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










Publication year range
2.
Nat Commun ; 13(1): 7444, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36460641

ABSTRACT

Mechanisms by which specific histone modifications regulate distinct gene networks remain little understood. We investigated how H3K79me2, a modification catalyzed by DOT1L and previously considered a general transcriptional activation mark, regulates gene expression during cardiogenesis. Embryonic cardiomyocyte ablation of Dot1l revealed that H3K79me2 does not act as a general transcriptional activator, but rather regulates highly specific transcriptional networks at two critical cardiogenic junctures: embryonic cardiogenesis, where it was particularly important for left ventricle-specific genes, and postnatal cardiomyocyte cell cycle withdrawal, with Dot1L mutants having more mononuclear cardiomyocytes and prolonged cardiomyocyte cell cycle activity. Mechanistic analyses revealed that H3K79me2 in two distinct domains, gene bodies and regulatory elements, synergized to promote expression of genes activated by DOT1L. Surprisingly, H3K79me2 in specific regulatory elements also contributed to silencing genes usually not expressed in cardiomyocytes. These results reveal mechanisms by which DOT1L successively regulates left ventricle specification and cardiomyocyte cell cycle withdrawal.


Subject(s)
Gene Regulatory Networks , Myocytes, Cardiac , Cell Division , Cell Cycle/genetics , Heart Ventricles
3.
Acta Biomater ; 152: 47-59, 2022 10 15.
Article in English | MEDLINE | ID: mdl-36041648

ABSTRACT

As the native regenerative potential of adult cardiac tissue is limited post-injury, stimulating endogenous repair mechanisms in the mammalian myocardium is a potential goal of regenerative medicine therapeutics. Injection of myocardial matrix hydrogels into the heart post-myocardial infarction (MI) has demonstrated increased cardiac muscle and promotion of pathways associated with cardiac development, suggesting potential promotion of cardiomyocyte turnover. In this study, the myocardial matrix hydrogel was shown to have native capability as an effective reactive oxygen species scavenger and protect against oxidative stress induced cell cycle inhibition in vitro. Encapsulation of cardiomyocytes demonstrated an enhanced turnover in in vitro studies, and in vivo assessments of myocardial matrix hydrogel treatment post-MI showed increased thymidine analog uptake in cardiomyocyte nuclei compared to saline controls. Overall, this study provides evidence that properties of the myocardial matrix material provide a microenvironment mitigating oxidative damage and supportive of cardiomyocytes undergoing DNA synthesis, toward possible DNA repair or cell cycle activation. STATEMENT OF SIGNIFICANCE: Loss of adult mammalian cardiomyocyte turnover is influenced by shifts in oxidative damage, which represents a potential mechanism for improving restoration of cardiac muscle after myocardial infarction (MI). Injection of a myocardial matrix hydrogel into the heart post-MI previously demonstrated increased cardiac muscle and promotion of pathways associated with cardiac development, suggesting potential in promoting proliferation of cardiomyocytes. In this study, the myocardial matrix hydrogel was shown to protect cells from oxidative stress and increase proliferation in vitro. In a rat MI model, greater presence of tissue free thiol content spared from oxidative damage, lesser mitochondrial superoxide content, and increased thymidine analog uptake in cardiomyocytes was found in matrix injected animals compared to saline controls. Overall, this study provides evidence that properties of the myocardial matrix material provide a microenvironment supportive of cardiomyocytes undergoing DNA synthesis, toward possible DNA repair or cell cycle activation.


Subject(s)
Myocardial Infarction , Myocytes, Cardiac , Animals , DNA/metabolism , Hydrogels/metabolism , Hydrogels/pharmacology , Mammals , Myocardial Infarction/metabolism , Myocardium/metabolism , Rats , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/pharmacology , Superoxides , Thymidine/metabolism , Thymidine/pharmacology
4.
Immunity ; 55(4): 606-622.e6, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35358427

ABSTRACT

Lymph node (LN) stromal cells play a crucial role in LN development and in supporting adaptive immune responses. However, their origin, differentiation pathways, and transcriptional programs are still elusive. Here, we used lineage-tracing approaches and single-cell transcriptome analyses to determine origin, transcriptional profile, and composition of LN stromal and endothelial progenitors. Our results showed that all major stromal cell subsets and a large proportion of blood endothelial cells originate from embryonic Hoxb6+ progenitors of the lateral plate mesoderm (LPM), whereas lymphatic endothelial cells arise from Pax3+ progenitors of the paraxial mesoderm (PXM). Single-cell RNA sequencing revealed the existence of different Cd34+ and Cxcl13+ stromal cell subsets and showed that embryonic LNs contain proliferating progenitors possibly representing the amplifying populations for terminally differentiated cells. Taken together, our work identifies the earliest embryonic sources of LN stromal and endothelial cells and demonstrates that stromal diversity begins already during LN development.


Subject(s)
Endothelial Cells , Endothelial Cells/metabolism , Lymph Nodes , Sequence Analysis, RNA , Single-Cell Analysis , Stromal Cells , Transcription Factors/metabolism
5.
Cell Rep ; 30(2): 571-582.e2, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31940497

ABSTRACT

Despite decades of studies suggesting that the in vivo adipocyte progenitor resides within the vascular niche, the exact nature of this progenitor remains controversial because distinct studies have attributed adipogenic properties to multiple vascular cell types. Using Cre recombinases labeling distinct vascular lineages, we conduct parallel lineage tracing experiments to assess their degree of contribution to de novo adipogenesis. Although we detect occasional adipocytes that were lineage traced by endothelial or mural recombinases, these are rare events. On the other hand, platelet-derived growth factor receptor alpha (PDGFRα)-expressing adventitial or capsular fibroblasts make a significant contribution to adipocytes in all depots and experimental settings tested. Our data also suggest that fibroblasts transition to an intermediate beige adipocyte phenotype prior to differentiating to a mature white adipocyte. These observations, together with histological analyses revealing that adipose tissue fibroblasts express the mural cell marker PDGFRß, harmonize a highly controversial field with implications for multiple human diseases, including the pandemic of obesity.


Subject(s)
Adipogenesis/genetics , Adipose Tissue/metabolism , Cell Lineage/physiology , Fibroblasts/metabolism , Obesity/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation , Humans
6.
JCI Insight ; 4(22)2019 11 14.
Article in English | MEDLINE | ID: mdl-31723062

ABSTRACT

Aging is a major risk factor for cardiovascular disease. Although the impact of aging has been extensively studied, little is known regarding the aging processes in cells of the heart. Here we analyzed the transcriptomes of hearts of 12-week-old and 18-month-old mice by single-nucleus RNA-sequencing. Among all cell types, aged fibroblasts showed most significant differential gene expression, increased RNA dynamics, and network entropy. Aged fibroblasts exhibited significantly changed expression patterns of inflammatory, extracellular matrix organization angiogenesis, and osteogenic genes. Functional analyses indicated deterioration of paracrine signatures between fibroblasts and endothelial cells in old hearts. Aged heart-derived fibroblasts had impaired endothelial cell angiogenesis and autophagy and augmented proinflammatory response. In particular, expression of Serpine1 and Serpine2 were significantly increased and secreted by old fibroblasts to exert antiangiogenic effects on endothelial cells, an effect that could be significantly prevented by using neutralizing antibodies. Moreover, we found an enlarged subpopulation of aged fibroblasts expressing osteoblast genes in the epicardial layer associated with increased calcification. Taken together this study provides system-wide insights and identifies molecular changes of aging cardiac fibroblasts, which may contribute to declined heart function.


Subject(s)
Aging/physiology , Fibroblasts , Heart/physiology , Myocardium/cytology , Transcriptome , Animals , Fibroblasts/chemistry , Fibroblasts/metabolism , Fibroblasts/physiology , Male , Mice , Serpins/genetics , Serpins/metabolism , Transcriptome/genetics , Transcriptome/physiology , Vascular Calcification/genetics , Vascular Calcification/metabolism
7.
Sci Transl Med ; 11(511)2019 09 25.
Article in English | MEDLINE | ID: mdl-31554736

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease where invasive pulmonary myofibroblasts secrete collagen and destroy lung integrity. Here, we show that interleukin-11 (IL11) is up-regulated in the lung of patients with IPF, associated with disease severity, and IL-11 is secreted from IPF fibroblasts. In vitro, IL-11 stimulates lung fibroblasts to become invasive actin alpha 2, smooth muscle-positive (ACTA2+), collagen-secreting myofibroblasts in an extracellular signal-regulated kinase (ERK)-dependent, posttranscriptional manner. In mice, fibroblast-specific transgenic expression or administration of murine IL-11 induces lung myofibroblasts and causes lung fibrosis. IL-11 receptor subunit alpha-1 (Il11ra1)-deleted mice, whose lung fibroblasts are unresponsive to profibrotic stimulation, are protected from fibrosis in the bleomycin mouse model of pulmonary fibrosis. We generated an IL-11-neutralizing antibody that blocks lung fibroblast activation downstream of multiple stimuli and reverses myofibroblast activation. In therapeutic studies, anti-IL-11 treatment diminished lung inflammation and reversed lung fibrosis while inhibiting ERK and SMAD activation in mice. These data prioritize IL-11 as a drug target for lung fibrosis and IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis/drug therapy , Interleukin-11/therapeutic use , Animals , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Bleomycin , Fibroblasts/pathology , Humans , Idiopathic Pulmonary Fibrosis/pathology , Interleukin-11 Receptor alpha Subunit/metabolism , Lung/pathology , Mice, Knockout , Severity of Illness Index , Signal Transduction , Up-Regulation
8.
Circulation ; 140(1): 55-66, 2019 07 02.
Article in English | MEDLINE | ID: mdl-30982350

ABSTRACT

BACKGROUND: Membrane contact sites are fundamental for transmission and translation of signals in multicellular organisms. The junctional membrane complexes in the cardiac dyads, where transverse (T) tubules are juxtaposed to the sarcoplasmic reticulum, are a prime example. T-tubule uncoupling and remodeling are well-known features of cardiac disease and heart failure. Even subtle alterations in the association between T-tubules and the junctional sarcoplasmic reticulum can cause serious cardiac disorders. NEXN (nexilin) has been identified as an actin-binding protein, and multiple mutations in the NEXN gene are associated with cardiac diseases, but the precise role of NEXN in heart function and disease is still unknown. METHODS: Nexn global and cardiomyocyte-specific knockout mice were generated. Comprehensive phenotypic and RNA sequencing and mass spectrometry analyses were performed. Heart tissue samples and isolated single cardiomyocytes were analyzed by electron and confocal microscopy. RESULTS: Global and cardiomyocyte-specific loss of Nexn in mice resulted in a rapidly progressive dilated cardiomyopathy. In vivo and in vitro analyses revealed that NEXN interacted with junctional sarcoplasmic reticulum proteins, was essential for optimal calcium transients, and was required for initiation of T-tubule invagination and formation. CONCLUSIONS: These results demonstrated that NEXN is a pivotal component of the junctional membrane complex and is required for initiation and formation of T-tubules, thus providing insight into mechanisms underlying cardiomyopathy in patients with mutations in NEXN.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Cell Membrane/metabolism , Intercellular Junctions/metabolism , Microfilament Proteins/deficiency , Muscle Fibers, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Animals , Calcium Channels, L-Type/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cell Membrane/genetics , Cell Membrane/pathology , Cells, Cultured , Intercellular Junctions/genetics , Intercellular Junctions/pathology , Mice , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/genetics , Muscle Fibers, Skeletal/pathology , Myocytes, Cardiac/pathology
9.
Circ Res ; 122(4): 583-590, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29269349

ABSTRACT

RATIONALE: Myocardial infarction is a major cause of adult mortality worldwide. The origin(s) of cardiac fibroblasts that constitute the postinfarct scar remain controversial, in particular the potential contribution of bone marrow lineages to activated fibroblasts within the scar. OBJECTIVE: The aim of this study was to establish the origin(s) of infarct fibroblasts using lineage tracing and bone marrow transplants and a robust marker for cardiac fibroblasts, the Collagen1a1-green fluorescent protein reporter. METHODS AND RESULTS: Using genetic lineage tracing or bone marrow transplant, we found no evidence for collagen-producing fibroblasts derived from hematopoietic or bone marrow lineages in hearts subjected to permanent left anterior descending coronary artery ligation. In fact, fibroblasts within the infarcted area were largely of epicardial origin. Intriguingly, collagen-producing fibrocytes from hematopoietic lineages were observed attached to the epicardial surface of infarcted and sham-operated hearts in which a suture was placed around the left anterior descending coronary artery. CONCLUSIONS: In this controversial field, our study demonstrated that the vast majority of infarct fibroblasts were of epicardial origin and not derived from bone marrow lineages, endothelial-to-mesenchymal transition, or blood. We also noted the presence of collagen-producing fibrocytes on the epicardial surface that resulted at least in part from the surgical procedure.


Subject(s)
Bone Marrow Cells/cytology , Cell Lineage , Myocardial Infarction/therapy , Myofibroblasts/cytology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation/adverse effects , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Pericardium/cytology
10.
Cardiovasc Res ; 114(3): 378-388, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29040414

ABSTRACT

AIMS: Luma is a recently discovered, evolutionarily conserved protein expressed in mammalian heart, which is associated with the LInker of Nucleoskeleton and Cytoskeleton (LINC) complex. The LINC complex structurally integrates the nucleus and the cytoplasm and plays a critical role in mechanotransduction across the nuclear envelope. Mutations in several LINC components in both humans and mice result in various cardiomyopathies, implying they play essential, non-redundant roles. A single amino acid substitution of serine 358 to leucine (S358L) in Luma is the unequivocal cause of a distinct form of arrhythmogenic cardiomyopathy. However, the role of Luma in heart has remained obscure. In addition, it also remains to be determined how the S358L mutation in Luma leads to cardiomyopathy. METHODS AND RESULTS: To determine the role of Luma in the heart, we first determined the expression pattern of Luma in mouse heart. Luma was sporadically expressed in cardiomyocytes throughout the heart, but was highly and uniformly expressed in cardiac fibroblasts and vascular smooth muscle cells. We also generated germline null Luma mice and discovered that germline null mutants were viable and exhibited normal cardiac function. Luma null mice also responded normally to pressure overload induced by transverse aortic constriction. In addition, localization and expression of other LINC complex components in both cardiac myocytes and fibroblasts was unaffected by global loss of Luma. Furthermore, we also generated and characterized Luma S358L knock-in mice, which displayed normal cardiac function and morphology. CONCLUSION: Our data suggest that Luma is dispensable for murine cardiac development and function and that the Luma S358L mutation alone may not cause cardiomyopathy in mice.


Subject(s)
Heart/embryology , Membrane Proteins/metabolism , Myocardium/metabolism , Animals , Arrhythmogenic Right Ventricular Dysplasia/genetics , Arrhythmogenic Right Ventricular Dysplasia/metabolism , Cells, Cultured , Cytoskeleton/metabolism , Female , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Heart/physiopathology , Humans , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Male , Mechanotransduction, Cellular , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/metabolism , Nuclear Matrix/metabolism
11.
Nature ; 552(7683): 110-115, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29160304

ABSTRACT

Fibrosis is a common pathology in cardiovascular disease. In the heart, fibrosis causes mechanical and electrical dysfunction and in the kidney, it predicts the onset of renal failure. Transforming growth factor ß1 (TGFß1) is the principal pro-fibrotic factor, but its inhibition is associated with side effects due to its pleiotropic roles. We hypothesized that downstream effectors of TGFß1 in fibroblasts could be attractive therapeutic targets and lack upstream toxicity. Here we show, using integrated imaging-genomics analyses of primary human fibroblasts, that upregulation of interleukin-11 (IL-11) is the dominant transcriptional response to TGFß1 exposure and required for its pro-fibrotic effect. IL-11 and its receptor (IL11RA) are expressed specifically in fibroblasts, in which they drive non-canonical, ERK-dependent autocrine signalling that is required for fibrogenic protein synthesis. In mice, fibroblast-specific Il11 transgene expression or Il-11 injection causes heart and kidney fibrosis and organ failure, whereas genetic deletion of Il11ra1 protects against disease. Therefore, inhibition of IL-11 prevents fibroblast activation across organs and species in response to a range of important pro-fibrotic stimuli. These results reveal a central role of IL-11 in fibrosis and we propose that inhibition of IL-11 is a potential therapeutic strategy to treat fibrotic diseases.


Subject(s)
Cardiovascular System/metabolism , Cardiovascular System/pathology , Fibrosis/metabolism , Fibrosis/pathology , Interleukin-11/metabolism , Animals , Autocrine Communication , Cells, Cultured , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/chemically induced , Heart , Humans , Interleukin-11/antagonists & inhibitors , Interleukin-11/genetics , Interleukin-11 Receptor alpha Subunit/deficiency , Interleukin-11 Receptor alpha Subunit/genetics , Kidney/pathology , Male , Mice , Mice, Knockout , Middle Aged , Myocardium/metabolism , Myocardium/pathology , Organ Dysfunction Scores , Protein Biosynthesis , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Transgenes/genetics
12.
Cell Stem Cell ; 20(5): 587-589, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28475883

ABSTRACT

In their reply, Sylvia Evans and colleagues argue that their lineage-labeling approach gives more precise tracing of the lineage contribution of mural cells in vivo than previous versions.


Subject(s)
Adipocytes/cytology , Adventitia/cytology , Stem Cells/cytology , Adipogenesis/genetics , Adipogenesis/physiology , Adipose Tissue, White/cytology , Animals , Fibroblasts/cytology , Flow Cytometry , Male , Pericytes/cytology
13.
Cell Stem Cell ; 20(3): 345-359.e5, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28111199

ABSTRACT

Pericytes are widely believed to function as mesenchymal stem cells (MSCs), multipotent tissue-resident progenitors with great potential for regenerative medicine. Cultured pericytes isolated from distinct tissues can differentiate into multiple cell types in vitro or following transplantation in vivo. However, the cell fate plasticity of endogenous pericytes in vivo remains unclear. Here, we show that the transcription factor Tbx18 selectively marks pericytes and vascular smooth muscle cells in multiple organs of adult mouse. Fluorescence-activated cell sorting (FACS)-purified Tbx18-expressing cells behaved as MSCs in vitro. However, lineage-tracing experiments using an inducible Tbx18-CreERT2 line revealed that pericytes and vascular smooth muscle cells maintained their identity in aging and diverse pathological settings and did not significantly contribute to other cell lineages. These results challenge the current view of endogenous pericytes as multipotent tissue-resident progenitors and suggest that the plasticity observed in vitro or following transplantation in vivo arises from artificial cell manipulations ex vivo.


Subject(s)
Mesenchymal Stem Cells/cytology , Organ Specificity , Pericytes/cytology , Adipocytes/cytology , Aging/genetics , Cell Lineage , Cicatrix/pathology , Fibroblasts/cytology , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/metabolism , Humans , Integrases/metabolism , Mesenchymal Stem Cells/metabolism , Muscle Development , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Neurons/cytology , Pericytes/metabolism , Phenotype , Receptor, Platelet-Derived Growth Factor beta/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
14.
Dev Cell ; 39(4): 392-394, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27875683

ABSTRACT

Adult cardiomyocytes are largely thought to lack proliferative and therefore regenerative potential. Reporting in Nature, Nakada et al. (2016) find that a hypoxic regime reduces mitochondrial metabolism and promotes proliferation in adult mouse cardiomyocytes, resulting in increased regeneration following myocardial infarction. These findings suggest the potential to transform post-MI care.


Subject(s)
Myocardial Infarction , Myocytes, Cardiac , Animals , Hypoxia , Mice , Oxidation-Reduction , Regeneration
15.
PLoS One ; 11(4): e0154413, 2016.
Article in English | MEDLINE | ID: mdl-27120339

ABSTRACT

The T-box transcription factor TBX18 is essential to mesenchymal cell differentiation in several tissues and Tbx18 loss-of-function results in dramatic organ malformations and perinatal lethality. Here we demonstrate for the first time that Tbx18 is required for the normal development of periductal smooth muscle stromal cells in prostate, particularly in the anterior lobe, with a clear impact on prostate health in adult mice. Prostate abnormalities are only subtly apparent in Tbx18 mutants at birth; to examine postnatal prostate development we utilized a relatively long-lived hypomorphic mutant and a novel conditional Tbx18 allele. Similar to the ureter, cells that fail to express Tbx18 do not condense normally into smooth muscle cells of the periductal prostatic stroma. However, in contrast to ureter, the periductal stromal cells in mutant prostate assume a hypertrophic, myofibroblastic state and the adjacent epithelium becomes grossly disorganized. To identify molecular events preceding the onset of this pathology, we compared gene expression in the urogenital sinus (UGS), from which the prostate develops, in Tbx18-null and wild type littermates at two embryonic stages. Genes that regulate cell proliferation, smooth muscle differentiation, prostate epithelium development, and inflammatory response were significantly dysregulated in the mutant urogenital sinus around the time that Tbx18 is first expressed in the wild type UGS, suggesting a direct role in regulating those genes. Together, these results argue that Tbx18 is essential to the differentiation and maintenance of the prostate periurethral mesenchyme and that it indirectly regulates epithelial differentiation through control of stromal-epithelial signaling.


Subject(s)
Gene Expression Regulation, Developmental , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Prostate/metabolism , Stromal Cells/metabolism , T-Box Domain Proteins/genetics , Alleles , Animals , Cell Communication , Cell Differentiation , Cell Proliferation , Ejaculatory Ducts/growth & development , Ejaculatory Ducts/metabolism , Ejaculatory Ducts/pathology , Embryo, Mammalian , Gene Expression Profiling , Male , Mice , Muscle, Smooth/growth & development , Muscle, Smooth/pathology , Myocytes, Smooth Muscle/pathology , Organogenesis/genetics , Prostate/growth & development , Prostate/pathology , Signal Transduction , Stromal Cells/pathology , T-Box Domain Proteins/deficiency , Ureter/growth & development , Ureter/metabolism , Ureter/pathology
16.
J Mol Med (Berl) ; 93(8): 823-30, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26169532

ABSTRACT

Cardiac fibroblasts are a major cell population of the heart and are characterized by their capacity to produce extracellular matrix (ECM). In hearts subjected to pressure overload, excessive fibroblast accumulation is responsible for fibrosis of the myocardium, a major clinical issue. Hence, understanding mechanisms generating fibroblasts in this context has become a key question in the cardiovascular field. Recent studies now point to the activation of resident fibroblasts as the underlying cause of fibrosis. However, de novo generation of fibroblasts from endothelium and circulating hematopoietic cells has also been proposed to significantly contribute to fibrosis. Here, we discuss the latest findings on fibroblast origins, with a particular emphasis on the pressure overload model, and the implication of these findings for the development of anti-fibrotic therapies that are currently lacking.


Subject(s)
Fibroblasts/cytology , Fibroblasts/pathology , Heart Failure/pathology , Myocardium/cytology , Myocardium/pathology , Animals , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibroblasts/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Heart/embryology , Heart Failure/metabolism , Humans , Myocardium/metabolism
17.
Dev Cell ; 33(5): 507-21, 2015 Jun 08.
Article in English | MEDLINE | ID: mdl-26028220

ABSTRACT

Transcriptional mediators of cell stress pathways, including HIF1α, ATF4, and p53, are key to normal development and play critical roles in disease, including ischemia and cancer. Despite their importance, mechanisms by which pathways mediated by these transcription factors interact with one another are not fully understood. In addressing the controversial role of HIF1α in cardiomyocytes (CMs) during heart development, we discovered a mid-gestational requirement for HIF1α for proliferation of hypoxic CMs, involving metabolic switching and a complex interplay among HIF1α, ATF4, and p53. Loss of HIF1α resulted in activation of ATF4 and p53, the latter inhibiting CM proliferation. Bioinformatic and biochemical analyses revealed unexpected mechanisms by which HIF1α intersects with ATF4 and p53 pathways. Our results highlight previously undescribed roles of HIF1α and interactions among major cell stress pathways that could be targeted to enhance proliferation of CMs in ischemia and may have relevance to other diseases, including cancer.


Subject(s)
Activating Transcription Factor 4/metabolism , Cell Proliferation , Embryo, Mammalian/cytology , Fetus/cytology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Hypoxia/physiopathology , Myocytes, Cardiac/cytology , Tumor Suppressor Protein p53/metabolism , Activating Transcription Factor 4/genetics , Animals , Biomarkers/metabolism , Blotting, Western , Cells, Cultured , Embryo, Mammalian/metabolism , Female , Fetus/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Immunoenzyme Techniques , Male , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Suppressor Protein p53/genetics
18.
J Clin Invest ; 124(7): 2921-34, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24937432

ABSTRACT

Activation and accumulation of cardiac fibroblasts, which result in excessive extracellular matrix deposition and consequent mechanical stiffness, myocyte uncoupling, and ischemia, are key contributors to heart failure progression. Recently, endothelial-to-mesenchymal transition (EndoMT) and the recruitment of circulating hematopoietic progenitors to the heart have been reported to generate substantial numbers of cardiac fibroblasts in response to pressure overload-induced injury; therefore, these processes are widely considered to be promising therapeutic targets. Here, using multiple independent murine Cre lines and a collagen1a1-GFP fusion reporter, which specifically labels fibroblasts, we found that following pressure overload, fibroblasts were not derived from hematopoietic cells, EndoMT, or epicardial epithelial-to-mesenchymal transition. Instead, pressure overload promoted comparable proliferation and activation of two resident fibroblast lineages, including a previously described epicardial population and a population of endothelial origin. Together, these data present a paradigm for the origins of cardiac fibroblasts during development and in fibrosis. Furthermore, these data indicate that therapeutic strategies for reducing pathogenic cardiac fibroblasts should shift from targeting presumptive EndoMT or infiltrating hematopoietically derived fibroblasts, toward common pathways upregulated in two endogenous fibroblast populations.


Subject(s)
Heart Failure/pathology , Myocardium/pathology , Animals , Biomarkers/metabolism , Blood Pressure , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cell Lineage , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Endocardium/metabolism , Endocardium/pathology , Epithelial-Mesenchymal Transition , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Gene Expression Profiling , Heart Failure/etiology , Heart Failure/physiopathology , Male , Mice , Mice, Knockout , Mice, Transgenic , Myocardium/metabolism , Pericardium/metabolism , Pericardium/pathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
19.
Dev Dyn ; 241(4): 675-83, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22354863

ABSTRACT

Deterioration of the intervertebral discs is an unfortunate consequence of aging. The intervertebral disc in mammals is composed of three parts: a jelly-like center called the nucleus pulposus, the cartilaginous annulus fibrosus, and anterior and posterior endplates that attach the discs to vertebrae. To understand the origin of the disc, we have investigated the intervertebral region of chickens. Surprisingly, our comparison of mouse and chicken discs revealed that chicken discs lack nuclei pulposi. In addition, the notochord, which in mice forms nuclei pulposi, was found to persist as a rod-like structure and express Shh throughout chicken embryogenesis. Our fate mapping data indicate that cells originating from the rostral half of each somite are responsible for forming the avian disc while cells in the caudal region of each somite form vertebrae. A histological analysis of mammalian and nonmammalian organisms suggests that nuclei pulposi are only present in mammals.


Subject(s)
Chickens/anatomy & histology , Intervertebral Disc , Animals , Biological Evolution , Intervertebral Disc/anatomy & histology , Intervertebral Disc/cytology , Mice
20.
Dev Cell ; 16(6): 822-32, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19531353

ABSTRACT

Adult skeletal muscle possesses a remarkable regenerative capacity, due to the presence of satellite cells, adult muscle stem cells. We used fate-mapping techniques in avian and mouse models to show that trunk (Pax3(+)) and cranial (MesP1(+)) skeletal muscle and satellite cells derive from separate genetic lineages. Similar lineage heterogeneity is seen within the head musculature and satellite cells, due to their shared, heterogenic embryonic origins. Lineage tracing experiments with Isl1Cre mice demonstrated the robust contribution of Isl1(+) cells to distinct jaw muscle-derived satellite cells. Transplantation of myofiber-associated, Isl1-derived satellite cells into damaged limb muscle contributed to muscle regeneration. In vitro experiments demonstrated the cardiogenic nature of cranial- but not trunk-derived satellite cells. Finally, overexpression of Isl1 in the branchiomeric muscles of chick embryos inhibited skeletal muscle differentiation in vitro and in vivo, suggesting that this gene plays a role in the specification of cardiovascular and skeletal muscle stem cell progenitors.


Subject(s)
Cell Lineage , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cells, Cultured , Chick Embryo , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Extremities/pathology , Gene Expression Regulation, Developmental , Genetic Heterogeneity , Head , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins , Mesoderm/cytology , Mesoderm/transplantation , Mice , Muscle Development , Muscles/pathology , Muscles/physiology , Myocardium/metabolism , PAX3 Transcription Factor , Paired Box Transcription Factors/metabolism , Quail/embryology , Regeneration , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...