlimb/blood supply , Humans , Ischemia/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Stem Cell Transplantation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism
13.
Arch Biochem Biophys ; 569: 1-9, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25659883

ABSTRACT

Sarcolemma damage and activation of various calcium channels are implicated in altered Ca(2+) homeostasis in muscle fibres of both Duchenne muscular dystrophy (DMD) sufferers and in the mdx mouse model of DMD. Previously we have demonstrated that also in mdx myoblasts extracellular nucleotides trigger elevated cytoplasmic Ca(2+) concentrations due to alterations of both ionotropic and metabotropic purinergic receptors. Here we extend these findings to show that the mdx mutation is associated with enhanced store-operated calcium entry (SOCE). Substantially increased rate of SOCE in mdx myoblasts in comparison to that in control cells correlated with significantly elevated STIM1 protein levels. These results reveal that mutation in the dystrophin-encoding Dmd gene may significantly impact cellular calcium response to metabotropic stimulation involving depletion of the intracellular calcium stores followed by activation of the store-operated calcium entry, as early as in undifferentiated myoblasts. These data are in agreement with the increasing number of reports showing that the dystrophic pathology resulting from dystrophin mutations may be developmentally regulated. Moreover, our results showing that aberrant responses to extracellular stimuli may contribute to DMD pathogenesis suggest that treatments inhibiting such responses might alter progression of this lethal disease.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Muscular Dystrophy, Duchenne/metabolism , Myoblasts, Skeletal/metabolism , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Calcium Signaling/drug effects , Cell Line , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/genetics , Myoblasts, Skeletal/drug effects , Stromal Interaction Molecule 1 , TRPC Cation Channels/metabolism , TRPC6 Cation Channel
14.
Stem Cells Dev ; 22(20): 2714-24, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23829870

ABSTRACT

With an incidence of ∼1:3,500 to 5,000 in male children, Duchenne muscular dystrophy (DMD) is an X-linked disorder in which progressive muscle degeneration occurs and affected boys usually die in their twenties or thirties. Cardiac involvement occurs in 90% of patients and heart failure accounts for up to 40% of deaths. To enable new therapeutics such as gene therapy and exon skipping to be tested in human cardiomyocytes, we produced human induced pluripotent stem cells (hiPSC) from seven patients harboring mutations across the DMD gene. Mutations were retained during differentiation and analysis indicated the cardiomyocytes showed a dystrophic gene expression profile. Antisense oligonucleotide-mediated skipping of exon 51 restored dystrophin expression to ∼30% of normal levels in hiPSC-cardiomyocytes carrying exon 47-50 or 48-50 deletions. Alternatively, delivery of a dystrophin minigene to cardiomyocytes with a deletion in exon 35 or a point mutation in exon 70 allowed expression levels similar to those seen in healthy cells. This demonstrates that DMD hiPSC-cardiomyocytes provide a novel tool to evaluate whether new therapeutics can restore dystrophin expression in the heart.


Subject(s)
Dystrophin/genetics , Gene Expression/drug effects , Induced Pluripotent Stem Cells/metabolism , Muscular Dystrophy, Duchenne/genetics , Mutation , Myocytes, Cardiac/drug effects , Oligonucleotides, Antisense/pharmacology , Base Sequence , Cell Differentiation , Child , Dystrophin/metabolism , Exons , Gene Transfer Techniques , Genetic Therapy , Humans , Induced Pluripotent Stem Cells/pathology , Male , Molecular Sequence Data , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/therapy , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Primary Cell Culture
SELECTION OF CITATIONS
SEARCH DETAIL
...