1
|
Herath M, Speer AL. Bioengineering of Intestinal Grafts. Gastroenterol Clin North Am 2024; 53:461-472. [PMID: 39068007 PMCID: PMC11284275 DOI: 10.1016/j.gtc.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Intestinal failure manifests as an impaired capacity of the intestine to sufficiently absorb vital nutrients and electrolytes essential for growth and well-being in pediatric and adult populations. Although parenteral nutrition remains the mainstay therapeutic approach, the pursuit of a definitive and curative strategy, such as regenerative medicine, is imperative. Substantial advancements in the field of engineered intestinal tissues present a promising avenue for addressing intestinal failure; nevertheless, extensive research is still necessary for effective translation from experimental benchwork to clinical bedside applications.
Collapse
Affiliation(s)
- Madushani Herath
- Program in Children's Regenerative Medicine, Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), 6431 Fannin Street, Suite 5.254, Houston, TX 77030, USA
| | - Allison L Speer
- Program in Children's Regenerative Medicine, Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), 6431 Fannin Street, Suite 5.254, Houston, TX 77030, USA.
| |
Collapse
|
2
|
Sun H, Liu F, Lin Z, Jiang Z, Wen X, Xu J, Zhang Z, Ma R. Silencing of NOTCH3 Signaling in Meniscus Smooth Muscle Cells Inhibits Fibrosis and Exacerbates Degeneration in a HEYL-Dependent Manner. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207020. [PMID: 37026620 PMCID: PMC10238196 DOI: 10.1002/advs.202207020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/08/2023] [Indexed: 06/04/2023]
Abstract
The mechanisms of meniscus fibrosis and novel ways to enhance fibrosis is unclear. This work reveals human meniscus fibrosis initiated at E24 weeks. Smooth muscle cell cluster is identified in embryonic meniscus, and the combined analysis with previous data suggests smooth muscle cell in embryonic meniscus as precursors of progenitor cells in the mature meniscus. NOTCH3 is constantly expressed in smooth muscle cells throughout embryogenesis to adulthood. Inhibition of NOTCH3 signaling in vivo inhibits meniscus fibrosis and exacerbates degeneration. Continuous histological sections show that HEYL, NOTCH3 downstream target gene, is expressed consistently with NOTCH3. HEYL knockdown in meniscus cells attenuated the COL1A1 upregulation by CTGF and TGF-β stimulation. Thus, this study discovers the existence of smooth muscle cells and fibers in the meniscus. Inhibition of NOTCH3 signaling in meniscus smooth muscle cells in a HEYL-dependent manner prevented meniscus fibrosis and exacerbated degeneration. Therefore, NOTCH3/HEYL signaling might be a potential therapeutic target for meniscus fibrosis.
Collapse
Affiliation(s)
- Hao Sun
- Department of Orthopaedic surgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120China
| | - Fangzhou Liu
- Department of Orthopaedic surgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120China
| | - Zhencan Lin
- Department of Orthopaedic surgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120China
| | - Zongrui Jiang
- Department of Joint SurgeryFirst Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Xingzhao Wen
- Department of Joint SurgeryFirst Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Jie Xu
- Department of Orthopaedic surgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120China
| | - Zhiqi Zhang
- Department of Joint SurgeryFirst Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Ruofan Ma
- Department of Orthopaedic surgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120China
| |
Collapse
|
3
|
Wen SM, Wen WC, Chao PHG. Zyxin and actin structure confer anisotropic YAP mechanotransduction. Acta Biomater 2022; 152:313-320. [PMID: 36089236 DOI: 10.1016/j.actbio.2022.08.079] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 08/24/2022] [Accepted: 08/31/2022] [Indexed: 11/01/2022]
Abstract
Tissues and the embedded cells experience anisotropic deformations due to their functions and anatomical locations. The resident cells, such as tenocytes and muscle cells, are often restricted by their extracellular matrix and organize parallel to their major loading direction, yet most studies on cellular responses to strains use isotropic substrates without predetermined organizations. To understand how confined cells sense and respond to anisotropic loading, we combine cell patterning and uniaxial stretch to have precise geometric control. Dynamic stretch parallel to the long axis of the cell activates YAP nuclear translocation, but not when stretched in the perpendicular direction. Looking at the initial cytoskeleton response, parallel stretch leads to actin breakage and repair within the first minute, mediated by zyxin, the focal adhesion protein. In addition, this zyxin-mediated repair response is controlled by focal adhesion kinase (FAK) and leads to YAP signaling. As these factors are intimately involved in a wide range of mechanical regulation, our findings point to new roles of zyxin and YAP in anisotropic mechanotransduction, and may provide additional perspectives in cellular adaptive responses and tissue homeostasis. STATEMENT OF SIGNIFICANCE: Structure and deformation of tissues control gene expression, migration, and proliferation of the resident cells. In an effort to understand the underlying mechanisms, we find that the transcription cofactor YAP respond to mechanical stretch in a direction-dependent manner. We demonstrate that parallel stretch induces actin cytoskeleton damage, focal adhesion kinase (FAK) activation, and zyxin relocation, which are involved in the anisotropic YAP signaling. Our findings provide additional perspectives in the interactions of tissue structure and cell mechanotransduction.
Collapse
Affiliation(s)
- Shin-Min Wen
- Department of Biomedical Engineering, School of Medicine and School of Engineering National Taiwan University
| | - Wen-Cih Wen
- Department of Biomedical Engineering, School of Medicine and School of Engineering National Taiwan University
| | - Pen-Hsiu Grace Chao
- Department of Biomedical Engineering, School of Medicine and School of Engineering National Taiwan University.
| |
Collapse
|
4
|
Role of smooth muscle progenitor cells in vascular mechanical injury and repair. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
5
|
Tullie L, Jones BC, De Coppi P, Li VSW. Building gut from scratch - progress and update of intestinal tissue engineering. Nat Rev Gastroenterol Hepatol 2022; 19:417-431. [PMID: 35241800 DOI: 10.1038/s41575-022-00586-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/31/2022] [Indexed: 12/18/2022]
Abstract
Short bowel syndrome (SBS), a condition defined by insufficient absorptive intestinal epithelium, is a rare disease, with an estimated prevalence up to 0.4 in 10,000 people. However, it has substantial morbidity and mortality for affected patients. The mainstay of treatment in SBS is supportive, in the form of intravenous parenteral nutrition, with the aim of achieving intestinal autonomy. The lack of a definitive curative therapy has led to attempts to harness innate developmental and regenerative mechanisms to engineer neo-intestine as an alternative approach to addressing this unmet clinical need. Exciting advances have been made in the field of intestinal tissue engineering (ITE) over the past decade, making a review in this field timely. In this Review, we discuss the latest advances in the components required to engineer intestinal grafts and summarize the progress of ITE. We also explore some key factors to consider and challenges to overcome when transitioning tissue-engineered intestine towards clinical translation, and provide the future outlook of ITE in therapeutic applications and beyond.
Collapse
Affiliation(s)
- Lucinda Tullie
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK.,Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Brendan C Jones
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK. .,Specialist Neonatal and Paediatric Surgery Unit, Great Ormond Street Hospital, London, UK.
| | - Vivian S W Li
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
6
|
Shukla PS, Xia R, Lin LH, Schwartz CJ. Gynaecological perivascular epithelioid cell tumour (PEComa): comparative analysis of proposed algorithms for prediction of clinical outcome. Histopathology 2021; 79:847-860. [PMID: 34157139 DOI: 10.1111/his.14434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/09/2021] [Accepted: 06/18/2021] [Indexed: 01/31/2023]
Abstract
AIMS Perivascular epithelioid cell tumours (PEComas) are rare mesenchymal tumours that coexpress smooth muscle and melanocytic markers. They have a predilection for gynaecological organs, where they present a unique diagnostic challenge, because of morphological and immunohistochemical overlap with more common smooth muscle and stromal tumours. Limited information regarding the natural history, owing to the rarity of this tumour, makes accurate risk stratification difficult. We aimed to review clinicopathological features of gynaecological PEComa and compare accuracy of five different classification systems for prediction of prognosis. METHODS AND RESULTS We have described the clinicopathological features of 13 new cases and tested five prognostic algorithms in a total of 67 cases of gynaecological PEComa. Receiver operating characteristic curves were constructed and areas under the curve (AUCs) were calculated to evaluate predictive accuracy. The modified gynaecological-specific algorithm showed high sensitivity and specificity and yielded the highest AUC (0.864). It's earlier version, the gynaecological-specific algorithm, suffered from lower specificity (AUC = 0.843). The post-hoc McNemar test confirmed significant differences between the performances of the modified gynaecological-specific algorithm and the gynaecological-specific algorithm (P = 0.008). The original Folpe algorithm for PEComas of all sites showed low specificity, had a lower AUC (0.591), and was inapplicable in 18% of cases. Its two later versions (the revised Folpe algorithm and the modified Folpe algorithm) also yielded lower AUCs (0.690 and 0.591, respectively). CONCLUSION We have shown that the modified gynaecological-specific algorithm predicts the clinical outcome of gynaecological PEComa with high accuracy, and have validated its use for prognostic stratification of gynaecological PEComa.
Collapse
Affiliation(s)
| | - Rong Xia
- NYU Langone Medical Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lawrence Hsu Lin
- NYU Langone Medical Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
7
|
Ruan J, Zhang L, Hu D, Qu X, Yang F, Chen F, He X, Shen J, Dong K, Sweet M, Sanchez C, Li D, Shou W, Zhou J, Cai CL. Novel Myh11 Dual Reporter Mouse Model Provides Definitive Labeling and Identification of Smooth Muscle Cells-Brief Report. Arterioscler Thromb Vasc Biol 2021; 41:815-821. [PMID: 33356387 DOI: 10.1161/atvbaha.120.315107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Myh11 encodes a myosin heavy chain protein that is specifically expressed in smooth muscle cells (SMCs) and is important for maintaining vascular wall stability. The goal of this study is to generate a Myh11 dual reporter mouse line for definitive visualization of MYH11+ SMCs in vivo. Approach and Results: We generated a Myh11 knock-in mouse model by inserting LoxP-nlacZ-4XpolyA-LoxP-H2B-GFP-polyA-FRT-Neo-FRT reporter cassette into the Myh11 gene locus. The nuclear (n) lacZ-4XpolyA cassette is flanked by 2 LoxP sites followed by H2B-GFP (histone 2B fused green fluorescent protein). Upon Cre-mediated recombination, nlacZ-stop cassette is removed thereby permitting nucleus localized H2B-GFP expression. Expression of the nuclear localized lacZ or H2B-GFP is under control of the endogenous Myh11 promoter. Nuclear lacZ was expressed specifically in SMCs at embryonic and adult stages. Following germline Cre-mediated deletion of nuclear lacZ, H2B-GFP was specifically expressed in the nuclei of SMCs. Comparison of nuclear lacZ expression with Wnt1Cre and Mef2cCre mediated-H2B-GFP expression revealed heterogenous origins of SMCs from neural crest and second heart field in the great arteries and coronary vessels adjacent to aortic root. CONCLUSIONS The Myh11 knock-in dual reporter mouse model offers an exceptional genetic tool to visualize and trace the origins of SMCs in mice.
Collapse
MESH Headings
- Age Factors
- Animals
- Cell Lineage
- Cell Tracking
- Female
- Gene Expression Regulation, Developmental
- Gene Knock-In Techniques
- Genes, Reporter
- Gestational Age
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Lac Operon
- Male
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Transgenic
- Muscle, Smooth, Vascular/embryology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myosin Heavy Chains/genetics
- Myosin Heavy Chains/metabolism
- Mice
Collapse
Affiliation(s)
- Jian Ruan
- School of Life Sciences, Shanghai University, China (J.R., F.C.)
- Department of Pediatrics, Herman Wells Center for Pediatric Research, Indiana University School of Medicine (J.R., L.Z., D.H., F.Y., M.S., C.S., W.S., C.-L.C.)
| | - Lu Zhang
- Department of Pediatrics, Herman Wells Center for Pediatric Research, Indiana University School of Medicine (J.R., L.Z., D.H., F.Y., M.S., C.S., W.S., C.-L.C.)
| | - Donghua Hu
- Department of Pediatrics, Herman Wells Center for Pediatric Research, Indiana University School of Medicine (J.R., L.Z., D.H., F.Y., M.S., C.S., W.S., C.-L.C.)
| | - Xianghu Qu
- Division of Pediatrics Cardiology, Vanderbilt University, Nashville, TN (X.Q.)
| | - Fan Yang
- Department of Pediatrics, Herman Wells Center for Pediatric Research, Indiana University School of Medicine (J.R., L.Z., D.H., F.Y., M.S., C.S., W.S., C.-L.C.)
| | - Fuxue Chen
- School of Life Sciences, Shanghai University, China (J.R., F.C.)
| | - Xiangqin He
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University (X.H., J.S., K.D., J.Z.)
| | - Jian Shen
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University (X.H., J.S., K.D., J.Z.)
| | - Kunzhe Dong
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University (X.H., J.S., K.D., J.Z.)
| | - Megan Sweet
- Department of Pediatrics, Herman Wells Center for Pediatric Research, Indiana University School of Medicine (J.R., L.Z., D.H., F.Y., M.S., C.S., W.S., C.-L.C.)
| | - Christina Sanchez
- Department of Pediatrics, Herman Wells Center for Pediatric Research, Indiana University School of Medicine (J.R., L.Z., D.H., F.Y., M.S., C.S., W.S., C.-L.C.)
| | - Deqiang Li
- Division of Cardiovascular Surgery, University of Maryland School of Medicine, Baltimore (D.L.)
| | - Weinian Shou
- Department of Pediatrics, Herman Wells Center for Pediatric Research, Indiana University School of Medicine (J.R., L.Z., D.H., F.Y., M.S., C.S., W.S., C.-L.C.)
| | - Jiliang Zhou
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University (X.H., J.S., K.D., J.Z.)
| | - Chen-Leng Cai
- Department of Pediatrics, Herman Wells Center for Pediatric Research, Indiana University School of Medicine (J.R., L.Z., D.H., F.Y., M.S., C.S., W.S., C.-L.C.)
| |
Collapse
|
8
|
Duddu S, Chakrabarti R, Ghosh A, Shukla PC. Hematopoietic Stem Cell Transcription Factors in Cardiovascular Pathology. Front Genet 2020; 11:588602. [PMID: 33193725 PMCID: PMC7596349 DOI: 10.3389/fgene.2020.588602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
Transcription factors as multifaceted modulators of gene expression that play a central role in cell proliferation, differentiation, lineage commitment, and disease progression. They interact among themselves and create complex spatiotemporal gene regulatory networks that modulate hematopoiesis, cardiogenesis, and conditional differentiation of hematopoietic stem cells into cells of cardiovascular lineage. Additionally, bone marrow-derived stem cells potentially contribute to the cardiovascular cell population and have shown potential as a therapeutic approach to treat cardiovascular diseases. However, the underlying regulatory mechanisms are currently debatable. This review focuses on some key transcription factors and associated epigenetic modifications that modulate the maintenance and differentiation of hematopoietic stem cells and cardiac progenitor cells. In addition to this, we aim to summarize different potential clinical therapeutic approaches in cardiac regeneration therapy and recent discoveries in stem cell-based transplantation.
Collapse
Affiliation(s)
| | | | | | - Praphulla Chandra Shukla
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
9
|
Markan KR, Boland LK, King-McAlpin AQ, Claflin KE, Leaman MP, Kemerling MK, Stonewall MM, Amendt BA, Ankrum JA, Potthoff MJ. Adipose TBX1 regulates β-adrenergic sensitivity in subcutaneous adipose tissue and thermogenic capacity in vivo. Mol Metab 2020; 36:100965. [PMID: 32240964 PMCID: PMC7115112 DOI: 10.1016/j.molmet.2020.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/07/2020] [Accepted: 02/11/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE T-box 1 (TBX1) has been identified as a genetic marker of beige adipose tissue. TBX1 is a mesodermal development transcription factor essential for tissue patterning and cell fate determination. However, whether it plays a role in the process of adipose beiging or how it functions in adipose tissue has not been reported. Here, we examined the function of TBX1 in adipose tissue as well as adipose-derived stem cells from mice and humans. METHODS Adipose-specific TBX1 transgenic (TBX1 AdipoTG) and adipose-specific TBX1 knockout (TBX1 AdipoKO) mice were generated to explore the function of TBX1 in the process of adipose beiging, metabolism and energy homeostasis in vivo. In vitro, we utilized a siRNA mediated approach to determine the function of TBX1 during adipogenesis in mouse and human stem cells. RESULTS Adipose-specific overexpression of TBX1 was not sufficient to fully induce beiging and prevent diet-induced obesity. However, adipose TBX1 expression was necessary to defend body temperature during cold through regulation of UCP1 and for maintaining β3-adrenergic sensitivity and glucose homeostasis in vivo. Loss of adipose TBX1 expression enhanced basal lipolysis and reduced the size of subcutaneous iWAT adipocytes. Reduction of TBX1 expression via siRNA significantly impaired adipogenesis of mouse stromal vascular cells but significantly enhanced adipogenesis in human adipose derived stem cells. CONCLUSIONS Adipose expression of TBX1 is necessary, but not sufficient, to defend body temperature during cold via proper UCP1 expression. Adipose TBX1 expression was also required for proper insulin signaling in subcutaneous adipose as well as for maintaining β-adrenergic sensitivity, but overexpression of TBX1 was not sufficient to induce adipocyte beiging or to prevent diet-induced obesity. TBX1 expression is enriched in adipose stem cells in which it has contrasting effects on adipogenesis in mouse versus human cells. Collectively, these data demonstrate the importance of adipose TBX1 in the regulation of beige adipocyte function, energy homeostasis, and adipocyte development.
Collapse
Affiliation(s)
- Kathleen R Markan
- Department of Neuroscience and Pharmacology, Iowa City, IA, 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA.
| | - Lauren K Boland
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA; Roy J. Carver Department of Biomedical Engineering, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Abdul Qaadir King-McAlpin
- Department of Neuroscience and Pharmacology, Iowa City, IA, 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA
| | - Kristin E Claflin
- Department of Neuroscience and Pharmacology, Iowa City, IA, 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA; Iowa Neurosciences Institute, Iowa City, IA, 52242, USA
| | - Michael P Leaman
- Department of Neuroscience and Pharmacology, Iowa City, IA, 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA
| | - Morgan K Kemerling
- Department of Neuroscience and Pharmacology, Iowa City, IA, 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA
| | - Madison M Stonewall
- Department of Neuroscience and Pharmacology, Iowa City, IA, 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA
| | - Brad A Amendt
- Department of Anatomy and Cell Biology and the Craniofacial Anomalies Research Center, Iowa City, IA, 52242, USA
| | - James A Ankrum
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA; Roy J. Carver Department of Biomedical Engineering, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Matthew J Potthoff
- Department of Neuroscience and Pharmacology, Iowa City, IA, 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA; Iowa Neurosciences Institute, Iowa City, IA, 52242, USA; Department of Veterans Affairs Medical Center, Iowa City, IA, 52242, USA
| |
Collapse
|
10
|
Bartoli-Leonard F, Wilkinson FL, Langford-Smith AWW, Alexander MY, Weston R. The Interplay of SIRT1 and Wnt Signaling in Vascular Calcification. Front Cardiovasc Med 2018; 5:183. [PMID: 30619890 PMCID: PMC6305318 DOI: 10.3389/fcvm.2018.00183] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/04/2018] [Indexed: 12/24/2022] Open
Abstract
Vascular calcification is a major health risk and is highly correlated with atherosclerosis, diabetes, and chronic kidney disease. The development of vascular calcification is an active and complex process linked with a multitude of signaling pathways, which regulate promoters and inhibitors of osteogenesis, the balance of which become deregulated in disease conditions. SIRT1, a protein deacetylase, known to be protective in inhibiting oxidative stress and inflammation within the vessel wall, has been shown as a possible key player in modulating the cell-fate determining canonical Wnt signaling pathways. Suppression of SIRT1 has been reported in patients suffering with cardiovascular pathologies, suggesting that the sustained acetylation of osteogenic factors could contribute to their activation and in turn, lead to the progression of calcification. There is clear evidence of the synergy between β-Catenin and elevated Runx2, and with Wnt signaling being β-Catenin dependent, further understanding is needed as to how these molecular pathways converge and interact, in order to provide novel insight into the mechanism by which smooth muscle cells switch to an osteogenic differentiation programme. Therefore, this review will describe the current concepts of pathological soft tissue mineralization, with a focus on the contribution of SIRT1 as a regulator of Wnt signaling and its targets, discussing SIRT1 as a potential target for manipulation and therapy.
Collapse
Affiliation(s)
- Francesca Bartoli-Leonard
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - Fiona L Wilkinson
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - Alex W W Langford-Smith
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - M Y Alexander
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - Ria Weston
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| |
Collapse
|
11
|
Hamanaka S, Umino A, Sato H, Hayama T, Yanagida A, Mizuno N, Kobayashi T, Kasai M, Suchy FP, Yamazaki S, Masaki H, Yamaguchi T, Nakauchi H. Generation of Vascular Endothelial Cells and Hematopoietic Cells by Blastocyst Complementation. Stem Cell Reports 2018; 11:988-997. [PMID: 30245211 PMCID: PMC6178562 DOI: 10.1016/j.stemcr.2018.08.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 08/22/2018] [Accepted: 08/22/2018] [Indexed: 01/06/2023] Open
Abstract
In the case of organ transplantation accompanied by vascular anastomosis, major histocompatibility complex mismatched vascular endothelial cells become a target for graft rejection. Production of a rejection-free, transplantable organ, therefore, requires simultaneous generation of vascular endothelial cells within the organ. To generate pluripotent stem cell (PSC)-derived vascular endothelial cells, we performed blastocyst complementation with a vascular endothelial growth factor receptor-2 homozygous mutant blastocyst. This mutation is embryonic lethal at embryonic (E) day 8.5–9.5 due to an early defect in endothelial and hematopoietic cells. The Flk-1 homozygous knockout chimeric mice survived to adulthood for over 1 year without any abnormality, and all vascular endothelial cells and hematopoietic cells were derived from the injected PSCs. This approach could be used in conjunction with other gene knockouts which induce organ deficiency to produce a rejection-free, transplantable organ in which all the organ's cells and vasculature are PSC derived. Flk-1-deficient PSCs did not contribute to vascular endothelial cells in chimeric mice Flk-1-deficient mice survived into adulthood by blastocyst complementation Both vascular endothelial cells and hematopoietic cells were generated from PSCs
Collapse
Affiliation(s)
- Sanae Hamanaka
- Division of Stem Cell Therapy, Distinguished Professor Unit, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Ayumi Umino
- Division of Stem Cell Therapy, Distinguished Professor Unit, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Hideyuki Sato
- Division of Stem Cell Therapy, Distinguished Professor Unit, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Tomonari Hayama
- Division of Stem Cell Therapy, Distinguished Professor Unit, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Ayaka Yanagida
- Division of Stem Cell Therapy, Distinguished Professor Unit, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Naoaki Mizuno
- Division of Stem Cell Therapy, Distinguished Professor Unit, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Toshihiro Kobayashi
- Division of Stem Cell Therapy, Distinguished Professor Unit, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Mariko Kasai
- Division of Stem Cell Therapy, Distinguished Professor Unit, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Fabian Patrik Suchy
- Institute for Stem Cell Biology and Regenerative Medicine, Department of Genetics, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Satoshi Yamazaki
- Division of Stem Cell Therapy, Distinguished Professor Unit, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Hideki Masaki
- Division of Stem Cell Therapy, Distinguished Professor Unit, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Tomoyuki Yamaguchi
- Division of Stem Cell Therapy, Distinguished Professor Unit, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Distinguished Professor Unit, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; Institute for Stem Cell Biology and Regenerative Medicine, Department of Genetics, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA.
| |
Collapse
|
12
|
Kahn CI, MacNeil M, Fanola CL, Whitney ER. Complex arterial patterning in an anatomical donor. TRANSLATIONAL RESEARCH IN ANATOMY 2018. [DOI: 10.1016/j.tria.2018.06.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
13
|
Deng Y, Lin C, Zhou HJ, Min W. Smooth muscle cell differentiation: Mechanisms and models for vascular diseases. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/s11515-017-1473-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
14
|
Wang J, Li W, Zheng X, Pang X, Du G. Research progress on the forkhead box C1. Oncotarget 2017; 9:12471-12478. [PMID: 29552326 PMCID: PMC5844762 DOI: 10.18632/oncotarget.22527] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 11/01/2017] [Indexed: 01/19/2023] Open
Abstract
FOXC1 is a vital member of FOX families which play important roles in biological processes including proliferation, differentiation, apoptosis, migration, invasion, metabolism, and longevity. Here we are focusing on roles of FOXC1 and their mechanisms in cancers. FOXC1 promoted progress of many cancers, such as breast cancer (especially basal-like breast cancer), hepatocellular carcinoma, gastric cancer and so on. FOXC1 was also found to be associated with drug resistance of cancers. FOXC1 promoted metastasis of cancers by increasing expression of MMP7, NEDD9 and Snail. Proliferation and invasion of cancers were increased by FOXC1 by mediating NF-κB, MST1R and KLF4 expression. FOXC1 was associated with development by regulating expression of FGF19 and MSX1. Recently, FOXC1 was found to be required for niche of stem cells or development of stem cells by mediating expression of Gli2, CXCL12, SCF, NFATC1, BMP and Myh7. Overall, FOXC1 exerts its functions by many mechanisms and may be used as a potential biomarker for diseases.
Collapse
Affiliation(s)
- Jinhua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China.,Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Wan Li
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Xiangjin Zheng
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Xiaocong Pang
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China.,Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
15
|
Locally Transplanted CD34+ Bone Marrow–Derived Cells Contribute to Vascular Healing After Vascular Injury. Transplant Proc 2017; 49:1467-1476. [DOI: 10.1016/j.transproceed.2017.01.081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/24/2017] [Indexed: 01/13/2023]
|
16
|
Collado MS, Cole BK, Figler RA, Lawson M, Manka D, Simmers MB, Hoang S, Serrano F, Blackman BR, Sinha S, Wamhoff BR. Exposure of Induced Pluripotent Stem Cell-Derived Vascular Endothelial and Smooth Muscle Cells in Coculture to Hemodynamics Induces Primary Vascular Cell-Like Phenotypes. Stem Cells Transl Med 2017. [PMID: 28628273 PMCID: PMC5689791 DOI: 10.1002/sctm.17-0004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Human induced pluripotent stem cells (iPSCs) can be differentiated into vascular endothelial (iEC) and smooth muscle (iSMC) cells. However, because iECs and iSMCs are not derived from an intact blood vessel, they represent an immature phenotype. Hemodynamics and heterotypic cell:cell communication play important roles in vascular cell phenotypic modulation. Here we tested the hypothesis that hemodynamic exposure of iECs in coculture with iSMCs induces an in vivo‐like phenotype. iECs and iSMCs were cocultured under vascular region‐specific blood flow hemodynamics, and compared to hemodynamic cocultures of blood vessel‐derived endothelial (pEC) and smooth muscle (pSMC) cells. Hemodynamic flow‐induced gene expression positively correlated between pECs and iECs as well as pSMCs and iSMCs. While endothelial nitric oxide synthase 3 protein was lower in iECs than pECs, iECs were functionally mature as seen by acetylated‐low‐density lipoprotein (LDL) uptake. SMC contractile protein markers were also positively correlated between pSMCs and iSMCs. Exposure of iECs and pECs to atheroprone hemodynamics with oxidized‐LDL induced an inflammatory response in both. Dysfunction of the transforming growth factor β (TGFβ) pathway is seen in several vascular diseases, and iECs and iSMCs exhibited a transcriptomic prolife similar to pECs and pSMCs, respectively, in their responses to LY2109761‐mediated transforming growth factor β receptor I/II (TGFβRI/II) inhibition. Although there are differences between ECs and SMCs derived from iPSCs versus blood vessels, hemodynamic coculture restores a high degree of similarity in their responses to pathological stimuli associated with vascular diseases. Thus, iPSC‐derived vascular cells exposed to hemodynamics may provide a viable system for modeling rare vascular diseases and testing new therapeutic approaches. Stem Cells Translational Medicine2017;6:1673–1683
Collapse
Affiliation(s)
| | | | | | - Mark Lawson
- HemoShear Therapeutics, LLC, Charlottesville, Virginia, USA
| | - David Manka
- HemoShear Therapeutics, LLC, Charlottesville, Virginia, USA
| | | | - Steve Hoang
- HemoShear Therapeutics, LLC, Charlottesville, Virginia, USA
| | - Felipe Serrano
- Department of Medicine and WT-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | | | - Sanjay Sinha
- Department of Medicine and WT-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
17
|
Shi X, Zhang W, Yin L, Chilian WM, Krieger J, Zhang P. Vascular precursor cells in tissue injury repair. Transl Res 2017; 184:77-100. [PMID: 28284670 PMCID: PMC5429880 DOI: 10.1016/j.trsl.2017.02.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 12/25/2016] [Accepted: 02/14/2017] [Indexed: 12/22/2022]
Abstract
Vascular precursor cells include stem cells and progenitor cells giving rise to all mature cell types in the wall of blood vessels. When tissue injury occurs, local hypoxia and inflammation result in the generation of vasculogenic mediators which orchestrate migration of vascular precursor cells from their niche environment to the site of tissue injury. The intricate crosstalk among signaling pathways coordinates vascular precursor cell proliferation and differentiation during neovascularization. Establishment of normal blood perfusion plays an essential role in the effective repair of the injured tissue. In recent years, studies on molecular mechanisms underlying the regulation of vascular precursor cell function have achieved substantial progress, which promotes exploration of vascular precursor cell-based approaches to treat chronic wounds and ischemic diseases in vital organ systems. Verification of safety and establishment of specific guidelines for the clinical application of vascular precursor cell-based therapy remain major challenges in the field.
Collapse
Affiliation(s)
- Xin Shi
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Weihong Zhang
- Department of Basic Medicine, School of Nursing, Zhengzhou University, Zhengzhou, Henan Province, PR China
| | - Liya Yin
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - William M Chilian
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Jessica Krieger
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Ping Zhang
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio.
| |
Collapse
|
18
|
Guo Q, Xu H, Yang X, Zhao D, Liu S, Sun X, Huang JA. Notch activation of Ca 2+-sensing receptor mediates hypoxia-induced pulmonary hypertension. Hypertens Res 2016; 40:117-129. [PMID: 27581537 DOI: 10.1038/hr.2016.118] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/15/2016] [Accepted: 07/25/2016] [Indexed: 12/13/2022]
Abstract
A recent study from our group demonstrated that the Ca2+-sensing receptor (CaSR) was upregulated and that the extracellular Ca2+-induced increase in the cytosolic Ca2+ concentration [Ca2+]cyt was enhanced in pulmonary arterial smooth muscle cells (PASMCs) from patients with idiopathic pulmonary arterial hypertension. Here, we examined whether hypoxia-induced activation of Notch signaling leads to the activation and upregulation of CaSR in hypoxia-induced pulmonary hypertension (HPH). The activation of Notch signaling with Jag-1, a Notch ligand, can activate the function and increase the expression of CaSR in acute and chronic hypoxic PASMCs. Downregulation of Notch3 with a siRNA attenuates the extracellular Ca2+-induced increase in [Ca2+]cyt and the increase in hypoxia-induced PASMC proliferation in acute hypoxic rat PASMCs. Furthermore, we tested the prevention and rescue effects of a γ-secretase inhibitor (DAPT) in HPH rats. For the Jag-1-treated group, right ventricular systolic pressure (RVSP), right heart hypertrophy (RV/LV+S ratio), and the level of right ventricular myocardial fibrosis were higher than the hypoxia alone group. Meanwhile, DAPT treatment prevented and rescued pulmonary hypertension in HPH rats. The Notch activation of CaSR mediates hypoxia-induced pulmonary hypertension. Understanding the new molecular mechanisms that regulate [Ca2+]cyt and PASMC proliferation is critical to elucidating the pathogenesis of HPH and the development of novel therapies for pulmonary hypertension.
Collapse
Affiliation(s)
- Qiang Guo
- Department of Medicine, Respiratory, Emergency and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hua Xu
- Department of Medicine, Respiratory, Emergency and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinjing Yang
- Department of Medicine, Respiratory, Emergency and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Daguo Zhao
- Department of Medicine, Respiratory, Emergency and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shenlang Liu
- Department of Medicine, Respiratory, Emergency and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xue Sun
- Department of Medicine, Respiratory, Emergency and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian-An Huang
- Department of Medicine, Respiratory, Emergency and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
19
|
Caplan AI. MSCs: The Sentinel and Safe-Guards of Injury. J Cell Physiol 2015; 231:1413-6. [PMID: 26565391 DOI: 10.1002/jcp.25255] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 11/11/2015] [Indexed: 01/08/2023]
Abstract
Mesenchymal stem cells (MSCs) were originally named because they could differentiate in a variety of mesenchymal phenotypes in culture. Evidence indicates that MSCs arise from perivascular cells, pericytes, when the blood vessels are broken or inflamed. These pericyte/MSCs are situated on every blood vessel in the body. The MSCs sense the micro-environment of the injury site and secrete site-specific factors that serve several important reparative functions: first, a curtain of molecules from the front of the MSCs provide a barrier from the interrogation of the over-aggressive immune system. Second, from the back of the MSCs, a different set of bioactive agents inhibit scar formation and establish a regenerative micro-environment. Third, if bacteria are sensed by the MSCs, they produce powerful protein antibiotics that kill the bacteria on contact. Last, the MSCs surround and encyst intruding solid objects like a piece of wood (a "splinter") or other foreign objects. The MSCs act as a combination paramedic and emergency room (ER) staff to survey the damage, isolate foreign components, stabilize the injured tissues, provide antibiotics and encysting protection before a slower, medicinal sequence can be initiated to regenerate the damaged tissue. The MSCs, thus, act as sentinels to safeguard the individual from intrusion and chronic injury. A societal treatment system has evolved, paramedics and ER procedures, which mirror in a macro-sense what MSCs orchestrate in a micro-sense. Key to this new understanding is that MSCs are not "stem cells," but rather as Medicinal Signaling Cells as the therapeutic agents.
Collapse
Affiliation(s)
- Arnold I Caplan
- Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
20
|
Wang G, Jacquet L, Karamariti E, Xu Q. Origin and differentiation of vascular smooth muscle cells. J Physiol 2015; 593:3013-30. [PMID: 25952975 PMCID: PMC4532522 DOI: 10.1113/jp270033] [Citation(s) in RCA: 212] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/19/2015] [Indexed: 12/18/2022] Open
Abstract
Vascular smooth muscle cells (SMCs), a major structural component of the vessel wall, not only play a key role in maintaining vascular structure but also perform various functions. During embryogenesis, SMC recruitment from their progenitors is an important step in the formation of the embryonic vascular system. SMCs in the arterial wall are mostly quiescent but can display a contractile phenotype in adults. Under pathophysiological conditions, i.e. vascular remodelling after endothelial dysfunction or damage, contractile SMCs found in the media switch to a secretory type, which will facilitate their ability to migrate to the intima and proliferate to contribute to neointimal lesions. However, recent evidence suggests that the mobilization and recruitment of abundant stem/progenitor cells present in the vessel wall are largely responsible for SMC accumulation in the intima during vascular remodelling such as neointimal hyperplasia and arteriosclerosis. Therefore, understanding the regulatory mechanisms that control SMC differentiation from vascular progenitors is essential for exploring therapeutic targets for potential clinical applications. In this article, we review the origin and differentiation of SMCs from stem/progenitor cells during cardiovascular development and in the adult, highlighting the environmental cues and signalling pathways that control phenotypic modulation within the vasculature.
![]()
Collapse
Affiliation(s)
- Gang Wang
- Department of Emergency Medicine, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Laureen Jacquet
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Eirini Karamariti
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Qingbo Xu
- Cardiovascular Division, King's College London BHF Centre, London, UK
| |
Collapse
|
21
|
Deb A. Stem Cells. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
22
|
Conlon N, Soslow RA, Murali R. Perivascular epithelioid tumours (PEComas) of the gynaecological tract. J Clin Pathol 2015; 68:418-26. [PMID: 25750268 DOI: 10.1136/jclinpath-2015-202945] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 12/11/2022]
Abstract
Perivascular epithelioid tumours (PEComas) of the gynaecological tract are rare tumours which were first recognised and diagnosed within the last 20 years. They represent a unique diagnostic challenge with regard to their accurate and reproducible distinction from more common entities such as smooth muscle tumours of the uterine corpus. In this review article, we trace the development of the concept of the PEComa tumour family, highlight what is known about extra-gynaecological tract PEComa at an immunohistochemical, molecular and therapeutic level and then present a summary of all reported cases of gynaecological tract PEComa to date. In the summary, we highlight rare subtypes of gynaecological tract PEComa and compare the performances of extant prognostic classification systems for malignancy in these tumours.
Collapse
Affiliation(s)
- Niamh Conlon
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Robert A Soslow
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Rajmohan Murali
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
23
|
Shi N, Chen SY. Mechanisms simultaneously regulate smooth muscle proliferation and differentiation. J Biomed Res 2013; 28:40-6. [PMID: 24474962 PMCID: PMC3904173 DOI: 10.7555/jbr.28.20130130] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 11/13/2013] [Indexed: 01/01/2023] Open
Abstract
Vascular smooth muscle cell (VSMC) differentiation and proliferation are two important physiological processes during vascular development. The phenotypic alteration from differentiated to proliferative VSMC contributes to the development of several major cardiovascular diseases including atherosclerosis, hypertension, restenosis after angioplasty or bypass, diabetic vascular complications, and transplantation arteriopathy. Since the VSMC phenotype in these pathological conditions resembles that of developing VSMC during embryonic development, understanding of the molecular mechanisms that control VSMC differentiation will provide fundamental insights into the pathological processes of these cardiovascular diseases. Although VSMC differentiation is usually accompanied by an irreversible cell cycle exit, VSMC proliferation and differentiation occur concurrently during embryonic development. The molecular mechanisms simultaneously regulating these two processes, however, remain largely unknown. Our recent study demonstrates that cell division cycle 7, a key regulator of cell cycle, promotes both VSMC differentiation and proliferation through different mechanisms during the initial phase of VSMC differentiation. Conversely, Krüppel-like factor 4 appears to be a repressor for both VSMC differentiation and proliferation. This review attempts to highlight the novel role of cell division cycle 7 in TGF-β-induced VSMC differentiation and proliferation. The role of Krüppel-like factor 4 in suppressing these two processes will also be discussed.
Collapse
Affiliation(s)
- Ning Shi
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA 30602, USA
| | - Shi-You Chen
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
24
|
Albertine KH, Dezawa M. A new age of regenerative medicine: fusion of tissue engineering and stem cell research. Anat Rec (Hoboken) 2013; 297:1-3. [PMID: 24293066 DOI: 10.1002/ar.22811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 09/16/2013] [Indexed: 01/21/2023]
Affiliation(s)
- Kurt H Albertine
- Editor-in-Chief, The Anatomical Record, Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah
| | | |
Collapse
|
25
|
Abstract
It is well known that the altered blood flow is related to vascular diseases, including atherosclerosis, restenosis, and arteriosclerosis, which preferentially located at areas with the disturbed blood flow, suggesting that altered biomechanical stress may exert their effect on the vascular disease. Recent evidence indicated the presence of abundant stem/progenitor cells in the vessel wall, in which laminar shear stress can stimulate these cells to differentiate towards endothelial lineage, while cyclic strain results in smooth muscle differentiation. In line with this, it was evidenced that altered biomechanical stress in stented vessels may lead to 'wrong' direction of vascular stem cell differentiation resulting in restenosis. However, the underlying mechanisms are not well understood. In this article, we will give an overview of the effect of the local flow pattern on stem/progenitor cell differentiation and the possible mechanism on how the blood flow influences stem cell behaviours in the development of vascular diseases.
Collapse
Affiliation(s)
- Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital, Jinan, China
| | | | | | | |
Collapse
|
26
|
Tsai TN, Kirton JP, Campagnolo P, Zhang L, Xiao Q, Zhang Z, Wang W, Hu Y, Xu Q. Contribution of stem cells to neointimal formation of decellularized vessel grafts in a novel mouse model. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:362-73. [PMID: 22613026 DOI: 10.1016/j.ajpath.2012.03.021] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 03/20/2012] [Accepted: 03/27/2012] [Indexed: 11/19/2022]
Abstract
Artificial vessel grafts are often used for the treatment of occluded blood vessels, but neointimal lesions commonly occur. To both elucidate and quantify which cell types contribute to the developing neointima, we established a novel mouse model of restenosis by grafting a decellularized vessel to the carotid artery. Typically, the graft developed neointimal lesions after 2 weeks, resulting in lumen closure within 4 weeks. Immunohistochemical staining revealed the presence of endothelial and smooth muscle cells, monocytes, and stem/progenitor cells at 2 weeks after implantation. Explanted cultures of neointimal tissues displayed heterogeneous outgrowth in stem cell medium. These lesional cells expressed a panel of stem/progenitor markers, including c-kit, stem cell antigen-1 (Sca-1), and CD34. Furthermore, these cells showed clonogenic and multilineage differentiation capacities. Isolated Sca-1(+) cells were able to differentiate into endothelial and smooth muscle cells in response to vascular endothelial growth factor (VEGF) or platelet-derived growth factor (PDGF)-BB stimulation in vitro. In vivo, local application of VEGF to the adventitial side of the decellularized vessel increased re-endothelialization and reduced neointimal formation in samples at 4 weeks after implantation. A population of stem/progenitor cells exists within developing neointima, which displays the ability to differentiate into both endothelial and smooth muscle cells and can contribute to restenosis. Our findings also indicate that drugs or cytokines that direct cell differentiation toward an endothelial lineage may be effective tools in the prevention or delay of restenosis.
Collapse
MESH Headings
- Animals
- Antigens, Ly/metabolism
- Blood Vessel Prosthesis
- Blood Vessel Prosthesis Implantation/methods
- Carotid Stenosis/pathology
- Carotid Stenosis/physiopathology
- Carotid Stenosis/prevention & control
- Carotid Stenosis/surgery
- Cell Differentiation
- Cells, Cultured
- Colony-Forming Units Assay
- Disease Models, Animal
- Endothelium, Vascular/pathology
- Graft Occlusion, Vascular/pathology
- Graft Occlusion, Vascular/prevention & control
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Muscle, Smooth, Vascular/pathology
- Neointima/pathology
- Neointima/prevention & control
- Stem Cells/pathology
- Stem Cells/physiology
- Tissue Scaffolds
- Transplantation Chimera
- Vascular Endothelial Growth Factor A/therapeutic use
Collapse
Affiliation(s)
- Tsung-Neng Tsai
- Cardiovascular Division, King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Li X, Chu JS, Yang L, Li S. Anisotropic effects of mechanical strain on neural crest stem cells. Ann Biomed Eng 2012; 40:598-605. [PMID: 22109803 DOI: 10.1007/s10439-011-0403-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 09/09/2011] [Indexed: 10/15/2022]
Abstract
Neural crest stem cells (NCSCs) are multipotent and play an important role during the development and tissue regeneration. However, the anisotropic effects of mechanical strain on NCSCs are not known. To investigate the anisotropic mechanosensing by NCSCs, NCSCs derived from induced pluripotent stem cells were cultured on micropatterned membranes, and subjected to cyclic uniaxial strain in the direction parallel or perpendicular to the microgrooves. Cell and nuclear shape were both regulated by micropatterning and mechanical strain. Among the unpatterned, parallel-patterned and perpendicular-patterned groups, mechanical strain caused an increase in histone deacetylase activity in the parallel-patterned group, accompanied by the increase of cell proliferation. In addition, mechanical strain increased the expression of contractile marker calponin-1 but not other differentiation markers in the unpatterned and parallel-patterned groups. These results demonstrated that NCSCs responded differently to the anisotropic mechanical environment. Understanding the mechanical regulation of NCSCs will reveal the role of mechanical factors in NCSC differentiation during development, and provide a basis for using NCSCs for tissue engineering.
Collapse
Affiliation(s)
- Xian Li
- 111 Project Laboratory of Biomechanics and Tissue Repair, Bioengineering College, Chongqing University, Chongqing 400044, China.
| | | | | | | |
Collapse
|
28
|
Maul TM, Chew DW, Nieponice A, Vorp DA. Mechanical stimuli differentially control stem cell behavior: morphology, proliferation, and differentiation. Biomech Model Mechanobiol 2011; 10:939-53. [PMID: 21253809 PMCID: PMC3208754 DOI: 10.1007/s10237-010-0285-8] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Accepted: 12/31/2010] [Indexed: 11/29/2022]
Abstract
Mesenchymal stem cell (MSC) therapy has demonstrated applications in vascular regenerative medicine. Although blood vessels exist in a mechanically dynamic environment, there has been no rigorous, systematic analysis of mechanical stimulation on stem cell differentiation. We hypothesize that mechanical stimuli, relevant to the vasculature, can differentiate MSCs toward smooth muscle (SMCs) and endothelial cells (ECs). This was tested using a unique experimental platform to differentially apply various mechanical stimuli in parallel. Three forces, cyclic stretch, cyclic pressure, and laminar shear stress, were applied independently to mimic several vascular physiologic conditions. Experiments were conducted using subconfluent MSCs for 5 days and demonstrated significant effects on morphology and proliferation depending upon the type, magnitude, frequency, and duration of applied stimulation. We have defined thresholds of cyclic stretch that potentiate SMC protein expression, but did not find EC protein expression under any condition tested. However, a second set of experiments performed at confluence and aimed to elicit the temporal gene expression response of a select magnitude of each stimulus revealed that EC gene expression can be increased with cyclic pressure and shear stress in a cell-contact-dependent manner. Further, these MSCs also appear to express genes from multiple lineages simultaneously which may warrant further investigation into post-transcriptional mechanisms for controlling protein expression. To our knowledge, this is the first systematic examination of the effects of mechanical stimulation on MSCs and has implications for the understanding of stem cell biology, as well as potential bioreactor designs for tissue engineering and cell therapy applications.
Collapse
Affiliation(s)
- Timothy M. Maul
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
- The McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive. Suite 300, Pittsburgh, PA 15219, USA
- The Center for Vascular Remodeling and Regeneration, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Douglas W. Chew
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
- The McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive. Suite 300, Pittsburgh, PA 15219, USA
- The Center for Vascular Remodeling and Regeneration, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Alejandro Nieponice
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
- The McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive. Suite 300, Pittsburgh, PA 15219, USA
- The Center for Vascular Remodeling and Regeneration, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - David A. Vorp
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
- The McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive. Suite 300, Pittsburgh, PA 15219, USA
- The Center for Vascular Remodeling and Regeneration, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
29
|
Wang A, Tang Z, Li X, Jiang Y, Tsou DA, Li S. Derivation of smooth muscle cells with neural crest origin from human induced pluripotent stem cells. Cells Tissues Organs 2011; 195:5-14. [PMID: 22005509 DOI: 10.1159/000331412] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The heterogeneity of vascular smooth muscle cells (SMCs) is related to their different developmental origins such as the neural crest and mesoderm. Derivation of SMCs from different origins will provide valuable in vitro models for the investigation of vascular development and diseases. From the perspective of regenerative medicine and tissue engineering, an expandable cell source of SMCs is required for the construction of tissue-engineered blood vessels. In this study, we developed a robust protocol to derive neural crest stem cells (NCSCs) from human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). NCSCs derived from ESCs and iPSCs were expandable with similar cell doubling times. NCSCs were capable of differentiating into neural and mesenchymal lineages. TGF-β1 induced the expression of SMC markers calponin-1, SM22α, and smooth muscle myosin heavy chain and resulted in the assembly of smooth muscle α-actin, calponin-1, and SM22α into stress fibers. This work provides a basis for using iPSCs to study SMC biology and deriving vascular cells for tissue engineering.
Collapse
Affiliation(s)
- Aijun Wang
- Department of Bioengineering, University of California, Berkeley, USA
| | | | | | | | | | | |
Collapse
|
30
|
Li X, Chu J, Wang A, Zhu Y, Chu WK, Yang L, Li S. Uniaxial mechanical strain modulates the differentiation of neural crest stem cells into smooth muscle lineage on micropatterned surfaces. PLoS One 2011; 6:e26029. [PMID: 22016804 PMCID: PMC3189240 DOI: 10.1371/journal.pone.0026029] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 09/15/2011] [Indexed: 11/19/2022] Open
Abstract
Neural crest stem cells (NCSCs) play an important role in the development and represent a valuable cell source for tissue engineering. However, how mechanical factors in vivo regulate NCSC differentiation is not understood. Here NCSCs were derived from induced pluripotent stem cells and used as a model to determine whether vascular mechanical strain modulates the differentiation of NCSCs into smooth muscle (SM) lineage. NCSCs were cultured on micropatterned membranes to mimic the organization of smooth muscle cells (SMCs), and subjected to cyclic uniaxial strain. Mechanical strain enhanced NCSC proliferation and ERK2 phosphorylation. In addition, mechanical strain induced contractile marker calponin-1 within 2 days and slightly induced SM myosin within 5 days. On the other hand, mechanical strain suppressed the differentiation of NCSCs into Schwann cells. The induction of calponin-1 by mechanical strain was inhibited by neural induction medium but further enhanced by TGF-β. For NCSCs pre-treated with TGF-β, mechanical strain induced the gene expression of both calponin-1 and SM myosin. Our results demonstrated that mechanical strain regulates the differentiation of NCSCs in a manner dependent on biochemical factors and the differentiation stage of NCSCs. Understanding the mechanical regulation of NCSC differentiation will shed light on the development and remodeling of vascular tissues, and how transplanted NCSCs respond to mechanical factors.
Collapse
Affiliation(s)
- Xian Li
- Bioengineering College, Chongqing University, Chongqing, China
- Department of Bioengineering, University of California, Berkeley, California, United States of America
| | - Julia Chu
- Department of Bioengineering, University of California, Berkeley, California, United States of America
| | - Aijun Wang
- Department of Bioengineering, University of California, Berkeley, California, United States of America
| | - Yiqian Zhu
- Department of Bioengineering, University of California, Berkeley, California, United States of America
| | - Wai Keung Chu
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Li Yang
- Bioengineering College, Chongqing University, Chongqing, China
| | - Song Li
- Department of Bioengineering, University of California, Berkeley, California, United States of America
| |
Collapse
|
31
|
Hsu S, Chu JS, Chen FF, Wang A, Li S. Effects of Fluid Shear Stress on a Distinct Population of Vascular Smooth Muscle Cells. Cell Mol Bioeng 2011; 4:627-636. [PMID: 22924082 DOI: 10.1007/s12195-011-0205-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Vascular smooth muscle cells (SMCs) are a major cell type involved in vascular remodeling. The various developmental origins of SMCs such as neural crest and mesoderm result in heterogeneity of SMCs, which plays an important role in the development of vascular remodeling and diseases. Upon vascular injury, SMCs are exposed to blood flow and subjected to fluid shear stress. Previous studies have shown that fluid shear stress inhibits SMC proliferation. However, the effect of shear stress on the subpopulation of SMCs from specific developmental origin and vascular bed is not well understood. Here we investigated how shear stress regulates human aortic SMCs positive for neural crest markers. DNA microarray analysis showed that shear stress modulates the expression of genes involved in cell proliferation, matrix synthesis, cell signaling, transcription and cytoskeleton organization. Further studies demonstrated that shear stress induced SMC proliferation and cyclin D1, downregulated cell cycle inhibitor p21, and activated Akt pathway. Inhibition of PI-3 kinase blocked these shear stress-induced changes. These results suggest that SMCs with neural crest characteristics may respond to shear stress in a different manner. This finding has significant implications in the remodeling and diseases of blood vessels.
Collapse
Affiliation(s)
- Steven Hsu
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | | | | | | | | |
Collapse
|
32
|
Abstract
Accumulating evidence indicates that the mobilization and recruitment of circulating or tissue-resident progenitor cells that give rise to endothelial cells (ECs) and smooth muscle cells (SMCs) can participate in atherosclerosis, neointima hyperplasia after arterial injury, and transplant arteriosclerosis. It is believed that endothelial progenitor cells do exist and can repair and rejuvenate the arteries under physiologic conditions; however, they may also contribute to lesion formation by influencing plaque stability in advanced atherosclerotic plaque under specific pathologic conditions. At the same time, smooth muscle progenitors, despite their capacity to expedite lesion formation during restenosis, may serve to promote atherosclerotic plaque stabilization by producing extracellular matrix proteins. This profound evidence provides support to the hypothesis that both endothelial and smooth muscle progenitors may act as a double-edged sword in the pathogenesis of arteriosclerosis. Therefore, the understanding of the regulatory networks that control endothelial and smooth muscle progenitor differentiation is undoubtedly fundamental both for basic research and for improving current therapeutic avenues for atherosclerosis. We update the progress in progenitor cell study related to the development of arteriosclerosis, focusing specifically on the role of progenitor cells in lesion formation and discuss the controversial issues that regard the origins, frequency, and impact of the progenitors in the disease.
Collapse
Affiliation(s)
- Paola Campagnolo
- Cardiovascular Division, King's College London BHF Centre, London, England
| | | | | |
Collapse
|
33
|
Torsney E, Xu Q. Resident vascular progenitor cells. J Mol Cell Cardiol 2010; 50:304-11. [PMID: 20850452 DOI: 10.1016/j.yjmcc.2010.09.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Revised: 09/03/2010] [Accepted: 09/08/2010] [Indexed: 12/22/2022]
Abstract
Homeostasis of the vessel wall is essential for maintaining its function, including blood pressure and patency of the lumen. In physiological conditions, the turnover rate of vascular cells, i.e. endothelial and smooth muscle cells, is low, but markedly increased in diseased situations, e.g. vascular injury after angioplasty. It is believed that mature vascular cells have an ability to proliferate to replace lost cells normally. On the other hand, recent evidence indicates stem/progenitor cells may participate in vascular repair and the formation of neointimal lesions in severely damaged vessels. It was found that all three layers of the vessels, the intima, media and adventitia, contain resident progenitor cells, including endothelial progenitor cells, mesenchymal stromal cells, Sca-1+ and CD34+ cells. Data also demonstrated that these resident progenitor cells could differentiate into a variety of cell types in response to different culture conditions. However, collective data were obtained mostly from in vitro culture assays and phenotypic marker studies. There are many unanswered questions concerning the mechanism of cell differentiation and the functional role of these cells in vascular repair and the pathogenesis of vascular disease. In the present review, we aim to summarize the data showing the presence of the resident progenitor cells, to highlight possible signal pathways orchestrating cell differentiation toward endothelial and smooth muscle cells, and to discuss the data limitations, challenges and controversial issues related to the role of progenitors. This article is part of a special issue entitled, "Cardiovascular Stem Cells Revisited".
Collapse
Affiliation(s)
- Evelyn Torsney
- Division of Cardiac & Vascular Sciences, St George's University of London, London, UK
| | | |
Collapse
|
34
|
Genetic variants promoting smooth muscle cell proliferation can result in diffuse and diverse vascular diseases: evidence for a hyperplastic vasculomyopathy. Genet Med 2010; 12:196-203. [PMID: 20130469 DOI: 10.1097/gim.0b013e3181cdd687] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Genetic predisposition to early onset of occlusive vascular diseases, including coronary artery disease, ischemic stroke, and Moyamoya disease, may represent varying presentations of a common underlying dysregulation of vascular smooth muscle cell proliferation. We discuss mutations in two genes, NF1 and ACTA2, which predispose affected individuals to diffuse and diverse vascular diseases. These patients show evidence of diffuse occlusive disease in multiple arterial beds or even develop seemingly diverse arterial pathologies, ranging from occlusions to arterial aneurysms. We also present the current evidence that both NF1 and ACTA2 mutations promote increased smooth muscle cell proliferation in vitro and in vivo, which leads us to propose that these diffuse and diverse vascular diseases are the outward signs of a more fundamental disease: a hyperplastic vasculomyopathy. We suggest that the concept of a hyperplastic vasculomyopathy offers a new approach not only to identifying mutated genes that lead to vascular diseases but also to counseling and possibly treating patients harboring such mutations. In other words, this framework may offer the opportunity to therapeutically target the inappropriate smooth muscle cell behavior that predisposes to a variety of vascular diseases throughout the arterial system.
Collapse
|
35
|
Orlandi A, Bennett M. Progenitor cell-derived smooth muscle cells in vascular disease. Biochem Pharmacol 2010; 79:1706-13. [DOI: 10.1016/j.bcp.2010.01.027] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 01/18/2010] [Accepted: 01/22/2010] [Indexed: 10/19/2022]
|
36
|
Jie W, Wang X, Huang L, Guo J, Kuang D, Zhu P, Li M, Zhao X, Duan Y, Wang G, Ao Q. Contribution of CXCR4(+)/PDGFRbeta(+) progenitor cells in hypoxic alveolar arterioles muscularization: role of myocardin. Cardiovasc Res 2010; 87:740-50. [PMID: 20484220 DOI: 10.1093/cvr/cvq147] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AIMS Bone marrow (BM) progenitor cells may contribute to vascular remodelling. The present study aimed to investigate the contribution of BM-derived CXCR4(+) (a CXC chemokine receptor) and PDGFRbeta(+) (platelet-derived growth factor receptor beta) progenitor cells in hypoxia-induced muscularization of alveolar arterioles. METHODS AND RESULTS Accumulation of GFP(+) (green fluorescent protein) cells was markedly increased in the pulmonary vasculature by the hypoxic (10% O(2,) 4 weeks) chimeric mice with transgenic GFP-tagged BM. After injection of BM-derived CXCR4(+)/PDGFRbeta(+) progenitor cells into C57BL/6J mice, followed by 6-week hypoxia, the cells were found to home to the alveolar arterioles and readily differentiated into smooth muscle cells (SMCs). Under the same hypoxic conditions, mice infused with myocardin lentiviral RNAi vector-transduced progenitor cells displayed lower myocardin expression in the muscularized alveolar arterioles, correlating with decreased pulmonary artery pressure and arteriole muscularization. In vitro experiments further confirmed that the differentiation of the progenitor cells into SMCs occurred under hypoxia (1% O(2)), and SMC differentiation could be suppressed when myocardin RNAi was administered. CONCLUSION Theses results suggest that myocardin may contribute to the differentiation of CXCR4(+)/PDGFRbeta(+) progenitor cells into SMCs induced by hypoxia, which leads to the muscularization of alveolar arterioles.
Collapse
Affiliation(s)
- Wei Jie
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Da Dao, Wuhan 430030, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Vo E, Hanjaya-Putra D, Zha Y, Kusuma S, Gerecht S. Smooth-Muscle-Like Cells Derived from Human Embryonic Stem Cells Support and Augment Cord-Like Structures In Vitro. Stem Cell Rev Rep 2010; 6:237-47. [DOI: 10.1007/s12015-010-9144-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
38
|
Barillot W, Tréguer K, Faucheux C, Fédou S, Thézé N, Thiébaud P. Induction and modulation of smooth muscle differentiation in Xenopus embryonic cells. Dev Dyn 2009; 237:3373-86. [PMID: 18855898 DOI: 10.1002/dvdy.21749] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
By comparison with skeletal or cardiac developmental programs, little is known regarding the specific factors that promote specification and differentiation of smooth muscle cells from pluripotent cells. We have analyzed the developmental expression of a subset of smooth muscle genes during Xenopus early development and showed that similar to mammals and avians, Xenopus smooth muscle myosin heavy chain (SM-MHC) is a highly specific marker of smooth muscle differentiation. Embryonic cells from animal pole explants of Xenopus blastula can be induced by basic fibroblast growth factor, Wnt, and bone morphogenetic protein signals to adopt the smooth muscle pathway. Explants from early embryos that contain neural crest cells can also differentiate into cells expressing smooth muscle genes. We examined the interplay of several transcription factors, that is SRF, myocardin, and GATA6, that induce the expression of SM-MHC in animal cap cells and found that myocardin-dependent expression of smooth muscle genes in animal cap cells is synergized by SRF but is strongly antagonized by GATA6.
Collapse
|
39
|
Costa-Silva B, da Costa MC, Melo FR, Neves CM, Alvarez-Silva M, Calloni GW, Trentin AG. Fibronectin promotes differentiation of neural crest progenitors endowed with smooth muscle cell potential. Exp Cell Res 2009; 315:955-67. [PMID: 19331824 DOI: 10.1016/j.yexcr.2009.01.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2008] [Revised: 01/11/2009] [Accepted: 01/17/2009] [Indexed: 12/12/2022]
Abstract
The neural crest (NC) is a model system used to investigate multipotency during vertebrate development. Environmental factors control NC cell fate decisions. Despite the well-known influence of extracellular matrix molecules in NC cell migration, the issue of whether they also influence NC cell differentiation has not been addressed at the single cell level. By analyzing mass and clonal cultures of mouse cephalic and quail trunk NC cells, we show for the first time that fibronectin (FN) promotes differentiation into the smooth muscle cell phenotype without affecting differentiation into glia, neurons, and melanocytes. Time course analysis indicated that the FN-induced effect was not related to massive cell death or proliferation of smooth muscle cells. Finally, by comparing clonal cultures of quail trunk NC cells grown on FN and collagen type IV (CLIV), we found that FN strongly increased both NC cell survival and the proportion of unipotent and oligopotent NC progenitors endowed with smooth muscle potential. In contrast, melanocytic progenitors were prominent in clonogenic NC cells grown on CLIV. Taken together, these results show that FN promotes NC cell differentiation along the smooth muscle lineage, and therefore plays an important role in fate decisions of NC progenitor cells.
Collapse
Affiliation(s)
- Bruno Costa-Silva
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário-Trindade, 88040-900 Florianópolis, S.C., Brazil
| | | | | | | | | | | | | |
Collapse
|
40
|
Kokudo T, Suzuki Y, Yoshimatsu Y, Yamazaki T, Watabe T, Miyazono K. Snail is required for TGFbeta-induced endothelial-mesenchymal transition of embryonic stem cell-derived endothelial cells. J Cell Sci 2008; 121:3317-24. [PMID: 18796538 DOI: 10.1242/jcs.028282] [Citation(s) in RCA: 252] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) plays important roles in various physiological and pathological processes, and is regulated by signaling pathways mediated by cytokines, including transforming growth factor beta (TGFbeta). Embryonic endothelial cells also undergo differentiation into mesenchymal cells during heart valve formation and aortic maturation. However, the molecular mechanisms that regulate such endothelial-mesenchymal transition (EndMT) remain to be elucidated. Here we show that TGFbeta plays important roles during mural differentiation of mouse embryonic stem cell-derived endothelial cells (MESECs). TGFbeta2 induced the differentiation of MESECs into mural cells, with a decrease in the expression of the endothelial marker claudin 5, and an increase in expression of the mural markers smooth muscle alpha-actin, SM22alpha and calponin, whereas a TGFbeta type I receptor kinase inhibitor inhibited EndMT. Among the transcription factors involved in EMT, Snail was induced by TGFbeta2 in MESECs. Tetracycline-regulated expression of Snail induced the differentiation of MESECs into mural cells, whereas knockdown of Snail expression abrogated TGFbeta2-induced mural differentiation of MESECs. These results indicate that Snail mediates the actions of endogenous TGFbeta signals that induce EndMT.
Collapse
Affiliation(s)
- Takashi Kokudo
- Department of Molecular Pathology, Graduate School of Medicine and the Global Center of Excellence Program for ;Integrative Life Science Based on the Study of Biosignaling Mechanisms', The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | |
Collapse
|
41
|
Matveeva NA, Latsis RV. Proliferation behavior of smooth muscle cells during ontogeny of the rat aorta. DOKLADY BIOLOGICAL SCIENCES : PROCEEDINGS OF THE ACADEMY OF SCIENCES OF THE USSR, BIOLOGICAL SCIENCES SECTIONS 2008; 419:143-5. [PMID: 18536286 DOI: 10.1134/s0012496608020221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- N A Matveeva
- Russian Cardiological Research and Technological Association, Ministry of Health of Russia, Tret'ya Cherepkovskaya 15a, Moscow, 121552, Russia
| | | |
Collapse
|
42
|
Wirz W, Antoine M, Tag CG, Gressner AM, Korff T, Hellerbrand C, Kiefer P. Hepatic stellate cells display a functional vascular smooth muscle cell phenotype in a three-dimensional co-culture model with endothelial cells. Differentiation 2008; 76:784-94. [PMID: 18177423 DOI: 10.1111/j.1432-0436.2007.00260.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hepatic stellate cells (HSCs) are pericytes of liver sinusoidal endothelial cells (LSECs) and activation of HSC into a myofibroblast-like phenotype (called transdifferentiation) is involved in several hepatic disease processes including neovascularization during liver metastasis, chronic and acute liver injury. While early smooth muscle cell (SMC) differentiation markers including SM alpha-actin and SM22alpha are expressed in a variety of non-SMC, expression of late-stage markers is far more restricted. Here, we found that in addition to early SMC markers, activated rat HSC express a large panel of characteristic late vascular SMC markers including SM myosin heavy chain, h1-calponin and h-caldesmon. Furthermore, myocardin, which is present exclusively in SMCs and cardiomyocytes and controls the transcription of a subset of early and late SMC markers, is highly expressed in activated HSC. We further studied activated HSC in a functional three-dimensional spheroidal co-culture system together with endothelial cells (EC). Co-culture spheroids of EC and SMC differentiate spontaneously and organize into a core of SMC and a surface layer of EC representing an inside-outside model of the physiological assembly of blood vessels. Replacing SMC by in vitro activated HSC resulted in a similar organized spheroid with differentiated, von-Willebrand factor producing, surface lining quiescent human umbilical vein endothelial cell and a core of HSC. In an in vitro angiogenesis assay, activated HSC induced quiescence in vascular EC-the hallmark of vascular SMC function. Co-spheroids of LSEC and activated HSC formed capillary-like sprouts in gel angiogenesis assays expressing the vascular EC marker VE-cadherin. Our findings indicate that activated HSC are capable to adapt a functional SMC phenotype and to induce formation of tubular sprouts by LSEC and vascular endothelial cells. Since tumors and tumor metastasis induce HSC activation, HSC may take part in tumor-induced neoangiogenesis by adapting SMC-like functions.
Collapse
Affiliation(s)
- W Wirz
- Institute of Clinical Chemistry and Pathobiochemistry, RWTH Aachen, D-52073 Germany
| | | | | | | | | | | | | |
Collapse
|
43
|
Tigges U, Hyer EG, Scharf J, Stallcup WB. FGF2-dependent neovascularization of subcutaneous Matrigel plugs is initiated by bone marrow-derived pericytes and macrophages. Development 2008; 135:523-32. [PMID: 18171688 DOI: 10.1242/dev.002071] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Vessel-like networks are quickly formed in subcutaneous FGF2-supplemented Matrigel plugs by two cell types: NG2(+) pericytes and F4/80(+) macrophages. Although not detected in these networks until 7 days after plug implantation, the appearance of CD31(+) endothelial cells marks the onset of vessel perfusion and the establishment of mature vessel morphology, with endothelial cells invested tightly by pericytes and more loosely by macrophages. Evidence that mature vessels develop from pericyte/macrophage networks comes from experiments in which 5-day plugs are transplanted into EGFP(+) recipients and allowed to mature. Fewer than 5% of pericytes in mature vessels are EGFP(+) in this paradigm, demonstrating their presence in the networks prior to plug transplantation. Endothelial cells represent the major vascular cell type recruited during later stages of vessel maturation. Bone marrow transplantation using EGFP(+) donors establishes that almost all macrophages and more than half of the pericytes in Matrigel vessels are derived from the bone marrow. By contrast, only 10% of endothelial cells exhibit a bone marrow origin. The vasculogenic, rather than angiogenic, nature of this neovascularization process is unique in that it is initiated by pericyte and macrophage progenitors, with endothelial cell recruitment occurring as a later step in the maturation process.
Collapse
Affiliation(s)
- Ulrich Tigges
- Burnham Institute for Medical Research, Cancer Research Center, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
44
|
Paye JMD, Partovian C, Simons M. Journeys in Coronary Angiogenesis. Angiogenesis 2008. [DOI: 10.1007/978-0-387-71518-6_47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
45
|
Abstract
The way in which multiple cell types organize themselves into a carefully sculpted, 3D labyrinth of vessels that regulate blood flow throughout the body has been a longstanding mystery. Clinicians familiar with congenital cardiovascular disease recognize how genetic variants and modest perturbations in this complex set of spatiotemporal interactions and stochastic processes can result in life-threatening anomalies. Although the mystery is not yet fully solved, we are poised at an exciting juncture, as insights from murine disease models are converging with advances in human genetics to shed new light on puzzling clinical phenotypes of vascular disease. The study by High et al. in this issue of the JCI establishes a model system that mimics clinical features of congenital cardiovascular disease and further defines the role of the Notch signaling pathway in the neural crest as an essential determinant of cardiovascular structure (see the related article beginning on page 353).
Collapse
Affiliation(s)
- Leonard M Anderson
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia 30310, USA
| | | |
Collapse
|
46
|
Le Ricousse-Roussanne S, Larghero J, Zini JM, Barateau V, Foubert P, Uzan G, Liu X, Lacassagne MN, Ternaux B, Robert I, Benbunan M, Vilquin JT, Vauchez K, Tobelem G, Marolleau JP. Ex vivo generation of mature and functional human smooth muscle cells differentiated from skeletal myoblasts. Exp Cell Res 2007; 313:1337-46. [PMID: 17362928 DOI: 10.1016/j.yexcr.2007.01.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2006] [Revised: 12/05/2006] [Accepted: 01/21/2007] [Indexed: 12/01/2022]
Abstract
We described the ex vivo production of mature and functional human smooth muscle cells (SMCs) derived from skeletal myoblasts. Initially, myoblasts expressed all myogenic cell-related markers such as Myf5, MyoD and Myogenin and differentiate into myotubes. After culture in a medium containing vascular endothelial growth factor (VEGF), these cells were shown to have adopted a differentiated SMC identity as demonstrated by alphaSMA, SM22alpha, calponin and smooth muscle-myosin heavy chain expression. Moreover, the cells cultured in the presence of VEGF did not express MyoD anymore and were unable to fuse in multinucleated myotubes. We demonstrated that myoblasts-derived SMCs (MDSMCs) interacted with endothelial cells to form, in vitro, a capillary-like network in three-dimensional collagen culture and, in vivo, a functional vascular structure in a Matrigel implant in nonobese diabetic-severe combined immunodeficient mice. Based on the easily available tissue source and their differentiation into functional SMCs, these data argue that skeletal myoblasts might represent an important tool for SMCs-based cell therapy.
Collapse
MESH Headings
- Animals
- Biomarkers/analysis
- Cell Differentiation
- Cells, Cultured
- Collagen/pharmacology
- Drug Combinations
- Epithelial Cells/physiology
- Female
- Humans
- Laminin/pharmacology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Muscle Fibers, Skeletal/physiology
- Myoblasts, Skeletal/drug effects
- Myoblasts, Skeletal/metabolism
- Myoblasts, Skeletal/physiology
- Myoblasts, Skeletal/transplantation
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- Myocytes, Smooth Muscle/transplantation
- Neovascularization, Physiologic
- Proteoglycans/pharmacology
- Tissue Culture Techniques
- Transplantation, Heterologous
- Vascular Endothelial Growth Factor A/pharmacology
Collapse
|
47
|
Abstract
The origins of vascular smooth muscle are far more diverse than previously thought. Lineage mapping studies show that the segmental organization of early vertebrate embryos leaves footprints on the adult vascular system in the form of a mosaic pattern of different smooth muscle types. Moreover, evolutionarily conserved tissue forming pathways produce vascular smooth muscle from a variety of unanticipated sources. A closer look at the diversity of smooth muscle origins in vascular development provides new perspectives about how blood vessels differ from one another and why they respond in disparate ways to common risk factors associated with vascular disease. The origins of vascular smooth muscle are far more diverse than previously thought. A closer look at the diversity of smooth muscle origins in vascular development provides new perspectives about how blood vessels differ from one another and why they respond in disparate ways to common risk factors associated with vascular disease.
Collapse
Affiliation(s)
- Mark W Majesky
- Department of Medicine, Carolina Cardiovascular Biology Center, University of North Carolina, Chapel Hill, NC 27599-7126, USA.
| |
Collapse
|
48
|
Li F, Luo Z, Huang W, Lu Q, Wilcox CS, Jose PA, Chen S. Response gene to complement 32, a novel regulator for transforming growth factor-beta-induced smooth muscle differentiation of neural crest cells. J Biol Chem 2007; 282:10133-7. [PMID: 17327222 DOI: 10.1074/jbc.c600225200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We previously developed a robust in vitro model system for vascular smooth muscle cell (VSMC) differentiation from neural crest cell line Monc-1 upon transforming growth factor-beta (TGF-beta) induction. Further studies demonstrated that both Smad and RhoA signaling are critical for TGF-beta-induced VSMC development. To identify downstream targets, we performed Affymetrix cDNA array analysis of Monc-1 cells and identified a gene named response gene to complement 32 (RGC-32) to be important for the VSMC differentiation. RGC-32 expression was increased 5-fold after 2 h and 50-fold after 24 h of TGF-beta induction. Knockdown of RGC-32 expression in Monc-1 cells by small interfering RNA significantly inhibited the expression of multiple smooth muscle marker genes, including SM alpha-actin (alpha-SMA), SM22alpha, and calponin. Of importance, the inhibition of RGC-32 expression correlated with the reduction of alpha-SMA while not inhibiting smooth muscle-unrelated c-fos gene expression, suggesting that RGC-32 is an important protein factor for VSMC differentiation from neural crest cells. Moreover, RGC-32 overexpression significantly enhanced TGF-beta-induced alpha-SMA, SM22alpha, and SM myosin heavy chain promoter activities in both Monc-1 and C3H10T1/2 cells. The induction of VSMC gene promoters by RGC-32 appears to be CArG-dependent. These data suggest that RGC-32 controls VSMC differentiation by regulating marker gene transcription in a CArG-dependent manner. Further studies revealed that both Smad and RhoA signaling are important for RGC-32 activation.
Collapse
Affiliation(s)
- Fengmin Li
- Department of Pediatrics, Georgetown University Medical Center, Washington, DC 20057, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Zhu P, Huang L, Ge X, Yan F, Wu R, Ao Q. Transdifferentiation of pulmonary arteriolar endothelial cells into smooth muscle-like cells regulated by myocardin involved in hypoxia-induced pulmonary vascular remodelling. Int J Exp Pathol 2007; 87:463-74. [PMID: 17222214 PMCID: PMC2517388 DOI: 10.1111/j.1365-2613.2006.00503.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Myocardin gene has been identified as a master regulator of smooth muscle cell differentiation. Smooth muscle cells play a critical role in the pathogenesis of hypoxia-induced pulmonary hypertension (PH) and pulmonary vascular remodelling (PVR). The purpose of this study was to investigate the change of myocardin gene expression in the pulmonary vessels of hypoxia-induced PH affected by Sildenafil treatment and the involvement of endothelial cells transdifferentiation into smooth muscle cells in the process of hypoxia-induced PH and PVR. Myocardin and relative markers were investigated in animal models and cultured endothelial cells. Mean pulmonary artery pressure (mPAP) was measured. Immunohistochemistry and immunofluorescence were used to show the expression of smooth muscle alpha-actin (SMA), in situ hybridization (ISH) and reverse transcription polymerase chain reaction (RT-PCR) were performed respectively to detect the myocardin and SMA expression at mRNA levels. Small interfering RNA (siRNA) induced suppression of myocardin in cultured cells. We confirmed that hypoxia induced the PH and PVR in rats. Sildenafil could attenuate the hypoxia-induced PH. We found that myocardin mRNA expression is upregulated significantly in the hypoxic pulmonary vessels and cultured cells but downregulated in PH with Sildenafil treatment. The porcine pulmonary artery endothelial cells (PAECs) transdifferentiate into smooth muscle-like cells in hypoxic culture while the transdifferentiation did not occur when SiRNA of myocardin was applied. Our results suggest that myocardin gene, as a marker of smooth muscle cell differentiation, was expressed in the pulmonary vessels in hypoxia-induced PH rats, which could be downregulated by Sildenafil treatment, as well as in hypoxic cultured endothelial cells. Hypoxia induced the transdifferentiation of endothelial cells of vessels into smooth muscle-like cells which was regulated by myocardin.
Collapse
MESH Headings
- Actins/analysis
- Actins/genetics
- Animals
- Arterioles
- Biomarkers/analysis
- Cell Differentiation
- Cells, Cultured
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Gene Expression Regulation
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypoxia/genetics
- Hypoxia/metabolism
- Immunohistochemistry/methods
- Male
- Models, Animal
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Piperazines/therapeutic use
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Purines
- RNA Interference
- Rats
- Rats, Sprague-Dawley
- Reverse Transcriptase Polymerase Chain Reaction
- Sildenafil Citrate
- Sulfones
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Vasodilator Agents/therapeutic use
Collapse
Affiliation(s)
- Pengcheng Zhu
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan City, China
- Key Laboratory of Pulmonary Diseases, Ministry of Health of ChinaWuhan City, China
| | - Lei Huang
- Department of Gynaecology and Obstetrics, The Central Hospital of WuhanWuhan City, China
| | - Xiaona Ge
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan City, China
- Key Laboratory of Pulmonary Diseases, Ministry of Health of ChinaWuhan City, China
| | - Fei Yan
- Department of Internal Medicine, Zhongshan Hospital of Hubei ProvinceWuhan City, China
| | - Renliang Wu
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan City, China
- Key Laboratory of Pulmonary Diseases, Ministry of Health of ChinaWuhan City, China
| | - Qilin Ao
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan City, China
- Key Laboratory of Pulmonary Diseases, Ministry of Health of ChinaWuhan City, China
| |
Collapse
|
50
|
De Coppi P, Callegari A, Chiavegato A, Gasparotto L, Piccoli M, Taiani J, Pozzobon M, Boldrin L, Okabe M, Cozzi E, Atala A, Gamba P, Sartore S. Amniotic fluid and bone marrow derived mesenchymal stem cells can be converted to smooth muscle cells in the cryo-injured rat bladder and prevent compensatory hypertrophy of surviving smooth muscle cells. J Urol 2007; 177:369-76. [PMID: 17162093 DOI: 10.1016/j.juro.2006.09.103] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2005] [Indexed: 12/25/2022]
Abstract
PURPOSE Wound healing of the cryo-injured bladder can bring about organ remodeling because of incomplete reconstitution of depleted smooth muscle cells. Stem cell transplantation could be beneficial to improve smooth muscle cell regeneration and/or modulate the remodeling process. The repair of bladder injury using adult-type stem cells would be useful for adult urological patients but unsuited for neonatal patients, in whom major benefits are likely to derive from fetal-type stem cells. MATERIALS AND METHODS The smooth muscle cell differentiation potential of fetal-type vs adult-type stem cells was evaluated by injecting green fluorescent protein labeled mesenchymal stem cells from rat amniotic fluid or bone marrow, respectively, in cryo-injured rat bladder walls. RESULTS At 30 days after transplantation only a few fetal-type or adult-type mesenchymal stem cells gave rise to enteric or vascular smooth muscle cells, whereas most mesenchymal stem cells appeared incapable of specific differentiation. In vitro co-culture experiments of smooth muscle cells with fetal-type or adult-type mesenchymal stem cells selectively labeled with distinct fluorochromes showed the presence of hybrid cells, suggesting that some mesenchymal stem cells can undergo cell fusion. Surprisingly the major effect of rat bone marrow or amniotic fluid mesenchymal stem cell transplantation seemed to be preventing cryo-injury induced hypertrophy of surviving smooth muscle cells. CONCLUSIONS In this model stem cell transplantation has a limited effect on smooth muscle cell regeneration. Instead it can regulate post-injury bladder remodeling, possibly via a paracrine mechanism.
Collapse
Affiliation(s)
- Paolo De Coppi
- Department of Pediatrics (Pediatric Oncoheamatology), University of Padua, Padua, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|