Go to The Journal of Clinical Investigation
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Transfers
  • Advertising
  • Job board
  • Contact
  • Current issue
  • Past issues
  • By specialty
    • COVID-19
    • Cardiology
    • Immunology
    • Metabolism
    • Nephrology
    • Oncology
    • Pulmonology
    • All ...
  • Videos
  • Collections
    • Resource and Technical Advances
    • Clinical Medicine
    • Reviews
    • Editorials
    • Perspectives
    • Top read articles
  • JCI This Month
    • Current issue
    • Past issues

  • Current issue
  • Past issues
  • Specialties
  • In-Press Preview
  • Concise Communication
  • Editorials
  • Viewpoint
  • Top read articles
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Transfers
  • Advertising
  • Job board
  • Contact

Stem cells

  • 63 Articles
  • 0 Posts
  • ← Previous
  • 1
  • 2
  • 3
  • 4
  • 5
  • 6
  • 7
  • Next →
Smooth muscle-derived progenitor cell myofibroblast differentiation through KLF4 downregulation promotes arterial remodeling and fibrosis
Sizhao Lu, … , Mark W. Majesky, Mary C.M. Weiser-Evans
Sizhao Lu, … , Mark W. Majesky, Mary C.M. Weiser-Evans
Published October 29, 2020
Citation Information: JCI Insight. 2020. https://doi.org/10.1172/jci.insight.139445.
View: Text | PDF

Smooth muscle-derived progenitor cell myofibroblast differentiation through KLF4 downregulation promotes arterial remodeling and fibrosis

  • Text
  • PDF
Abstract

Resident vascular adventitial SCA1(+) progenitor (AdvSca1) cells are essential in vascular development and injury. However, the heterogeneity of AdvSca1 cells presents a unique challenge in understanding signaling pathways orchestrating their behavior in homeostasis and injury responses. Using smooth muscle cell (SMC) lineage tracing models, we identified a subpopulation of AdvSca1 cells (AdvSca1-SM) originating from mature SMCs that undergo reprogramming in situ and exhibit a multipotent phenotype. Here we employed lineage tracing and RNA sequencing to define the signaling pathways regulating SMC-to-AdvSca1-SM cell reprogramming and AdvSca1-SM progenitor cell phenotype. Unbiased hierarchical clustering revealed that genes related to hedgehog/WNT/beta-catenin signaling are significantly enriched in AdvSca1-SM cells, emphasizing the importance of this signaling axis in the reprogramming event. Leveraging AdvSca1-SM-specific expression of Gli1, we generated Gli1-CreERT2-ROSA26-YFP reporter mice to selectively track AdvSca1-SM cells. We demonstrated that physiologically relevant vascular injury or AdvSca1-SM cell-specific Klf4 depletion facilitated the proliferation and differentiation of AdvSca1-SM cells to a pro-fibrotic myofibroblast phenotype rather than macrophages. Surprisingly, AdvSca1-SM cells selectively contributed to adventitial remodeling and fibrosis, but little to neointima formation. Together, these findings strongly support therapeutics aimed at preserving the AdvSca1-SM cell phenotype as a viable anti-fibrotic approach.

Authors

Sizhao Lu, Austin J. Jolly, Keith A. Strand, Allison M. Dubner, Marie F. Mutryn, Karen S. Moulton, Raphael A. Nemenoff, Mark W. Majesky, Mary C.M. Weiser-Evans

×

PDGFRA in vascular adventitial MSCs promotes neointima formation in arteriovenous fistula in chronic kidney disease
Ke Song, … , Luan Truong, Jizhong Cheng
Ke Song, … , Luan Truong, Jizhong Cheng
Published October 1, 2020
Citation Information: JCI Insight. 2020. https://doi.org/10.1172/jci.insight.137298.
View: Text | PDF

PDGFRA in vascular adventitial MSCs promotes neointima formation in arteriovenous fistula in chronic kidney disease

  • Text
  • PDF
Abstract

Chronic kidney disease (CKD) induces the failure of arteriovenous fistulas (AVF) and promotes the differentiation of vascular adventitial GLI1+ mesenchymal stem cells (GMCs). However, the roles of GMCs in forming neointima in AVFs remains unknown. GMCs isolated from CKD mice showed increased potential capacity of differentiation into myofibroblast-like cells. Increased activation of expression of PDGFRA and hedgehog (HH) signaling were detected in adventitial cells of AVFs from ESRD patients and CKD mice. PDGFRA was translocated and accumulated in early endosome when hedgehog signaling stimulates was activated. In endosome, PDGFRA mediated activation of TGFB1/SMAD signaling promoting GMCs differentiation into myofibroblast, extracellular matrix deposition, and vascular fibrosis. These responses resulted in neointima formation and AVF failure. Knockout (KO) of Pdgfra or inhibition of HH signaling in GMCs suppressed the differentiation of GMCs into myofibroblasts. In vivo, specific KO of Pdgfra inhibited GMC activation and vascular fibrosis, resulting in suppression of neointima formation and improvement of AVF patency despite CKD. Our findings could yield strategies for maintaining AVF functions.

Authors

Ke Song, Ying Qing, Qunying Guo, Eric K. Peden, Changyi Chen, William E. Mitch, Luan Truong, Jizhong Cheng

×

Elevating EGFR-MAPK program by a non-conventional Cdc42 enhances intestinal epithelial survival and regeneration
Xiao Zhang, … , Ivaylo I. Ivanov, Nan Gao
Xiao Zhang, … , Ivaylo I. Ivanov, Nan Gao
Published July 20, 2020
Citation Information: JCI Insight. 2020. https://doi.org/10.1172/jci.insight.135923.
View: Text | PDF

Elevating EGFR-MAPK program by a non-conventional Cdc42 enhances intestinal epithelial survival and regeneration

  • Text
  • PDF
Abstract

The regulatory mechanisms enabling the intestinal epithelium to maintain a high degree of regenerative capacity during mucosal injury remain unclear. Ex vivo survival and clonogenicity of intestinal stem cells (ISCs) strictly required Cdc42-mediated growth response and Cdc42-deficient enteroids undergo rapid apoptosis. Mechanistically, Cdc42 engaging with EGFR was required for EGF-stimulated receptor-mediated endocytosis and sufficient to promote MAPK signaling. Proteomics and kinase analysis revealed that a physiological, but non-conventionally, spliced Cdc42 variant 2 (V2), exhibited stronger MAPK-activating capability. Human CDC42-V2 is transcriptionally elevated in some colon tumor tissues. Accordingly, mice engineered to overexpress Cdc42-V2 in intestinal epithelium showed elevated MAPK signaling, enhanced regeneration, and reduced mucosal damage in response to irradiation. Overproducing Cdc42-V2 specifically in mouse ISCs enhanced intestinal regeneration following injury. Thus, the intrinsic Cdc42-MAPK program is required for intestinal epithelial regeneration while elevating this signaling cascade is capable of initiating protection from genotoxic injury.

Authors

Xiao Zhang, Sheila Bandyopadhyay, Leandro P. Araujo, Kevin Tong, Juan Flores, Daniel Laubitz, Yanlin Zhao, George Yap, Jingren Wang, Qingze Zou, Ronaldo P. Ferraris, Lanjing Zhang, Wenwei Hu, Edward M. Bonder, Pawel R. Kiela, Robert J. Coffey, Michael Verzi, Ivaylo I. Ivanov, Nan Gao

×

MYSM1 maintains ribosomal protein gene expression in hematopoietic stem cells to prevent hematopoietic dysfunction
Jad I. Belle, … , David Langlais, Anastasia Nijnik
Jad I. Belle, … , David Langlais, Anastasia Nijnik
Published July 9, 2020
Citation Information: JCI Insight. 2020;5(13):e125690. https://doi.org/10.1172/jci.insight.125690.
View: Text | PDF

MYSM1 maintains ribosomal protein gene expression in hematopoietic stem cells to prevent hematopoietic dysfunction

  • Text
  • PDF
Abstract

Ribosomopathies are congenital disorders caused by mutations in the genes encoding ribosomal and other functionally related proteins. They are characterized by anemia, other hematopoietic and developmental abnormalities, and p53 activation. Ribosome assembly requires coordinated expression of many ribosomal protein (RP) genes; however, the regulation of RP gene expression, especially in hematopoietic stem cells (HSCs), remains poorly understood. MYSM1 is a transcriptional regulator essential for HSC function and hematopoiesis. We established that HSC dysfunction in Mysm1 deficiency is driven by p53; however, the mechanisms of p53 activation remained unclear. Here, we describe the transcriptome of Mysm1-deficient mouse HSCs and identify MYSM1 genome-wide DNA binding sites. We establish a direct role for MYSM1 in RP gene expression and show a reduction in protein synthesis in Mysm1–/– HSCs. Loss of p53 in mice fully rescues Mysm1–/– anemia phenotype but not RP gene expression, indicating that RP gene dysregulation is a direct outcome of Mysm1 deficiency and an upstream mediator of Mysm1–/– phenotypes through p53 activation. We characterize a patient with a homozygous nonsense MYSM1 gene variant, and we demonstrate reduced protein synthesis and increased p53 levels in patient hematopoietic cells. Our work provides insights into the specialized mechanisms regulating RP gene expression in HSCs and establishes a common etiology of MYSM1 deficiency and ribosomopathy syndromes.

Authors

Jad I. Belle, HanChen Wang, Amanda Fiore, Jessica C. Petrov, Yun Hsiao Lin, Chu-Han Feng, Thi Tuyet Mai Nguyen, Jacky Tung, Philippe M. Campeau, Uta Behrends, Theresa Brunet, Gloria Sarah Leszinski, Philippe Gros, David Langlais, Anastasia Nijnik

×

Selective LXR agonist DMHCA corrects retinal and bone marrow dysfunction in type 2 diabetes
Cristiano P. Vieira, … , Julia V. Busik, Maria B. Grant
Cristiano P. Vieira, … , Julia V. Busik, Maria B. Grant
Published July 9, 2020
Citation Information: JCI Insight. 2020;5(13):e137230. https://doi.org/10.1172/jci.insight.137230.
View: Text | PDF

Selective LXR agonist DMHCA corrects retinal and bone marrow dysfunction in type 2 diabetes

  • Text
  • PDF
Abstract

In diabetic dyslipidemia, cholesterol accumulates in the plasma membrane, decreasing fluidity and thereby suppressing the ability of cells to transduce ligand-activated signaling pathways. Liver X receptors (LXRs) make up the main cellular mechanism by which intracellular cholesterol is regulated and play important roles in inflammation and disease pathogenesis. N, N-dimethyl-3β-hydroxy-cholenamide (DMHCA), a selective LXR agonist, specifically activates the cholesterol efflux arm of the LXR pathway without stimulating triglyceride synthesis. In this study, we use a multisystem approach to understand the effects and molecular mechanisms of DMHCA treatment in type 2 diabetic (db/db) mice and human circulating angiogenic cells (CACs), which are hematopoietic progenitor cells with vascular reparative capacity. We found that DMHCA is sufficient to correct retinal and BM dysfunction in diabetes, thereby restoring retinal structure, function, and cholesterol homeostasis; rejuvenating membrane fluidity in CACs; hampering systemic inflammation; and correcting BM pathology. Using single-cell RNA sequencing on lineage–sca1+c-Kit+ (LSK) hematopoietic stem cells (HSCs) from untreated and DMHCA-treated diabetic mice, we provide potentially novel insights into hematopoiesis and reveal DMHCA’s mechanism of action in correcting diabetic HSCs by reducing myeloidosis and increasing CACs and erythrocyte progenitors. Taken together, these findings demonstrate the beneficial effects of DMHCA treatment on diabetes-induced retinal and BM pathology.

Authors

Cristiano P. Vieira, Seth D. Fortmann, Masroor Hossain, Ana Leda Longhini, Sandra S. Hammer, Bright Asare-Bediako, David K. Crossman, Micheli S. Sielski, Yvonne Adu-Agyeiwaah, Mariana Dupont, Jason L. Floyd, Sergio Li Calzi, Todd Lydic, Robert S. Welner, Gary J. Blanchard, Julia V. Busik, Maria B. Grant

×

Endogenous CCN family member WISP-1 inhibits trauma-induced heterotopic ossification
Ginny Ching-Yun Hsu, … , Benjamin Levi, Aaron W. James
Ginny Ching-Yun Hsu, … , Benjamin Levi, Aaron W. James
Published June 2, 2020
Citation Information: JCI Insight. 2020. https://doi.org/10.1172/jci.insight.135432.
View: Text | PDF

Endogenous CCN family member WISP-1 inhibits trauma-induced heterotopic ossification

  • Text
  • PDF
Abstract

Heterotopic ossification (HO) is defined as abnormal differentiation of local stromal cells of mesenchymal origin resulting in pathologic cartilage and bone matrix deposition. CCN family members are matricellular proteins that have diverse regulatory functions on cell proliferation and differentiation, including the regulation of chondrogenesis. However, little is known regarding CCN family member expression or function in HO. Here, a combination of bulk and single cell RNA sequencing defined the dynamic temporospatial pattern of CCN family member induction within a mouse model of trauma-induced HO. Among CCN family proteins, Wisp1(also known as Ccn4) was most upregulated during the evolution of HO, and Wisp1 expression corresponded with chondrogenic gene profile. Immunohistochemistry confirmed WISP1/CCN4 expression across traumatic and genetic HO mouse models, as well as in human HO samples. Transgenic Wisp1LacZ/LacZ knockin animals showed an increase in endochondral ossification in HO after trauma. Finally, the transcriptome of Wisp1 null tenocytes revealed enrichment in signaling pathways such as STAT3 and PCP signaling that may explain increased HO in the context of Wisp1 deficiency. In sum, CCN family members, and in particular Wisp1, are spatiotemporally associated with and negatively regulate trauma-induced HO formation.

Authors

Ginny Ching-Yun Hsu, Simone Marini, Stefano Negri, Yiyun Wang, Jiajia Xu, Chase A. Pagani, Charles Hwang, David M. Stepien, Carolyn A. Meyers, Sarah Miller, Edward McCarthy, Karen M. Lyons, Benjamin Levi, Aaron W. James

×

Scleraxis is required for the growth of adult tendons in response to mechanical loading
Jonathan P. Gumucio, … , Eithne Comerford, Christopher L. Mendias
Jonathan P. Gumucio, … , Eithne Comerford, Christopher L. Mendias
Published May 28, 2020
Citation Information: JCI Insight. 2020. https://doi.org/10.1172/jci.insight.138295.
View: Text | PDF

Scleraxis is required for the growth of adult tendons in response to mechanical loading

  • Text
  • PDF
Abstract

Scleraxis is a basic helix-loop-helix transcription factor that plays a central role in promoting tenocyte proliferation and matrix synthesis during embryonic tendon development. However, the role of scleraxis in the growth and adaptation of adult tendons is not known. We hypothesized that scleraxis is required for tendon growth in response to mechanical loading, and that scleraxis promotes the specification of progenitor cells into tenocytes. We conditionally deleted scleraxis in adult mice using a tamoxifen-inducible Cre-recombinase expressed from the Rosa26 locus (ScxΔ), and then induced tendon growth in Scx+ and ScxΔ adult mice via plantaris tendon mechanical overload. Compared to the wild type Scx+ group, ScxΔ mice demonstrated blunted tendon growth. Transcriptional and proteomic analyses revealed significant reductions in cell proliferation, protein synthesis, and extracellular matrix genes and proteins. Our results indicate that scleraxis is required for mechanically-stimulated adult tendon growth by causing the commitment of CD146+ pericytes into the tenogenic lineage, and by promoting the initial expansion of newly committed tenocytes and the production of extracellular matrix proteins.

Authors

Jonathan P. Gumucio, Martin M. Schonk, Yalda A. Kharaz, Eithne Comerford, Christopher L. Mendias

×

Duchenne muscular dystrophy hiPSC-derived myoblast drug screen identifies compounds that ameliorate disease in mdx mice
Congshan Sun, … , Kathryn R. Wagner, Gabsang Lee
Congshan Sun, … , Kathryn R. Wagner, Gabsang Lee
Published April 28, 2020
Citation Information: JCI Insight. 2020. https://doi.org/10.1172/jci.insight.134287.
View: Text | PDF

Duchenne muscular dystrophy hiPSC-derived myoblast drug screen identifies compounds that ameliorate disease in mdx mice

  • Text
  • PDF
Abstract

Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy. When human induced pluripotent stem cells (hiPSCs) were differentiated into myoblasts, the myoblasts derived from DMD patients’ hiPSCs (DMD hiPSC-derived myoblasts) exhibited an identifiable DMD relevant phenotype: myogenic fusion deficiency. Based on this model, we developed a DMD hiPSC-derived myoblast screening platform employing a high-content imaging (BD pathway 855) approach to generate parameters describing morphological as well as myogenic marker protein expression. Following treatment of the cells with 1524 compounds from the Johns Hopkins Clinical Compound Library, compounds that enhanced myogenic fusion of DMD hiPSC-derived myoblasts were identified. The final hits were ginsenoside Rd and fenofibrate. Transcriptional profiling revealed that ginsenoside Rd is functionally related to FLT3 signaling, while fenofibrate is linked to TGF-β signaling. Preclinical tests in mdx mice showed that treatment with these two hit compounds can significantly ameliorate some of the skeletal muscle phenotypes caused by dystrophin deficiency, supporting their therapeutic potential. Further study with hiPSC-derived cardiomyocytes revealed that fenofibrate could inhibit mitochondria-induced apoptosis in the DMD hiPSC-derived cardiomyocytes. We have developed a platform based on DMD hiPSC-derived myoblasts for drug screening and identified two promising small molecules with in vivo efficacy.

Authors

Congshan Sun, In Young Choi, Yazmin I. Rovira Gonzalez, Peter Andersen, C. Conover Talbot Jr., Shama R. Iyer, Richard M. Lovering, Kathryn R. Wagner, Gabsang Lee

×

Myocyte-derived Myomaker expression is required for regenerative fusion but exacerbates membrane instability in dystrophic myofibers
Michael J. Petrany, … , Sakthivel Sadayappan, Douglas P. Millay
Michael J. Petrany, … , Sakthivel Sadayappan, Douglas P. Millay
Published April 20, 2020
Citation Information: JCI Insight. 2020. https://doi.org/10.1172/jci.insight.136095.
View: Text | PDF

Myocyte-derived Myomaker expression is required for regenerative fusion but exacerbates membrane instability in dystrophic myofibers

  • Text
  • PDF
Abstract

Muscle progenitor cell fusion is required for the formation and regeneration of multinucleated skeletal muscle fibers. Chronic muscle regeneration in Duchenne muscular dystrophy (DMD) is characterized by ongoing fusion of satellite cell (SC) progeny, but the effects of fusion on disease and the mechanisms by which fusion is accomplished in this setting are not fully understood. Using the mdx mouse model of DMD, we deleted the fusogenic protein Myomaker in SCs or myofibers. Following deletion in SCs, mice displayed a complete lack of myocyte fusion, resulting in severe muscle loss, enhanced fibrosis, and significant functional decline. Reduction of Myomaker in mature myofibers in mdx mice, however, led to minimal alterations in fusion dynamics. Unexpectedly, myofiber-specific deletion of myomaker resulted in improvement of disease phenotype, with enhanced function and decreased muscle damage. Our data indicate that Myomaker has divergent effects on dystrophic disease severity depending upon its compartment of expression. These findings show that myocyte fusion is absolutely required for effective regeneration in DMD, but persistent Myomaker expression in myofibers due to ongoing fusion may have unintended deleterious consequences for muscle integrity. Thus, sustained activation of a component of the myogenic program in dystrophic myofibers exacerbates disease.

Authors

Michael J. Petrany, Taejeong Song, Sakthivel Sadayappan, Douglas P. Millay

×

CIC is a critical regulator of neuronal differentiation
Inah Hwang, … , Hongwu Zheng, Jihye Paik
Inah Hwang, … , Hongwu Zheng, Jihye Paik
Published March 31, 2020
Citation Information: JCI Insight. 2020. https://doi.org/10.1172/jci.insight.135826.
View: Text | PDF

CIC is a critical regulator of neuronal differentiation

  • Text
  • PDF
Abstract

Capicua (CIC), a member of the high mobility group (HMG)-box superfamily of transcriptional repressors, is frequently mutated in human oligodendrogliomas. But its function in brain development and tumorigenesis remains poorly understood. Here, we report that brain-specific deletion of Cic compromises developmental transition of neuroblast to immature neurons in mouse hippocampus and compromises normal neuronal differentiation. Combined gene expression and ChIP-seq analyses identified VGF as an important CIC-repressed transcriptional surrogate involved in neuronal lineage regulation. Aberrant VGF expression promotes neural progenitor cell proliferation by suppressing their differentiation. Mechanistically, we demonstrated that CIC represses VGF expression by tethering SIN3-HDAC to form a transcriptional corepressor complex. Mass spectrometry analysis of CIC-interacting proteins further identified BRG1 containing mSWI/SNF complex whose function is necessary for transcriptional repression by CIC. Together, this study uncovers a novel regulatory pathway of CIC-dependent neuronal differentiation and may implicate these molecular mechanisms in CIC-dependent brain tumorigenesis.

Authors

Inah Hwang, Heng Pan, Jun Yao, Olivier Elemento, Hongwu Zheng, Jihye Paik

×
  • ← Previous
  • 1
  • 2
  • 3
  • 4
  • 5
  • 6
  • 7
  • Next →

No posts were found with this tag.

Advertisement

Copyright © 2022 American Society for Clinical Investigation
ISSN 2379-3708

Sign up for email alerts