1
|
Rahmawati FN, Takakura N. Development and aging of resident endothelial stem cells in pre-existing blood vessels. Exp Hematol 2025:104795. [PMID: 40311858 DOI: 10.1016/j.exphem.2025.104795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/09/2025] [Accepted: 04/13/2025] [Indexed: 05/03/2025]
Abstract
Organ-specific somatic stem cells play an important role in supporting tissue turnover and facilitating regeneration on injury. Hematopoietic stem cells are one of the most established organ-specific somatic cells that have been frequently used for transplantation therapy. Recently, there has been a growing interest in other organ-specific somatic cells, including vascular endothelial stem cells (VESCs). We have previously reported on the use of CD157 and CD200 as markers to isolate VESCs from adult mouse organs, particularly the liver. In this review, we aimed to summarize, based on our previous research, how CD157⁺CD200⁺ VESCs in the liver develop from the fetal stage to postnatal life, what transcriptional regulatory mechanisms govern them, and how VESCs change with aging.
Collapse
Affiliation(s)
- Fitriana N Rahmawati
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.
| | - Nobuyuki Takakura
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Immunology Frontier Research Center, Osaka University, Suita, Japan; Center for Infectious Disease Education and Research, Osaka University, Suita, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Japan.
| |
Collapse
|
2
|
Wang X, Yao F, Yang L, Han D, Zeng Y, Huang Z, Yang C, Lin B, Chen X. Macrophage extracellular vesicle-packaged miR-23a-3p impairs maintenance and angiogenic capacity of human endothelial progenitor cells in neonatal hyperoxia-induced lung injury. Stem Cell Res Ther 2024; 15:295. [PMID: 39256862 PMCID: PMC11389047 DOI: 10.1186/s13287-024-03920-z] [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] [Received: 06/25/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Premature infants requiring mechanical ventilation and supplemental oxygen for respiratory support are at increased risk for bronchopulmonary dysplasia (BPD), wherein inflammation have been proposed as a driver of hyperoxia-induced injuries, including persistent loss of endothelial progenitor cells (EPCs), impaired vascularization and eventual alveolar simplification in BPD lungs. However, the underlying mechanisms linking these phenomena remain poorly defined. METHODS We used clodronate liposomes to deplete macrophages in a mouse model of neonatal hyperoxia-induced lung injury to evaluate if EPC loss in BPD lungs could be an effect of macrophage infiltration. We further generated in vitro culture systems initiated with cord blood (CB)-derived CD34+ EPCs and neonatal macrophages either polarized from CB-derived monocytes or isolated from tracheal aspirates of human preterm infants requiring mechanical ventilation and oxygen supplementation, to identify EV-transmitted molecular mechanism that is critical for inhibitory actions of hyperoxic macrophages on EPCs. RESULTS Initial experiments using mouse model identified the crucial role of macrophage infiltration in eliciting significant reduction of c-Kit+ EPCs in BPD lungs. Further examination of this concept in human system, we found that hyperoxia-exposed neonatal macrophages hamper human CD34+ EPC maintenance and impair endothelial function in the differentiated progeny via the EV transmission of miR-23a-3p. Notably, treatment with antagomiR-23a-3p to silence miR-23a-3p in vivo enhances c-Kit+ EPC maintenance, and increases capillary density, and consequently mitigates simplified alveolarization in BPD lungs. CONCLUSION Our findings highlight the importance of pulmonary intercellular communication in the pathophysiology of BPD, by identifying a linkage through vesicle transfer of miR-23a-3p from hyperoxic macrophages to EPCs, and thus demonstrating potential for novel therapeutic target in BPD.
Collapse
Affiliation(s)
- Xuan Wang
- Laboratory of Neonatology, Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518000, China
| | - Fang Yao
- Laboratory of Neonatology, Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518000, China
| | - Lingling Yang
- Laboratory of Neonatology, Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518000, China
| | - Dongshan Han
- Laboratory of Neonatology, Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518000, China
| | - Yali Zeng
- Laboratory of Neonatology, Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518000, China
- The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Zilu Huang
- Laboratory of Neonatology, Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518000, China
| | - Chuanzhong Yang
- Laboratory of Neonatology, Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518000, China
- The First Clinical Medical School, Southern Medical University, Guangzhou, China
- Shenzhen Key Laboratory of Maternal and Child Health and Diseases, Shenzhen, China
| | - Bingchun Lin
- Laboratory of Neonatology, Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518000, China.
| | - Xueyu Chen
- Laboratory of Neonatology, Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518000, China.
- The First Clinical Medical School, Southern Medical University, Guangzhou, China.
- Shenzhen Key Laboratory of Maternal and Child Health and Diseases, Shenzhen, China.
| |
Collapse
|
3
|
Zhang Y, Guan Z, Gong H, Ni Z, Xiao Q, Guo X, Xu Q. The Role of Progenitor Cells in the Pathogenesis of Arteriosclerosis. CARDIOLOGY DISCOVERY 2024; 4:231-244. [DOI: 10.1097/cd9.0000000000000130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The increasing incidence of arteriosclerosis has become a significant global health burden. Arteriosclerosis is characterized by the thickening and hardening of arterial walls, which can lead to the narrowing or complete blockage of blood vessels. However, the pathogenesis of the disease remains incompletely understood. Recent research has shown that stem and progenitor cells found in the bone marrow and local vessel walls play a role in the development of arteriosclerosis by differentiating into various types of vascular cells, including endothelial cells, smooth muscle cells, fibroblasts, and inflammatory cells. This review aims to provide a comprehensive understanding of the role of stem and progenitor cells in the pathogenesis of arteriosclerosis, shedding light on the underlying mechanisms and potential therapeutic approaches for this disease.
Collapse
Affiliation(s)
- Yuesheng Zhang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Ziyin Guan
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Hui Gong
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Zhichao Ni
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Xiaogang Guo
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
4
|
Lin Y, Gil CH, Banno K, Yokoyama M, Wingo M, Go E, Prasain N, Liu Y, Hato T, Naito H, Wakabayashi T, Sominskaia M, Gao M, Chen K, Geng F, Salinero JMG, Chen S, Shelley WC, Yoshimoto M, Li Calzi S, Murphy MP, Horie K, Grant MB, Schreiner R, Redmond D, Basile DP, Rafii S, Yoder MC. ABCG2-Expressing Clonal Repopulating Endothelial Cells Serve to Form and Maintain Blood Vessels. Circulation 2024; 150:451-465. [PMID: 38682338 PMCID: PMC11300167 DOI: 10.1161/circulationaha.122.061833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 03/05/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Most organs are maintained lifelong by resident stem/progenitor cells. During development and regeneration, lineage-specific stem/progenitor cells can contribute to the growth or maintenance of different organs, whereas fully differentiated mature cells have less regenerative potential. However, it is unclear whether vascular endothelial cells (ECs) are also replenished by stem/progenitor cells with EC-repopulating potential residing in blood vessels. It has been reported recently that some EC populations possess higher clonal proliferative potential and vessel-forming capacity compared with mature ECs. Nevertheless, a marker to identify vascular clonal repopulating ECs (CRECs) in murine and human individuals is lacking, and, hence, the mechanism for the proliferative, self-renewal, and vessel-forming potential of CRECs is elusive. METHODS We analyzed colony-forming, self-renewal, and vessel-forming potential of ABCG2 (ATP binding cassette subfamily G member 2)-expressing ECs in human umbilical vessels. To study the contribution of Abcg2-expressing ECs to vessel development and regeneration, we developed Abcg2CreErt2;ROSA TdTomato mice and performed lineage tracing during mouse development and during tissue regeneration after myocardial infarction injury. RNA sequencing and chromatin methylation chromatin immunoprecipitation followed by sequencing were conducted to study the gene regulation in Abcg2-expressing ECs. RESULTS In human and mouse vessels, ECs with higher ABCG2 expression (ABCECs) possess higher clonal proliferative potential and in vivo vessel-forming potential compared with mature ECs. These cells could clonally contribute to vessel formation in primary and secondary recipients after transplantation. These features of ABCECs meet the criteria of CRECs. Results from lineage tracing experiments confirm that Abcg2-expressing CRECs (AbcCRECs) contribute to arteries, veins, and capillaries in cardiac tissue development and vascular tissue regeneration after myocardial infarction. Transcriptome and epigenetic analyses reveal that a gene expression signature involved in angiogenesis and vessel development is enriched in AbcCRECs. In addition, various angiogenic genes, such as Notch2 and Hey2, are bivalently modified by trimethylation at the 4th and 27th lysine residue of histone H3 (H3K4me3 and H3K27me3) in AbcCRECs. CONCLUSIONS These results are the first to establish that a single prospective marker identifies CRECs in mice and human individuals, which holds promise to provide new cell therapies for repair of damaged vessels in patients with endothelial dysfunction.
Collapse
Affiliation(s)
- Yang Lin
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Chang-Hyun Gil
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kimihiko Banno
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Physiology II, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Masataka Yokoyama
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Matthew Wingo
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ellen Go
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Pediatrics, Division of Pediatric Rheumatology, Riley Hospital for Children, Indianapolis, IN
| | - Nutan Prasain
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ying Liu
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Takashi Hato
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hisamichi Naito
- Department of Vascular Physiology, Kanazawa University School of Medicine, Kanazawa, Japan
| | - Taku Wakabayashi
- Department of Vascular Physiology, Kanazawa University School of Medicine, Kanazawa, Japan
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Wills Eye Hospital, Mid Atlantic Retina, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Musia Sominskaia
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Meng Gao
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Kevin Chen
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Fuqiang Geng
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jesus Maria Gomez Salinero
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sisi Chen
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - W. Christopher. Shelley
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Momoko Yoshimoto
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Immunobiology, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Sergio Li Calzi
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham, Alabama USA
| | - Michael P. Murphy
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kyoji Horie
- Department of Physiology II, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Maria B. Grant
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham, Alabama USA
| | - Ryan Schreiner
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David Redmond
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David P. Basile
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Mervin C. Yoder
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Jeong H, Kim RI, Koo H, Choi YH, Kim M, Roh H, Park SG, Sung JH, Kim KL, Suh W. Stem cell factor and cKIT modulate endothelial glycolysis in hypoxia. Cardiovasc Res 2024; 120:745-755. [PMID: 38507654 DOI: 10.1093/cvr/cvae058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 10/05/2023] [Accepted: 12/12/2023] [Indexed: 03/22/2024] Open
Abstract
AIMS In hypoxia, endothelial cells (ECs) proliferate, migrate, and form new vasculature in a process called angiogenesis. Recent studies have suggested that ECs rely on glycolysis to meet metabolic needs for angiogenesis in ischaemic tissues, and several studies have investigated the molecular mechanisms integrating angiogenesis and endothelial metabolism. Here, we investigated the role of stem cell factor (SCF) and its receptor, cKIT, in regulating endothelial glycolysis during hypoxia-driven angiogenesis. METHODS AND RESULTS SCF and cKIT signalling increased the glucose uptake, lactate production, and glycolysis in human ECs under hypoxia. Mechanistically, SCF and cKIT signalling enhanced the expression of genes encoding glucose transporter 1 (GLUT1) and glycolytic enzymes via Akt- and ERK1/2-dependent increased translation of hypoxia inducible factor 1A (HIF1A). In hypoxic conditions, reduction of glycolysis and HIF-1α expression using chemical inhibitors significantly reduced the SCF-induced in vitro angiogenesis in human ECs. Compared with normal mice, mice with oxygen-induced retinopathy (OIR), characterized by ischaemia-driven pathological retinal neovascularization, displayed increased levels of SCF, cKIT, HIF-1α, GLUT1, and glycolytic enzymes in the retina. Moreover, cKIT-positive neovessels in the retina of mice with OIR showed elevated expression of GLUT1 and glycolytic enzymes. Further, blocking SCF and cKIT signalling using anti-SCF neutralizing IgG and cKIT mutant mice significantly reduced the expression of HIF-1α, GLUT1, and glycolytic enzymes and decreased the pathological neovascularization in the retina of mice with OIR. CONCLUSION We demonstrated that SCF and cKIT signalling regulate angiogenesis by controlling endothelial glycolysis in hypoxia and elucidated the SCF/cKIT/HIF-1α axis as a novel metabolic regulation pathway during hypoxia-driven pathological angiogenesis.
Collapse
Affiliation(s)
- Hayoung Jeong
- Department of Global Innovative Drug, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea
- College of Pharmacy, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul 06974, Korea
| | - Ryul-I Kim
- Department of Global Innovative Drug, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea
- College of Pharmacy, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul 06974, Korea
| | - Hyunwoo Koo
- College of Pharmacy, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul 06974, Korea
| | - Yang Hee Choi
- Department of Global Innovative Drug, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea
- College of Pharmacy, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul 06974, Korea
| | - Minju Kim
- Department of Global Innovative Drug, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea
- College of Pharmacy, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul 06974, Korea
| | - Hyejin Roh
- Department of Global Innovative Drug, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea
- College of Pharmacy, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul 06974, Korea
| | - Sang Gyu Park
- College of Pharmacy, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon 16499, Korea
| | - Jong-Hyuk Sung
- College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Korea
| | - Koung Li Kim
- College of Pharmacy, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul 06974, Korea
| | - Wonhee Suh
- Department of Global Innovative Drug, Graduate School of Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea
- College of Pharmacy, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul 06974, Korea
| |
Collapse
|
6
|
Matsui Y, Muramatsu F, Nakamura H, Noda Y, Matsumoto K, Kishima H, Takakura N. Brain-derived endothelial cells are neuroprotective in a chronic cerebral hypoperfusion mouse model. Commun Biol 2024; 7:338. [PMID: 38499610 PMCID: PMC10948829 DOI: 10.1038/s42003-024-06030-x] [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] [Received: 04/10/2023] [Accepted: 03/08/2024] [Indexed: 03/20/2024] Open
Abstract
Whether organ-specific regeneration is induced by organ-specific endothelial cells (ECs) remains unelucidated. The formation of white matter lesions due to chronic cerebral hypoperfusion causes cognitive decline, depression, motor dysfunction, and even acute ischemic stroke. Vascular ECs are an important target for treating chronic cerebral hypoperfusion. Brain-derived ECs transplanted into a mouse chronic cerebral hypoperfusion model showed excellent angiogenic potential. They were also associated with reducing both white matter lesions and brain dysfunction possibly due to the high expression of neuroprotective humoral factors. The in vitro coculture of brain cells with ECs from several diverse organs suggested the function of brain-derived endothelium is affected within a brain environment due to netrin-1 and Unc 5B systems. We found brain CD157-positive ECs were more proliferative and beneficial in a mouse model of chronic cerebral hypoperfusion than CD157-negative ECs upon inoculation. We propose novel methods to improve the symptoms of chronic cerebral hypoperfusion using CD157-positive ECs.
Collapse
Affiliation(s)
- Yuichi Matsui
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Fumitaka Muramatsu
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Hajime Nakamura
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshimi Noda
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Kinnosuke Matsumoto
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Haruhiko Kishima
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Nobuyuki Takakura
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.
- World Premier Institute Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan.
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan.
| |
Collapse
|
7
|
Li Y, Liu Z, Han X, Liang F, Zhang Q, Huang X, Shi X, Huo H, Han M, Liu X, Zhu H, He L, Shen L, Hu X, Wang J, Wang QD, Smart N, Zhou B, He B. Dynamics of Endothelial Cell Generation and Turnover in Arteries During Homeostasis and Diseases. Circulation 2024; 149:135-154. [PMID: 38084582 DOI: 10.1161/circulationaha.123.064301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 10/06/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND Endothelial cell (EC) generation and turnover by self-proliferation contributes to vascular repair and regeneration. The ability to accurately measure the dynamics of EC generation would advance our understanding of cellular mechanisms of vascular homeostasis and diseases. However, it is currently challenging to evaluate the dynamics of EC generation in large vessels such as arteries because of their infrequent proliferation. METHODS By using dual recombination systems based on Cre-loxP and Dre-rox, we developed a genetic system for temporally seamless recording of EC proliferation in vivo. We combined genetic recording of EC proliferation with single-cell RNA sequencing and gene knockout to uncover cellular and molecular mechanisms underlying EC generation in arteries during homeostasis and disease. RESULTS Genetic proliferation tracing reveals that ≈3% of aortic ECs undergo proliferation per month in adult mice during homeostasis. The orientation of aortic EC division is generally parallel to blood flow in the aorta, which is regulated by the mechanosensing protein Piezo1. Single-cell RNA sequencing analysis reveals 4 heterogeneous aortic EC subpopulations with distinct proliferative activity. EC cluster 1 exhibits transit-amplifying cell features with preferential proliferative capacity and enriched expression of stem cell markers such as Sca1 and Sox18. EC proliferation increases in hypertension but decreases in type 2 diabetes, coinciding with changes in the extent of EC cluster 1 proliferation. Combined gene knockout and proliferation tracing reveals that Hippo/vascular endothelial growth factor receptor 2 signaling pathways regulate EC proliferation in large vessels. CONCLUSIONS Genetic proliferation tracing quantitatively delineates the dynamics of EC generation and turnover, as well as EC division orientation, in large vessels during homeostasis and disease. An EC subpopulation in the aorta exhibits more robust cell proliferation during homeostasis and type 2 diabetes, identifying it as a potential therapeutic target for vascular repair and regeneration.
Collapse
Affiliation(s)
- Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Zixin Liu
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Ximeng Han
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Feng Liang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Qianyu Zhang
- School of Life Science and Technology, ShanghaiTech University, China (Q.Z., M.H., B.Z.)
| | - Xiuzhen Huang
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Xin Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Huanhuan Huo
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Maoying Han
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
- School of Life Science and Technology, ShanghaiTech University, China (Q.Z., M.H., B.Z.)
| | - Xiuxiu Liu
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Huan Zhu
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Lingjuan He
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China (L.H.)
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Xinyang Hu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (X.H., J.W.)
| | - Jian'an Wang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (X.H., J.W.)
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (Q.D.W.)
| | - Nicola Smart
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, UK (N.S.)
| | - Bin Zhou
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
- School of Life Science and Technology, ShanghaiTech University, China (Q.Z., M.H., B.Z.)
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, China (B.Z.)
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| |
Collapse
|
8
|
Lin A, Brittan M, Baker AH, Dimmeler S, Fisher EA, Sluimer JC, Misra A. Clonal Expansion in Cardiovascular Pathology. JACC Basic Transl Sci 2024; 9:120-144. [PMID: 38362345 PMCID: PMC10864919 DOI: 10.1016/j.jacbts.2023.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 02/17/2024]
Abstract
Clonal expansion refers to the proliferation and selection of advantageous "clones" that are better suited for survival in a Darwinian manner. In recent years, we have greatly enhanced our understanding of cell clonality in the cardiovascular context. However, our knowledge of the underlying mechanisms behind this clonal selection is still severely limited. There is a transpiring pattern of clonal expansion of smooth muscle cells and endothelial cells-and, in some cases, macrophages-in numerous cardiovascular diseases irrespective of their differing microenvironments. These findings indirectly suggest the possible existence of stem-like vascular cells which are primed to respond during disease. Subsequent clones may undergo further phenotypic changes to adopt either protective or detrimental roles. By investigating these clone-forming vascular cells, we may be able to harness this inherent clonal nature for future therapeutic intervention. This review comprehensively discusses what is currently known about clonal expansion across the cardiovascular field. Comparisons of the clonal nature of vascular cells in atherosclerosis (including clonal hematopoiesis of indeterminate potential), pulmonary hypertension, aneurysm, blood vessel injury, ischemia- and tumor-induced angiogenesis, and cerebral cavernous malformations are evaluated. Finally, we discuss the potential clinical implications of these findings and propose that proper understanding and specific targeting of these clonal cells may provide unique therapeutic options for the treatment of these cardiovascular conditions.
Collapse
Affiliation(s)
- Alexander Lin
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, New South Wales, Australia
| | - Mairi Brittan
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew H. Baker
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- CARIM School for Cardiovascular Sciences, Department of Pathology, Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Edward A. Fisher
- Department of Medicine/Division of Cardiology, New York University Grossman School of Medicine, New York, New York, USA
- Cardiovascular Research Center, New York University Grossman School of Medicine, New York, New York, USA
| | - Judith C. Sluimer
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- CARIM School for Cardiovascular Sciences, Department of Pathology, Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
| | - Ashish Misra
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
9
|
Shimizu S, Iba T, Naito H, Rahmawati FN, Konishi H, Jia W, Muramatsu F, Takakura N. Aging impairs the ability of vascular endothelial stem cells to generate endothelial cells in mice. Angiogenesis 2023; 26:567-580. [PMID: 37563497 PMCID: PMC10542733 DOI: 10.1007/s10456-023-09891-8] [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] [Received: 02/21/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023]
Abstract
Tissue-resident vascular endothelial stem cells (VESCs), marked by expression of CD157, possess long-term repopulating potential and contribute to vascular regeneration and homeostasis in mice. Stem cell exhaustion is regarded as one of the hallmarks of aging and is being extensively studied in several types of tissue-resident stem cells; however, how aging affects VESCs has not been clarified yet. In the present study, we isolated VESCs from young and aged mice to compare their potential to differentiate into endothelial cells in vitro and in vivo. Here, we report that the number of liver endothelial cells (ECs) including VESCs was lower in aged (27-28 month-old) than young (2-3 month-old) mice. In vitro culture of primary VESCs revealed that the potential to generate ECs is impaired in aged VESCs isolated from liver and lung relative to young VESCs. Orthotopic transplantation of VESCs showed that aged VESCs and their progeny expand less efficiently than their young counterparts when transplanted into aged mice, but they are equally functional in young recipients. Gene expression analysis indicated that inflammatory signaling was more activated in aged ECs including VESCs. Using single-cell RNA sequencing data from the Tabula Muris Consortium, we show that T cells and monocyte/macrophage lineage cells including Kupffer cells are enriched in the aged liver. These immune cells produce IL-1β and several chemokines, suggesting the possible involvement of age-associated inflammation in the functional decline of VESCs with age.
Collapse
Affiliation(s)
- Shota Shimizu
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Tomohiro Iba
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
- Department of Physiology, Kanazawa University School of Medicine, Ishikawa, Japan
| | - Hisamichi Naito
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
- Department of Physiology, Kanazawa University School of Medicine, Ishikawa, Japan
| | - Fitriana Nur Rahmawati
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Hirotaka Konishi
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Weizhen Jia
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Fumitaka Muramatsu
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Nobuyuki Takakura
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka, 565-0871, Japan.
- World Premier Institute Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan.
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan.
| |
Collapse
|
10
|
Link PA, Farkas L, Heise RL. Using extracellular matrix derived from sugen-chronic hypoxia lung tissue to study pulmonary arterial hypertension. Front Pharmacol 2023; 14:1192798. [PMID: 37731734 PMCID: PMC10507686 DOI: 10.3389/fphar.2023.1192798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 08/09/2023] [Indexed: 09/22/2023] Open
Abstract
Pulmonary arterial hypertension has characteristic changes to the mechanical environment, extracellular matrix, and cellular proliferation. In order to develop a culture system to investigate extracellular matrix (ECM) compositional-dependent changes in pulmonary arterial hypertension, we decellularized and characterized protein and lipid profiles from healthy and Sugen-Chronic Hypoxia rat lungs. Significant changes in lipid profiles were observed in intact Sugen-Hypoxia lungs compared with healthy controls. Decellularized lung matrix retained lipids in measurable quantities in both healthy and Sugen-Chronic Hypoxia samples. Proteomics revealed significantly changed proteins associated with pulmonary arterial hypertension in the decellularized Sugen-Chronic Hypoxia lung ECM. We then investigated the potential role of healthy vs. Sugen-Chronic Hypoxia ECM with controlled substrate stiffness to determine if the ECM composition regulated endothelial cell morphology and phenotype. CD117+ rat lung endothelial cell clones were plated on the variable stiffness gels and cellular proliferation, morphology, and gene expression were quantified. Sugen-Chronic Hypoxia ECM on healthy stiffness gels produced significant changes in cellular gene expression levels of Bmp2, Col1α1, Col3α1 and Fn1. The signaling and cell morphology observed at low substrate stiffness suggests early changes to the ECM composition can initiate processes associated with disease progression. These data suggest that Sugen-Chronic Hypoxia ECM can be used to investigate cell-ECM interactions relevant to pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Patrick A. Link
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Laszlo Farkas
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Rebecca L. Heise
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
11
|
Shan HJ, Jiang K, Zhao MZ, Deng WJ, Cao WH, Li JJ, Li KR, She C, Luo WF, Yao J, Zhou XZ, Zhang D, Cao C. SCF/c-Kit-activated signaling and angiogenesis require Gαi1 and Gαi3. Int J Biol Sci 2023; 19:1910-1924. [PMID: 37063428 PMCID: PMC10092767 DOI: 10.7150/ijbs.82855] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/03/2023] [Indexed: 04/18/2023] Open
Abstract
The stem cell factor (SCF) binds to c-Kit in endothelial cells, thus activating downstream signaling and angiogenesis. Herein, we examined the role of G protein subunit alpha inhibitory (Gαi) proteins in this process. In MEFs and HUVECs, Gαi1/3 was associated with SCF-activated c-Kit, promoting c-Kit endocytosis, and binding of key adaptor proteins, subsequently transducing downstream signaling. SCF-induced Akt-mTOR and Erk activation was robustly attenuated by Gαi1/3 silencing or knockout (KO), or due to dominant negative mutations but was strengthened substantially following ectopic overexpression of Gαi1/3. SCF-induced HUVEC proliferation, migration, and capillary tube formation were suppressed after Gαi1/3 silencing or KO, or due to dominant negative mutations. In vivo, endothelial knockdown of Gαi1/3 by intravitreous injection of endothelial-specific shRNA adeno-associated virus (AAV) potently reduced SCF-induced signaling and retinal angiogenesis in mice. Moreover, mRNA and protein expressions of SCF increased significantly in the retinal tissues of streptozotocin-induced diabetic retinopathy (DR) mice. SCF silencing, through intravitreous injection of SCF shRNA AAV, inhibited pathological retinal angiogenesis and degeneration of retinal ganglion cells in DR mice. Finally, the expression of SCF and c-Kit increased in proliferative retinal tissues of human patients with proliferative DR. Taken together, Gαi1/3 mediate SCF/c-Kit-activated signaling and angiogenesis.
Collapse
Affiliation(s)
- Hua-jian Shan
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Kun Jiang
- Vascular Surgery Department, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, China
| | - Ming-zhi Zhao
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wen-jing Deng
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
| | - Wen-hao Cao
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jia-jun Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Ke-ran Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Chang She
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei-feng Luo
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
| | - Jin Yao
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao-zhong Zhou
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Dan Zhang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cong Cao
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
12
|
Hysenaj L, Little S, Kulhanek K, Magnen M, Bahl K, Gbenedio OM, Prinz M, Rodriguez L, Andersen C, Rao AA, Shen A, Lone JC, Lupin-Jimenez LC, Bonser LR, Serwas NK, Mick E, Khalid MM, Taha TY, Kumar R, Li JZ, Ding VW, Matsumoto S, Maishan M, Sreekumar B, Simoneau C, Nazarenko I, Tomlinson MG, Khan K, von Gottberg A, Sigal A, Looney MR, Fragiadakis GK, Jablons DM, Langelier CR, Matthay M, Krummel M, Erle DJ, Combes AJ, Sil A, Ott M, Kratz JR, Roose JP. SARS-CoV-2 infection of airway organoids reveals conserved use of Tetraspanin-8 by Ancestral, Delta, and Omicron variants. Stem Cell Reports 2023; 18:636-653. [PMID: 36827975 PMCID: PMC9948283 DOI: 10.1016/j.stemcr.2023.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/25/2023] Open
Abstract
Ancestral SARS coronavirus-2 (SARS-CoV-2) and variants of concern (VOC) caused a global pandemic with a spectrum of disease severity. The mechanistic explaining variations related to airway epithelium are relatively understudied. Here, we biobanked airway organoids (AO) by preserving stem cell function. We optimized viral infection with H1N1/PR8 and comprehensively characterized epithelial responses to SARS-CoV-2 infection in phenotypically stable AO from 20 different subjects. We discovered Tetraspanin-8 (TSPAN8) as a facilitator of SARS-CoV-2 infection. TSPAN8 facilitates SARS-CoV-2 infection rates independently of ACE2-Spike interaction. In head-to-head comparisons with Ancestral SARS-CoV-2, Delta and Omicron VOC displayed lower overall infection rates of AO but triggered changes in epithelial response. All variants shared highest tropism for ciliated and goblet cells. TSPAN8-blocking antibodies diminish SARS-CoV-2 infection and may spur novel avenues for COVID-19 therapy.
Collapse
Affiliation(s)
- Lisiena Hysenaj
- Department of Anatomy, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Samantha Little
- Department of Anatomy, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Kayla Kulhanek
- Department of Anatomy, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Melia Magnen
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kriti Bahl
- Department of Anatomy, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Oghenekevwe M Gbenedio
- Department of Anatomy, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Morgan Prinz
- Department of Anatomy, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Lauren Rodriguez
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA, USA
| | - Christopher Andersen
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA, USA
| | - Arjun Arkal Rao
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alan Shen
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Leonard C Lupin-Jimenez
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA, USA
| | - Luke R Bonser
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Nina K Serwas
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eran Mick
- Division of Infectious Diseases, University of California, San Francisco, San Francisco, CA, USA; Division of Pulmonary and Critical Care, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Mir M Khalid
- Gladstone Institute of Virology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Taha Y Taha
- Gladstone Institute of Virology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Renuka Kumar
- Gladstone Institute of Virology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Jack Z Li
- Department of Surgery, Division of Cardiothoracic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Vivianne W Ding
- Department of Surgery, Division of Cardiothoracic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Shotaro Matsumoto
- Cardiovascular Research Institute, Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mazharul Maishan
- Cardiovascular Research Institute, Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bharath Sreekumar
- Gladstone Institute of Virology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Camille Simoneau
- Gladstone Institute of Virology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Irina Nazarenko
- Institute for Infection Prevention and Hospital Epidemiology, University of Freiburg, Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany
| | - Michael G Tomlinson
- School of Biosciences, University of Birmingham, Birmingham, UK; Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Midlands, UK
| | - Khajida Khan
- Africa Health Research Institute, Durban, South Africa; School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Anne von Gottberg
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa; SAMRC Antibody Immunity Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Alex Sigal
- Africa Health Research Institute, Durban, South Africa; School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa; Max Planck Institute for Infection Biology, Berlin, Germany; Centre for the AIDS Program of Research, Durban, South Africa
| | - Mark R Looney
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Division of Pulmonary and Critical Care, San Francisco, San Francisco, CA, USA
| | - Gabriela K Fragiadakis
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David M Jablons
- Division of Pulmonary and Critical Care, San Francisco, San Francisco, CA, USA; Department of Surgery, Division of Cardiothoracic Surgery, University of California, San Francisco, San Francisco, CA, USA; Cardiovascular Research Institute, Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Charles R Langelier
- Division of Infectious Diseases, University of California, San Francisco, San Francisco, CA, USA; Division of Pulmonary and Critical Care, San Francisco, San Francisco, CA, USA; Gladstone Institute of Virology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Michael Matthay
- Division of Pulmonary and Critical Care, San Francisco, San Francisco, CA, USA; Cardiovascular Research Institute, Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew Krummel
- Department of Anatomy, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David J Erle
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA, USA; Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Division of Pulmonary and Critical Care, San Francisco, San Francisco, CA, USA
| | - Alexis J Combes
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Anita Sil
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Melanie Ott
- Gladstone Institute of Virology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, CA 94143, USA; Quantitative Biosciences Institute COVID-19 Research Group, University of California, San Francisco, San Francisco, CA, USA
| | - Johannes R Kratz
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA, USA; Department of Surgery, Division of Cardiothoracic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Jeroen P Roose
- Department of Anatomy, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA.
| |
Collapse
|
13
|
Wakabayashi T, Naito H. Cellular heterogeneity and stem cells of vascular endothelial cells in blood vessel formation and homeostasis: Insights from single-cell RNA sequencing. Front Cell Dev Biol 2023; 11:1146399. [PMID: 37025170 PMCID: PMC10070846 DOI: 10.3389/fcell.2023.1146399] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/06/2023] [Indexed: 04/08/2023] Open
Abstract
Vascular endothelial cells (ECs) that constitute the inner surface of blood vessels are essential for new vessel formation and organ homeostasis. ECs display remarkable phenotypic heterogeneity across different organs and the vascular tree during angiogenesis and homeostasis. Recent advances in single cell RNA sequencing (scRNA-seq) technologies have allowed a new understanding of EC heterogeneity in both mice and humans. In particular, scRNA-seq has identified new molecular signatures for arterial, venous and capillary ECs in different organs, as well as previously unrecognized specialized EC subtypes, such as the aerocytes localized in the alveolar capillaries of the lung. scRNA-seq has also revealed the gene expression profiles of specialized tissue-resident EC subtypes that are capable of clonal expansion and contribute to adult angiogenesis, a process of new vessel formation from the pre-existing vasculature. These specialized tissue-resident ECs have been identified in various different mouse tissues, including aortic endothelium, liver, heart, lung, skin, skeletal muscle, retina, choroid, and brain. Transcription factors and signaling pathways have also been identified in the specialized tissue-resident ECs that control angiogenesis. Furthermore, scRNA-seq has also documented responses of ECs in diseases such as cancer, age-related macular degeneration, Alzheimer's disease, atherosclerosis, and myocardial infarction. These new findings revealed by scRNA-seq have the potential to provide new therapeutic targets for different diseases associated with blood vessels. In this article, we summarize recent advances in the understanding of the vascular endothelial cell heterogeneity and endothelial stem cells associated with angiogenesis and homeostasis in mice and humans, and we discuss future prospects for the application of scRNA-seq technology.
Collapse
Affiliation(s)
- Taku Wakabayashi
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
- Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, United States
- *Correspondence: Taku Wakabayashi, ; Hisamichi Naito,
| | - Hisamichi Naito
- Department of Vascular Physiology, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa, Japan
- *Correspondence: Taku Wakabayashi, ; Hisamichi Naito,
| |
Collapse
|
14
|
Sun H, Morihara R, Feng T, Bian Z, Yu H, Hu X, Hu X, Bian Y, Sasaki R, Fukui Y, Takemoto M, Yunoki T, Nakano Y, Abe K, Yamashita T. Human Cord Blood-Endothelial Progenitor Cells Alleviate Intimal Hyperplasia of Arterial Damage in a Rat Stroke Model. Cell Transplant 2023; 32:9636897231193069. [PMID: 37615293 PMCID: PMC10467372 DOI: 10.1177/09636897231193069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 06/28/2023] [Accepted: 07/18/2023] [Indexed: 08/25/2023] Open
Abstract
Human cord blood-endothelial progenitor cells (hCB-EPCs) isolated from the human umbilical cord can be used to repair damaged arteries. In this study, we used an animal model with pathological changes that mimics artery wall damage caused by stent retrievers in humans. We injected hCB-EPCs to investigate their effect on endothelial hyperplasia and dysfunction during intimal repair. Four groups were established based on the length of reperfusion (3 and 28 days), as well as the presence or absence of hCB-EPC therapy. Damage to the internal carotid artery was evaluated by hematoxylin-eosin and immunohistochemical staining. Stroke volume was not significantly different between non-EPC and EPC groups although EPC treatment alleviated intimal hyperplasia 28 days after intimal damage. Vascular endothelial growth factor (VEGF) and eNOS expression were significantly higher in the EPC-treated group than in the non-EPC group 3 days after intimal damage. In addition, MMP9 and 4HNE expression in the EPC-treated group was significantly lower than in the non-EPC group. Ultimately, this study found that venous transplantation of hCB-EPCs could inhibit neointimal hyperplasia, alleviate endothelial dysfunction, suppress intimal inflammation, and reduce oxidative stress during healing of intimal damage.
Collapse
Affiliation(s)
- Hongming Sun
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ryuta Morihara
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tian Feng
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Zhihong Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Haibo Yu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Xiao Hu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Xinran Hu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yuting Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ryo Sasaki
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yusuke Fukui
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Mami Takemoto
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Taijun Yunoki
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yumiko Nakano
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Koji Abe
- National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
15
|
Yu QC, Geng A, Preusch CB, Chen Y, Peng G, Xu Y, Jia Y, Miao Y, Xue H, Gao D, Bao L, Pan W, Chen J, Garcia KC, Cheung TH, Zeng YA. Activation of Wnt/β-catenin signaling by Zeb1 in endothelial progenitors induces vascular quiescence entry. Cell Rep 2022; 41:111694. [DOI: 10.1016/j.celrep.2022.111694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 08/05/2022] [Accepted: 10/31/2022] [Indexed: 11/23/2022] Open
|
16
|
Hashemi G, Dight J, Khosrotehrani K, Sormani L. Melanoma Tumour Vascularization and Tissue-Resident Endothelial Progenitor Cells. Cancers (Basel) 2022; 14:4216. [PMID: 36077754 PMCID: PMC9454996 DOI: 10.3390/cancers14174216] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/20/2022] Open
Abstract
The aggressiveness of solid cancers, such as melanoma, relies on their metastatic potential. It has become evident that this key cause of mortality is largely conferred by the tumour-associated stromal cells, especially endothelial cells. In addition to their essential role in the formation of the tumour vasculature, endothelial cells significantly contribute to the establishment of the tumour microenvironment, thus enabling the dissemination of cancer cells. Melanoma tumour vascularization occurs through diverse biological processes. Vasculogenesis is the formation of de novo blood vessels from endothelial progenitor cells (EPCs), and recent research has shown the role of EPCs in melanoma tumour vascularization. A more detailed understanding of the complex role of EPCs and how they contribute to the abnormal vessel structures in tumours is of importance. Moreover, anti-angiogenic drugs have a limited effect on melanoma tumour vascularization, and the role of these drugs on EPCs remains to be clarified. Overall, targeting cancer vasculature remains a challenge, and the role of anti-angiogenic drugs and combination therapies in melanoma, a focus of this review, is an area of extensive exploration.
Collapse
Affiliation(s)
| | | | - Kiarash Khosrotehrani
- Experimental Dermatology Group, Dermatology Research Centre, The UQ Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Laura Sormani
- Experimental Dermatology Group, Dermatology Research Centre, The UQ Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
17
|
Moradi SZ, Jalili F, Hoseinkhani Z, Mansouri K. Regenerative Medicine and Angiogenesis; Focused on Cardiovascular Disease. Adv Pharm Bull 2022; 12:686-699. [PMID: 36415645 PMCID: PMC9675929 DOI: 10.34172/apb.2022.072] [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: 07/12/2020] [Revised: 03/26/2021] [Accepted: 09/27/2021] [Indexed: 10/11/2023] Open
Abstract
Cardiovascular disease (CVD) is a major concern for health with high mortality rates around the world. CVD is often associated with partial or full occlusion of the blood vessel network. Changes in lifestyle can be useful for management early-stage disease but in the advanced stage, surgical interventions or pharmacological are needed to increase the blood flow through the affected tissue or to reduce the energy requirements. Regeneration medicine is a new science that has provided many different options for treating various diseases, especially in CVD over the years. Stem cell therapy, gene therapy, and tissue engineering are some of the powerful branches of the field that have given patients great hope in improving their condition. In this review, we attempted to examine the beneficial effects, challenges, and contradictory effects of angiogenesis in vivo, and in vitro models' studies of CVD. We hope that this information will be able to help other researchers to design new effective structures and open new avenues for the treatment of CVD with the help of angiogenesis and regeneration medicine in the future.
Collapse
Affiliation(s)
- Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Faramarz Jalili
- Gradute Studies Student, Sobey School of Business, Saint Mary‚S University, Halifax, NS,Canada
| | - Zohreh Hoseinkhani
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Molecular Medicine Department, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
18
|
Tacconi C, Plein A, Colletto C, Villa E, Denti L, Barone C, Javanmardi Y, Moeendarbary E, Azzoni E, Fantin A, Ruhrberg C. KIT is dispensable for physiological organ vascularisation in the embryo. Angiogenesis 2022; 25:343-353. [PMID: 35416527 PMCID: PMC9249691 DOI: 10.1007/s10456-022-09837-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 03/25/2022] [Indexed: 11/08/2022]
Abstract
Blood vessels form vast networks in all vertebrate organs to sustain tissue growth, repair and homeostatic metabolism, but they also contribute to a range of diseases with neovascularisation. It is, therefore, important to define the molecular mechanisms that underpin blood vessel growth. The receptor tyrosine kinase KIT is required for the normal expansion of hematopoietic progenitors that arise during embryogenesis from hemogenic endothelium in the yolk sac and dorsal aorta. Additionally, KIT has been reported to be expressed in endothelial cells during embryonic brain vascularisation and has been implicated in pathological angiogenesis. However, it is neither known whether KIT expression is widespread in normal organ endothelium nor whether it promotes blood vessel growth in developing organs. Here, we have used single-cell analyses to show that KIT is expressed in endothelial cell subsets of several organs, both in the adult and in the developing embryo. Knockout mouse analyses revealed that KIT is dispensable for vascularisation of growing organs in the midgestation embryo, including the lung, liver and brain. By contrast, vascular changes emerged during late-stage embryogenesis in these organs from KIT-deficient embryos, concurrent with severe erythrocyte deficiency and growth retardation. These findings suggest that KIT is not required for developmental tissue vascularisation in physiological conditions, but that KIT deficiency causes foetal anaemia at late gestation and thereby pathological vascular remodelling.
Collapse
Affiliation(s)
- Carlotta Tacconi
- Department of Biosciences, University of Milan, Via G. Celoria 26, 20133, Milan, Italy
| | - Alice Plein
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Chiara Colletto
- Department of Biosciences, University of Milan, Via G. Celoria 26, 20133, Milan, Italy
| | - Emanuela Villa
- Department of Biosciences, University of Milan, Via G. Celoria 26, 20133, Milan, Italy
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Laura Denti
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Cristiana Barone
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Yousef Javanmardi
- UCL Department of Mechanical Engineering, University College London, London, UK
| | - Emad Moeendarbary
- UCL Department of Mechanical Engineering, University College London, London, UK
| | - Emanuele Azzoni
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Alessandro Fantin
- Department of Biosciences, University of Milan, Via G. Celoria 26, 20133, Milan, Italy.
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| |
Collapse
|
19
|
Tao J, Cao X, Yu B, Qu A. Vascular Stem/Progenitor Cells in Vessel Injury and Repair. Front Cardiovasc Med 2022; 9:845070. [PMID: 35224067 PMCID: PMC8866648 DOI: 10.3389/fcvm.2022.845070] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular repair upon vessel injury is essential for the maintenance of arterial homeostasis and function. Stem/progenitor cells were demonstrated to play a crucial role in regeneration and replenishment of damaged vascular cells during vascular repair. Previous studies revealed that myeloid stem/progenitor cells were the main sources of tissue regeneration after vascular injury. However, accumulating evidences from developing lineage tracing studies indicate that various populations of vessel-resident stem/progenitor cells play specific roles in different process of vessel injury and repair. In response to shear stress, inflammation, or other risk factors-induced vascular injury, these vascular stem/progenitor cells can be activated and consequently differentiate into different types of vascular wall cells to participate in vascular repair. In this review, mechanisms that contribute to stem/progenitor cell differentiation and vascular repair are described. Targeting these mechanisms has potential to improve outcome of diseases that are characterized by vascular injury, such as atherosclerosis, hypertension, restenosis, and aortic aneurysm/dissection. Future studies on potential stem cell-based therapy are also highlighted.
Collapse
Affiliation(s)
- Jiaping Tao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- The Key Laboratory of Cardiovascular Remodeling-Related Diseases, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Xuejie Cao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- The Key Laboratory of Cardiovascular Remodeling-Related Diseases, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Baoqi Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- The Key Laboratory of Cardiovascular Remodeling-Related Diseases, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
- *Correspondence: Baoqi Yu
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- The Key Laboratory of Cardiovascular Remodeling-Related Diseases, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
- Aijuan Qu
| |
Collapse
|
20
|
Saito N, Shirado T, Funabashi-Eto H, Wu Y, Mori M, Asahi R, Yoshimura K. Purification and characterization of human adipose-resident microvascular endothelial progenitor cells. Sci Rep 2022; 12:1775. [PMID: 35110646 PMCID: PMC8811023 DOI: 10.1038/s41598-022-05760-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/20/2021] [Indexed: 12/18/2022] Open
Abstract
Human adipose tissue is a rich source of adipose-derived stem cells (ASCs) and vascular endothelial progenitor cells (EPCs). However, no standardized method has been established for the isolation and purification of adipose-derived EPCs (AEPCs). The aim of this study was to establish a method for the isolation and purification of AEPCs. The stromal vascular fraction (SVF) was extracted from human lipoaspirates, and the CD45−CD31+ fraction of the SVF was collected by magnetic-activated cell sorting (MACS). The CD45−CD31+ fraction was cultured for 4.5 days, followed by a second MACS separation to collect the CD31+ fraction. Purified AEPCs were expanded without being overwhelmed by proliferating ASCs, indicating that a high level (> 95%) of AEPC purification is a key factor for their successful isolation and expansion. AEPCs exhibited typical endothelial markers, including CD31, von Willebrand factor, and the isolectin-B4 binding capacity. AEPCs formed colonies, comparable to cultured human umbilical vein endothelial cells (HUVECs). Both AEPCs and HUVECs formed capillary-like networks in the tube formation assay, with no significant difference in network lengths. We are the first to establish a purification and expansion method to isolate these cells. Because adipose tissue is a clinically accessible and abundant tissue, AEPCs may have potential advantages as a therapeutic tool for regenerative medicine.
Collapse
Affiliation(s)
- Natsumi Saito
- Department of Plastic Surgery, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Takako Shirado
- Department of Plastic Surgery, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hitomi Funabashi-Eto
- Department of Plastic Surgery, Federation of National Public Service Personnel Mutual Aid Associations, Hamanomachi Hospital, 3-3-1, Nagahama, Chuou-ku, Fukuoka, 810-8539, Japan
| | - Yunyan Wu
- Department of Plastic Surgery, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Masanori Mori
- Department of Plastic Surgery, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Rintaro Asahi
- Department of Plastic Surgery, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Kotaro Yoshimura
- Department of Plastic Surgery, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| |
Collapse
|
21
|
Arpino JM, Yin H, Prescott EK, Staples SCR, Nong Z, Li F, Chevalier J, Balint B, O’Neil C, Mortuza R, Milkovich S, Lee JJ, Lorusso D, Sandig M, Hamilton DW, Holdsworth DW, Poepping TL, Ellis CG, Pickering JG. Low-flow intussusception and metastable VEGFR2 signaling launch angiogenesis in ischemic muscle. SCIENCE ADVANCES 2021; 7:eabg9509. [PMID: 34826235 PMCID: PMC8626079 DOI: 10.1126/sciadv.abg9509] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Efforts to promote sprouting angiogenesis in skeletal muscles of individuals with peripheral artery disease have not been clinically successful. We discovered that, contrary to the prevailing view, angiogenesis following ischemic muscle injury in mice was not driven by endothelial sprouting. Instead, real-time imaging revealed the emergence of wide-caliber, primordial conduits with ultralow flow that rapidly transformed into a hierarchical neocirculation by transluminal bridging and intussusception. This process was accelerated by inhibiting vascular endothelial growth factor receptor-2 (VEGFR2). We probed this response by developing the first live-cell model of transluminal endothelial bridging using microfluidics. Endothelial cells subjected to ultralow shear stress could reposition inside the flowing lumen as pillars. Moreover, the low-flow lumen proved to be a privileged location for endothelial cells with reduced VEGFR2 signaling capacity, as VEGFR2 mechanosignals were boosted. These findings redefine regenerative angiogenesis in muscle as an intussusceptive process and uncover a basis for its launch.
Collapse
Affiliation(s)
- John-Michael Arpino
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Hao Yin
- Robarts Research Institute, Western University, London, Canada
| | - Emma K. Prescott
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Sabrina C. R. Staples
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Zengxuan Nong
- Robarts Research Institute, Western University, London, Canada
| | - Fuyan Li
- Robarts Research Institute, Western University, London, Canada
| | - Jacqueline Chevalier
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Brittany Balint
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Caroline O’Neil
- Robarts Research Institute, Western University, London, Canada
| | | | - Stephanie Milkovich
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Jason J. Lee
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
- Department of Medicine, Western University, London, Canada
| | - Daniel Lorusso
- Robarts Research Institute, Western University, London, Canada
| | - Martin Sandig
- Department of Anatomy and Cell Biology, Western University, London, Canada
| | | | - David W. Holdsworth
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Tamie L. Poepping
- Department of Physics and Astronomy, Western University, London, Canada
| | - Christopher G. Ellis
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
- Department of Medicine, Western University, London, Canada
| | - J. Geoffrey Pickering
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
- Department of Medicine, Western University, London, Canada
- Department of Biochemistry, Western University, London, Canada
- Corresponding author.
| |
Collapse
|
22
|
Chambers SE, Pathak V, Pedrini E, Soret L, Gendron N, Guerin CL, Stitt AW, Smadja DM, Medina RJ. Current concepts on endothelial stem cells definition, location, and markers. Stem Cells Transl Med 2021; 10 Suppl 2:S54-S61. [PMID: 34724714 PMCID: PMC8560200 DOI: 10.1002/sctm.21-0022] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/12/2021] [Accepted: 03/25/2021] [Indexed: 12/20/2022] Open
Abstract
Ischemic vascular disease is a major cause of mortality and morbidity worldwide, and regeneration of blood vessels in perfusion-deficient tissues is a worthwhile therapeutic goal. The idea of delivering endothelial stem/progenitor cells to repair damaged vasculature, reperfuse hypoxic tissue, prevent cell death, and consequently diminish tissue inflammation and fibrosis has a strong scientific basis and clinical value. Various labs have proposed endothelial stem/progenitor cell candidates. This has created confusion, as there are profound differences between these cell definitions based on isolation methodology, characterization, and reparative biology. Here, a stricter definition based on stem cell biology principles is proposed. Although preclinical studies have often been promising, results from clinical trials have been highly contradictory and served to highlight multiple challenges associated with disappointing therapeutic benefit. This article reviews recent accomplishments in the field and discusses current difficulties when developing endothelial stem cell therapies. Emerging evidence that disputes the classic view of the bone marrow as the source for these cells and supports the vascular wall as the niche for these tissue-resident endothelial stem cells is considered. In addition, novel markers to identify endothelial stem cells, including CD157, EPCR, and CD31low VEGFR2low IL33+ Sox9+ , are described.
Collapse
Affiliation(s)
- Sarah E.J. Chambers
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - Varun Pathak
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - Edoardo Pedrini
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - Lou Soret
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Hematology department and Biosurgical research lab (Carpentier Foundation)Assistance Publique Hôpitaux de Paris.Centre‐Université de Paris (APHP‐CUP)ParisFrance
| | - Nicolas Gendron
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Hematology department and Biosurgical research lab (Carpentier Foundation)Assistance Publique Hôpitaux de Paris.Centre‐Université de Paris (APHP‐CUP)ParisFrance
| | - Coralie L. Guerin
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Cytometry Platform, Institut CurieParisFrance
| | - Alan W. Stitt
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - David M. Smadja
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Hematology department and Biosurgical research lab (Carpentier Foundation)Assistance Publique Hôpitaux de Paris.Centre‐Université de Paris (APHP‐CUP)ParisFrance
| | - Reinhold J. Medina
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| |
Collapse
|
23
|
Wang X, Wang R, Jiang L, Xu Q, Guo X. Endothelial repair by stem and progenitor cells. J Mol Cell Cardiol 2021; 163:133-146. [PMID: 34743936 DOI: 10.1016/j.yjmcc.2021.10.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 12/19/2022]
Abstract
The integrity of the endothelial barrier is required to maintain vascular homeostasis and fluid balance between the circulatory system and surrounding tissues and to prevent the development of vascular disease. However, the origin of the newly developed endothelial cells is still controversial. Stem and progenitor cells have the potential to differentiate into endothelial cell lines and stimulate vascular regeneration in a paracrine/autocrine fashion. The one source of new endothelial cells was believed to come from the bone marrow, which was challenged by the recent findings. By administration of new techniques, including genetic cell lineage tracing and single cell RNA sequencing, more solid data were obtained that support the concept of stem/progenitor cells for regenerating damaged endothelium. Specifically, it was found that tissue resident endothelial progenitors located in the vessel wall were crucial for endothelial repair. In this review, we summarized the latest advances in stem and progenitor cell research in endothelial regeneration through findings from animal models and discussed clinical data to indicate the future direction of stem cell therapy.
Collapse
Affiliation(s)
- Xuyang Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruilin Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liujun Jiang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
24
|
Dight J, Zhao J, Styke C, Khosrotehrani K, Patel J. Resident vascular endothelial progenitor definition and function: the age of reckoning. Angiogenesis 2021; 25:15-33. [PMID: 34499264 PMCID: PMC8813834 DOI: 10.1007/s10456-021-09817-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/05/2021] [Indexed: 02/07/2023]
Abstract
The cardiovascular system is composed around the central function of the endothelium that lines the inner surfaces of its vessels. In recent years, the existence of a progenitor population within the endothelium has been validated through the study of endothelial colony-forming cells (ECFCs) in human peripheral blood and certain vascular beds. However, our knowledge on endothelial populations in vivo that can give rise to ECFCs in culture has been limited. In this review we report and analyse recent attempts at describing progenitor populations in vivo from murine studies that reflect the self-renewal and stemness capacity observed in ECFCs. We pinpoint seminal discoveries within the field, which have phenotypically defined, and functionally scrutinised these endothelial progenitors. Furthermore, we review recent publications utilising single-cell sequencing technologies to better understand the endothelium in homeostasis and pathology.
Collapse
Affiliation(s)
- James Dight
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
| | - Jilai Zhao
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
| | - Cassandra Styke
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
| | - Kiarash Khosrotehrani
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia.
| | - Jatin Patel
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia. .,Cancer and Ageing Research Program, School of Biomedical Sciences, Queensland University of Technology, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia.
| |
Collapse
|
25
|
Jiang L, Chen T, Sun S, Wang R, Deng J, Lyu L, Wu H, Yang M, Pu X, Du L, Chen Q, Hu Y, Hu X, Zhou Y, Xu Q, Zhang L. Nonbone Marrow CD34 + Cells Are Crucial for Endothelial Repair of Injured Artery. Circ Res 2021; 129:e146-e165. [PMID: 34474592 DOI: 10.1161/circresaha.121.319494] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Liujun Jiang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Ting Chen
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu).,Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, Zhejiang Province, China (T. Chen)
| | - Shasha Sun
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu).,Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China. (S. Sun, M. Yang, Q. Chen, L. Zhang)
| | - Ruilin Wang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Jiacheng Deng
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Lingxia Lyu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Hong Wu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Mei Yang
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China. (S. Sun, M. Yang, Q. Chen, L. Zhang)
| | - Xiangyuan Pu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Luping Du
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Qishan Chen
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China. (S. Sun, M. Yang, Q. Chen, L. Zhang)
| | - Yanhua Hu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Xiaosheng Hu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Yijiang Zhou
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu).,Centre for Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (Q. Xu)
| | - Li Zhang
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China. (S. Sun, M. Yang, Q. Chen, L. Zhang)
| |
Collapse
|
26
|
Wang G, Wen B, Ren X, Li E, Zhang Y, Guo M, Xu Y, Whitsett JA, Kalin TV, Kalinichenko VV. Generation of Pulmonary Endothelial Progenitor Cells for Cell-based Therapy Using Interspecies Mouse-Rat Chimeras. Am J Respir Crit Care Med 2021; 204:326-338. [PMID: 33705684 DOI: 10.1164/rccm.202003-0758oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale: Although pulmonary endothelial progenitor cells (EPCs) hold promise for cell-based therapies for neonatal pulmonary disorders, whether EPCs can be derived from pluripotent embryonic stem cells (ESCs) or induced pluripotent stem cells remains unknown.Objectives: To investigate the heterogeneity of pulmonary EPCs and derive functional EPCs from pluripotent ESCs.Methods: Single-cell RNA sequencing of neonatal human and mouse lung was used to identify the heterogeneity of pulmonary EPCs. CRISPR/Cas9 gene editing was used to genetically label and purify mouse pulmonary EPCs. Functional properties of the EPCs were assessed after cell transplantation into neonatal mice with S52F Foxf1 mutation, a mouse model of alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV). Interspecies mouse-rat chimeras were produced through blastocyst complementation to generate EPCs from pluripotent ESCs for cell therapy in ACDMPV mice.Measurements and Main Results: We identified a unique population of EPCs, FOXF1+cKIT+ EPCs, as a subset of recently described general capillary cells (gCAPs) expressing SMAD7, ZBTB20, NFIA, and DLL4 but lacking mature arterial, venous, and lymphatic markers. FOXF1+cKIT+ gCAPs are reduced in ACDMPV, and their transcriptomic signature is conserved in mouse and human lungs. After cell transplantation into the neonatal circulation of ACDMPV mice, FOXF1+cKIT+ gCAPs engraft into the pulmonary vasculature, stimulate angiogenesis, improve oxygenation, and prevent alveolar simplification. FOXF1+cKIT+ gCAPs, produced from ESCs in interspecies chimeras, are fully competent to stimulate neonatal lung angiogenesis and alveolarization in ACDMPV mice.Conclusions: Cell-based therapy using donor or ESC/induced pluripotent stem cell-derived FOXF1+cKIT+ endothelial progenitors may be considered for treatment of human ACDMPV.
Collapse
Affiliation(s)
| | | | | | - Enhong Li
- Center for Lung Regenerative Medicine
| | | | | | - Yan Xu
- Division of Pulmonary Biology, and
| | | | | | - Vladimir V Kalinichenko
- Center for Lung Regenerative Medicine.,Division of Pulmonary Biology, and.,Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, Ohio
| |
Collapse
|
27
|
Kolesnichenko OA, Whitsett JA, Kalin TV, Kalinichenko VV. Therapeutic Potential of Endothelial Progenitor Cells in Pulmonary Diseases. Am J Respir Cell Mol Biol 2021; 65:473-488. [PMID: 34293272 DOI: 10.1165/rcmb.2021-0152tr] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Compromised alveolar development and pulmonary vascular remodeling are hallmarks of pediatric lung diseases such as bronchopulmonary dysplasia (BPD) and alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV). Although advances in surfactant therapy, corticosteroids, and anti-inflammatory drugs have improved clinical management of preterm infants, still those who suffer with severe vascular complications lack viable treatment options. Paucity of the alveolar capillary network in ACDMPV causes respiratory distress and leads to mortality in a vast majority of ACDMPV infants. The discovery of endothelial progenitor cells (EPCs) in 1997 brought forth the paradigm of postnatal vasculogenesis and hope for promoting vascularization in fragile patient populations, such as those with BPD and ACDMPV. The identification of diverse EPC populations, both hematopoietic and nonhematopoietic in origin, provided a need to identify progenitor cell selective markers which are linked to progenitor properties needed to develop cell-based therapies. Focusing to the future potential of EPCs for regenerative medicine, this review will discuss various aspects of EPC biology, beginning with the identification of hematopoietic, nonhematopoietic, and tissue-resident EPC populations. We will review knowledge related to cell surface markers, signature gene expression, key transcriptional regulators, and will explore the translational potential of EPCs for cell-based therapy for BPD and ACDMPV. The ability to produce pulmonary EPCs from patient-derived induced pluripotent stem cells (iPSCs) in vitro, holds promise for restoring vascular growth and function in the lungs of patients with pediatric pulmonary disorders.
Collapse
Affiliation(s)
- Olena A Kolesnichenko
- Cincinnati Children's Hospital Medical Center, 2518, Cincinnati, Ohio, United States
| | - Jeffrey A Whitsett
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Tanya V Kalin
- Cincinnati Children\'s Hospital Medical Center, 2518, Pediatrics, Cincinnati, Ohio, United States
| | - Vladimir V Kalinichenko
- Cincinnati Children's Hospital Medical Center, Pediatrics, Division of Pulmonary Biology, Cincinnati, Ohio, United States;
| |
Collapse
|
28
|
Jiang L, Sun X, Deng J, Hu Y, Xu Q. Different Roles of Stem/Progenitor Cells in Vascular Remodeling. Antioxid Redox Signal 2021; 35:192-203. [PMID: 33107320 DOI: 10.1089/ars.2020.8199] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Significance: Since the discovery of vascular stem cells, there has been considerable advancement in comprehending the nature and functions of these cells. Due to their differentiation potential to repair endothelial cells and to participate in lesion formation during vascular remodeling, it is crucial to elucidate vascular stem cell behaviors and the mechanisms underlying this process, which could provide new chances for the design of clinical therapeutic application of stem cells. Recent Advances: Over the past decades, major progress has been made on progenitor/vascular stem cells in the field of cardiovascular research. Vascular stem cells are mostly latent in their niches and can be bioactivated in response to damage and get involved in endothelial repair and smooth muscle cell aggregation to generate neointima. Accumulating evidence has been shown recently, using genetic lineage tracing mouse models, to particularly provide solutions to the nature of vascular stem cells and to monitor both cell migration and the process of differentiation during physiological angiogenesis and in vascular diseases. Critical Issues: This article reviews and summarizes the current research progress of vascular stem cells in this field and highlights future prospects for stem cell research in regenerative medicine. Future Directions: Despite recent advances and achievements of stem cells in cardiovascular research, the nature and cell fate of vascular stem cells remain elusive. Further comprehensive studies using new techniques including genetic cell lineage tracing and single-cell RNA sequencing are essential to fully illuminate the role of stem cells in vascular development and diseases. Antioxid. Redox Signal. 35, 192-203.
Collapse
Affiliation(s)
- Liujun Jiang
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaolei Sun
- Vascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiacheng Deng
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanhua Hu
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
29
|
Hysenaj L, Little S, Kulhanek K, Gbenedio OM, Rodriguez L, Shen A, Lone JC, Lupin-Jimenez LC, Bonser LR, Serwas NK, Bahl K, Mick E, Li JZ, Ding VW, Matsumoto S, Maishan M, Simoneau C, Fragiadakis G, Jablons DM, Langelier CR, Matthay M, Ott M, Krummel M, Combes AJ, Sil A, Erle DJ, Kratz JR, Roose JP. SARS-CoV-2 infection studies in lung organoids identify TSPAN8 as novel mediator. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.06.01.446640. [PMID: 34100012 PMCID: PMC8183007 DOI: 10.1101/2021.06.01.446640] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SARS coronavirus-2 (SARS-CoV-2) is causing a global pandemic with large variation in COVID-19 disease spectrum. SARS-CoV-2 infection requires host receptor ACE2 on lung epithelium, but epithelial underpinnings of variation are largely unknown. We capitalized on comprehensive organoid assays to report remarkable variation in SARS-CoV-2 infection rates of lung organoids from different subjects. Tropism is highest for TUBA- and MUC5AC-positive organoid cells, but levels of TUBA-, MUC5A-, or ACE2- positive cells do not predict infection rate. We identify surface molecule Tetraspanin 8 (TSPAN8) as novel mediator of SARS-CoV-2 infection, which is not downregulated by this specific virus. TSPAN8 levels, prior to infection, strongly correlate with infection rate and TSPAN8-blocking antibodies diminish SARS-CoV-2 infection. We propose TSPAN8 as novel functional biomarker and potential therapeutic target for COVID-19.
Collapse
Affiliation(s)
- Lisiena Hysenaj
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| | - Samantha Little
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| | - Kayla Kulhanek
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| | - Oghenekevwe M. Gbenedio
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| | - Lauren Rodriguez
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California 94143, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, California 94143, USA
| | - Alan Shen
- UCSF CoLabs, University of California San Francisco, San Francisco, California 94143, USA
| | - Jean-Christophe Lone
- School of Life Science, University of Essex, Wivenhoe Park,Colchester C04 3SQ, United Kingdom
| | | | - Luke R. Bonser
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California 94143, USA
| | - Nina K. Serwas
- Department of Pathology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Kriti Bahl
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| | - Eran Mick
- Division of Infectious Diseases, University of California, San Francisco, San Francisco, California 94143, USA and Department of Surgery, Division of Cardiothoracic Surgery, University of California, San Francisco, San Francisco, California 94143, USA
| | - Jack Z. Li
- Cardiovascular Research Institute, Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, California 94143, USA
| | - Vivianne W. Ding
- Cardiovascular Research Institute, Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, California 94143, USA
| | - Shotaro Matsumoto
- Gladstone Institute of Virology, Department of Medicine, University of California San Francisco, California 94143, USA
| | - Mazharul Maishan
- Gladstone Institute of Virology, Department of Medicine, University of California San Francisco, California 94143, USA
| | - Camille Simoneau
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Gabriela Fragiadakis
- UCSF CoLabs, University of California San Francisco, San Francisco, California 94143, USA
| | - David M. Jablons
- Cardiovascular Research Institute, Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, California 94143, USA
| | - Charles R. Langelier
- Division of Infectious Diseases, University of California, San Francisco, San Francisco, California 94143, USA and Department of Surgery, Division of Cardiothoracic Surgery, University of California, San Francisco, San Francisco, California 94143, USA
- Chan Zuckerberg Biohub, San Francisco, California 94158, USA
| | - Michael Matthay
- Gladstone Institute of Virology, Department of Medicine, University of California San Francisco, California 94143, USA
| | - Melanie Ott
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Matthew Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Alexis J. Combes
- UCSF CoLabs, University of California San Francisco, San Francisco, California 94143, USA
| | - Anita Sil
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California 94143, USA
| | - David J. Erle
- UCSF CoLabs, University of California San Francisco, San Francisco, California 94143, USA
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California 94143, USA
| | - Johannes R. Kratz
- Cardiovascular Research Institute, Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, California 94143, USA
| | - Jeroen P. Roose
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
30
|
Aquino JB, Sierra R, Montaldo LA. Diverse cellular origins of adult blood vascular endothelial cells. Dev Biol 2021; 477:117-132. [PMID: 34048734 DOI: 10.1016/j.ydbio.2021.05.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/26/2021] [Accepted: 05/14/2021] [Indexed: 12/11/2022]
Abstract
During embryonic stages, vascular endothelial cells (ECs) originate from the mesoderm, at specific extraembryonic and embryonic regions, through a process called vasculogenesis. In the adult, EC renewal/replacement mostly depend on local resident ECs or endothelial progenitor cells (EPCs). Nevertheless, contribution from circulating ECs/EPCs was also reported. In addition, cells lacking from EC/EPC markers with in vitro extended plasticity were shown to originate endothelial-like cells (ELCs). Most of these cells consist of mesenchymal stromal progenitors, which would eventually get mobilized from the bone marrow after injury. Based on that, current knowledge on different mouse and human bone marrow stromal cell (BM-SC) subpopulations, able to contribute with mesenchymal stromal/stem cells (MSCs), is herein reviewed. Such analyses underline an unexpected heterogeneity among sinusoidal LepR+ stromal/CAR cells. For instance, in a recent report a subgroup of LepR+ stromal/CAR progenitors, which express GLAST and is traced in Wnt1Cre;R26RTom mice, was found to contribute with ELCs in vivo. These GLAST + Wnt1+ BM-SCs were shown to get mobilized to the peripheral blood and to contribute with liver regeneration. Other sources of ELCs, such as adipose, neural and dental pulp tissues, were also published. Finally, mechanisms likely involved in the enhanced cellular plasticity properties of bone marrow/adipose tissue stromal cells, able to originate ELCs, are assessed. In the future, strategies to analyze the in vivo expression profile of stromal cells, with MSC properties, in combination with screening of active genomic regions at the single cell-level, during early postnatal development and/or after injury, will likely help understanding properties of these ELC sources.
Collapse
Affiliation(s)
- Jorge B Aquino
- CONICET-Universidad Austral, Instituto de Investigaciones en Medicina Traslacional (IIMT), Developmental Biology & Regenerative Medicine Laboratory, Argentina.
| | - Romina Sierra
- CONICET-Universidad Austral, Instituto de Investigaciones en Medicina Traslacional (IIMT), Developmental Biology & Regenerative Medicine Laboratory, Argentina
| | - Laura A Montaldo
- CONICET-Universidad Austral, Instituto de Investigaciones en Medicina Traslacional (IIMT), Developmental Biology & Regenerative Medicine Laboratory, Argentina
| |
Collapse
|
31
|
Jelinkova S, Sleiman Y, Fojtík P, Aimond F, Finan A, Hugon G, Scheuermann V, Beckerová D, Cazorla O, Vincenti M, Amedro P, Richard S, Jaros J, Dvorak P, Lacampagne A, Carnac G, Rotrekl V, Meli AC. Dystrophin Deficiency Causes Progressive Depletion of Cardiovascular Progenitor Cells in the Heart. Int J Mol Sci 2021; 22:ijms22095025. [PMID: 34068508 PMCID: PMC8125982 DOI: 10.3390/ijms22095025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/30/2021] [Accepted: 05/07/2021] [Indexed: 11/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating condition shortening the lifespan of young men. DMD patients suffer from age-related dilated cardiomyopathy (DCM) that leads to heart failure. Several molecular mechanisms leading to cardiomyocyte death in DMD have been described. However, the pathological progression of DMD-associated DCM remains unclear. In skeletal muscle, a dramatic decrease in stem cells, so-called satellite cells, has been shown in DMD patients. Whether similar dysfunction occurs with cardiac muscle cardiovascular progenitor cells (CVPCs) in DMD remains to be explored. We hypothesized that the number of CVPCs decreases in the dystrophin-deficient heart with age and disease state, contributing to DCM progression. We used the dystrophin-deficient mouse model (mdx) to investigate age-dependent CVPC properties. Using quantitative PCR, flow cytometry, speckle tracking echocardiography, and immunofluorescence, we revealed that young mdx mice exhibit elevated CVPCs. We observed a rapid age-related CVPC depletion, coinciding with the progressive onset of cardiac dysfunction. Moreover, mdx CVPCs displayed increased DNA damage, suggesting impaired cardiac muscle homeostasis. Overall, our results identify the early recruitment of CVPCs in dystrophic hearts and their fast depletion with ageing. This latter depletion may participate in the fibrosis development and the acceleration onset of the cardiomyopathy.
Collapse
MESH Headings
- Aging/genetics
- Aging/pathology
- Animals
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Cardiovascular System/metabolism
- Cardiovascular System/pathology
- DNA Damage/genetics
- Disease Models, Animal
- Dystrophin/deficiency
- Dystrophin/genetics
- Gene Expression Regulation/genetics
- Humans
- Mice
- Mice, Inbred mdx/genetics
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Proto-Oncogene Proteins c-kit/genetics
- Stem Cells/metabolism
- Stem Cells/pathology
Collapse
Affiliation(s)
- Sarka Jelinkova
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A3, 62500 Brno, Czech Republic; (S.J.); (P.F.); (D.B.); (P.D.)
- ICRC, St Anne’s University Hospital, Pekařská 53, 65691 Brno, Czech Republic;
| | - Yvonne Sleiman
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Petr Fojtík
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A3, 62500 Brno, Czech Republic; (S.J.); (P.F.); (D.B.); (P.D.)
- ICRC, St Anne’s University Hospital, Pekařská 53, 65691 Brno, Czech Republic;
| | - Franck Aimond
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Amanda Finan
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Gerald Hugon
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Valerie Scheuermann
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Deborah Beckerová
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A3, 62500 Brno, Czech Republic; (S.J.); (P.F.); (D.B.); (P.D.)
- ICRC, St Anne’s University Hospital, Pekařská 53, 65691 Brno, Czech Republic;
| | - Olivier Cazorla
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Marie Vincenti
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
- Pediatric and Adult Congenital Cardiology Department, M3C Regional Reference CHD Center, CHU Montpellier, 371 Avenue du Doyen Giraud, 34295 Montpellier, France
| | - Pascal Amedro
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
- Pediatric and Adult Congenital Cardiology Department, M3C Regional Reference CHD Center, CHU Montpellier, 371 Avenue du Doyen Giraud, 34295 Montpellier, France
| | - Sylvain Richard
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Josef Jaros
- ICRC, St Anne’s University Hospital, Pekařská 53, 65691 Brno, Czech Republic;
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5/A1, 62500 Brno, Czech Republic
| | - Petr Dvorak
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A3, 62500 Brno, Czech Republic; (S.J.); (P.F.); (D.B.); (P.D.)
| | - Alain Lacampagne
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Gilles Carnac
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Vladimir Rotrekl
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A3, 62500 Brno, Czech Republic; (S.J.); (P.F.); (D.B.); (P.D.)
- ICRC, St Anne’s University Hospital, Pekařská 53, 65691 Brno, Czech Republic;
- Correspondence: (V.R.); (A.C.M.); Tel.: +420-549-498-002 (V.R.); +33-4-67-41-52-44 (A.C.M.); Fax: +420-549-491-327 (V.R.); +33-4-67-41-52-42 (A.C.M.)
| | - Albano C. Meli
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
- Correspondence: (V.R.); (A.C.M.); Tel.: +420-549-498-002 (V.R.); +33-4-67-41-52-44 (A.C.M.); Fax: +420-549-491-327 (V.R.); +33-4-67-41-52-42 (A.C.M.)
| |
Collapse
|
32
|
Wu H, Zhou X, Gong H, Ni Z, Xu Q. Perivascular tissue stem cells are crucial players in vascular disease. Free Radic Biol Med 2021; 165:324-333. [PMID: 33556462 DOI: 10.1016/j.freeradbiomed.2021.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 12/21/2022]
Abstract
Perivascular tissue including adipose layer and adventitia have been considered to play pivotal roles in vascular development and disease progression. Recent studies showed that abundant stem/progenitorcells (SPCs) are present in perivascular tissues. These SPCs exhibit capability to proliferate and differentiate into specific terminal cells. Adult perivascular SPCs are quiescent in normal condition, once activated by specific molecules (e.g., cytokines), they migrate toward the lumen side where they differentiate into both smooth muscle cells (SMCs) and endothelial cells (ECs), thus promoting intima hyperplasia or endothelial regeneration. In addition, perivascular SPCs can also regulate vascular diseases via other ways including but not limited to paracrine effects, matrix protein modulation and microvessel formation. Perivascular SPCs have also been shown to possess therapeutic potentials due to the capability to differentiate into vascular cells and regenerate vascular structures. This review summarizes current knowledge on resident SPCs features and discusses the potential benefits of SPCs therapy in vascular diseases.
Collapse
Affiliation(s)
- Hong Wu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Xuhao Zhou
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Hui Gong
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Zhichao Ni
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, China.
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, China.
| |
Collapse
|
33
|
Skeletal Muscle Subpopulation Rearrangements upon Rhabdomyosarcoma Development through Single-Cell Mass Cytometry. J Clin Med 2021; 10:jcm10040823. [PMID: 33671425 PMCID: PMC7922544 DOI: 10.3390/jcm10040823] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/08/2021] [Accepted: 02/15/2021] [Indexed: 11/16/2022] Open
Abstract
The embryonal rhabdomyosarcoma (eRMS) is a soft tissue sarcoma commonly affecting the head and neck, the extremities and the genitourinary tract. To contribute to revealing the cell types that may originate this tumor, we exploited mass cytometry, a single-cell technique that, by using heavy-metal-tagged antibodies, allows the accurate monitoring of the changes occurring in the mononuclear cell composition of skeletal muscle tissue during tumor development. To this end, we compared cell populations of healthy muscles with those from spatiotemporal-induced eRMS tumors in a mouse model (LSL-KrasG12D/+;Tp53Fl/Fl) that can be used to develop rhabdomyosarcoma by means of infection with an adenovirus vector expressing Cre (Ad-Cre) recombinase. By monitoring different time points after tumor induction, we were able to analyze tumor progression and composition, identifying fibro/adipogenic progenitors (FAPs) as the cell type that, in this model system, had a pivotal role in tumor development. In vitro studies highlighted that both FAPs and satellite cells (SCs), upon infection with the Ad-Cre, acquired the potential to develop rhabdomyosarcomas when transplanted into immunocompromised mice. However, only infected FAPs had an antigen profile that was similar to embryonal rhabdomyosarcoma cells. Overall, our analysis supports the involvement of FAPs in eRMS development.
Collapse
|
34
|
Abstract
Blood vessel formation is a key feature in physiologic and pathologic processes. Once considered a homogeneous cell population that functions as a passive physical barrier between blood and tissue, endothelial cells (ECs) are now recognized to be quite "heterogeneous." While numerous attempts to enhance endothelial repair and replacement have been attempted using so called "endothelial progenitor cells" it is now clear that a better understanding of the origin, location, and activation of stem and progenitor cells of the resident vascular endothelium is required before attempting exogenous cell therapy approaches. This chapter provides an overview for performance of single-cell clonogenic studies of human umbilical cord blood circulating endothelial colony-forming cells (ECFC) that represent distinct precursors for the endothelial lineage with vessel forming potential.
Collapse
|
35
|
Grant D, Wanner N, Frimel M, Erzurum S, Asosingh K. Comprehensive phenotyping of endothelial cells using flow cytometry 2: Human. Cytometry A 2020; 99:257-264. [PMID: 33369145 DOI: 10.1002/cyto.a.24293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In vascular research, clinical samples and samples from animal models are often used together to foster translation of preclinical findings to humans. General concepts of endothelia and murine-specific endothelial phenotypes were discussed in part 1 of this two part series. Here, in part 2, we present a comprehensive overview of human-specific endothelial phenotypes. Pan-endothelial cell markers, organ specific endothelial antigens, and flow cytometric immunophenotyping of blood-borne endothelial cells are reviewed.
Collapse
Affiliation(s)
- Dillon Grant
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nicholas Wanner
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Matthew Frimel
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Serpil Erzurum
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kewal Asosingh
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA.,Flow Cytometry Core Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
36
|
Grant D, Wanner N, Frimel M, Erzurum S, Asosingh K. Comprehensive phenotyping of endothelial cells using flow cytometry 1: Murine. Cytometry A 2020; 99:251-256. [PMID: 33345421 DOI: 10.1002/cyto.a.24292] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 12/21/2022]
Abstract
The endothelium forms a selective barrier between circulating blood or lymph and surrounding tissue. Endothelial cells play an essential role in vessel homeostasis, and identification of these cells is critical in vascular biology research. However, characteristics of endothelial cells differ depending on the location and type of blood or lymph vessel. Endothelial cell subsets are numerous and often identified using different flow cytometric markers, making immunophenotyping these cells complex. In part 1 of this two part review series, we present a comprehensive overview of markers for the flow cytometric identification and phenotyping of murine endothelial subsets. These subsets can be distinguished using a panel of cell surface and intracellular markers shared by all endothelial cells in combination with additional markers of specialized endothelial cell types. This review can be used to determine the best markers for identifying and phenotyping desired murine endothelial cell subsets.
Collapse
Affiliation(s)
- Dillon Grant
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nicholas Wanner
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Matthew Frimel
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Serpil Erzurum
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kewal Asosingh
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA.,Flow Cytometry Core Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
37
|
Deng J, Ni Z, Gu W, Chen Q, Nowak WN, Chen T, Issa Bhaloo S, Zhang Z, Hu Y, Zhou B, Zhang L, Xu Q. Single-cell gene profiling and lineage tracing analyses revealed novel mechanisms of endothelial repair by progenitors. Cell Mol Life Sci 2020; 77:5299-5320. [PMID: 32166394 PMCID: PMC11104897 DOI: 10.1007/s00018-020-03480-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/18/2020] [Accepted: 02/07/2020] [Indexed: 12/20/2022]
Abstract
Stem/progenitor cells (SPCs) have been implicated to participate in vascular repair. However, the exact role of SPCs in endothelial repair of large vessels still remains controversial. This study aimed to delineate the cellular heterogeneity and possible functional role of endogenous vascular SPCs in large vessels. Using single-cell RNA-sequencing (scRNA-seq) and genetic lineage tracing mouse models, we uncovered the cellular heterogeneity of SPCs, i.e., c-Kit+ cells in the mouse aorta, and found that endogenous c-Kit+ cells acquire endothelial cell fate in the aorta under both physiological and pathological conditions. While c-Kit+ cells contribute to aortic endothelial turnover in the atheroprone regions during homeostasis, recipient c-Kit+ cells of nonbone marrow source replace both luminal and microvessel endothelial cells in transplant arteriosclerosis. Single-cell pseudotime analysis of scRNA-seq data and in vitro cell experiments suggest that vascular SPCs display endothelial differentiation potential and undergo metabolic reprogramming during cell differentiation, in which AKT/mTOR-dependent glycolysis is critical for endothelial gene expression. These findings demonstrate a critical role for c-Kit lineage cells in aortic endothelial turnover and replacement, and may provide insights into therapeutic strategies for vascular diseases.
Collapse
Affiliation(s)
- Jiacheng Deng
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Zhichao Ni
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Wenduo Gu
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Qishan Chen
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Witold Norbert Nowak
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Ting Chen
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Shirin Issa Bhaloo
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Zhongyi Zhang
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Yanhua Hu
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai, 200031, China
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK.
| |
Collapse
|
38
|
Mechanisms of Endothelial Regeneration and Vascular Repair and Their Application to Regenerative Medicine. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:52-65. [PMID: 33069720 PMCID: PMC7560161 DOI: 10.1016/j.ajpath.2020.10.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022]
Abstract
Endothelial barrier integrity is required for maintaining vascular homeostasis and fluid balance between the circulation and surrounding tissues and for preventing the development of vascular disease. Despite comprehensive understanding of the molecular mechanisms and signaling pathways that mediate endothelial injury, the regulatory mechanisms responsible for endothelial regeneration and vascular repair are incompletely understood and constitute an emerging area of research. Endogenous and exogenous reparative mechanisms serve to reverse vascular damage and restore endothelial barrier function through regeneration of a functional endothelium and re-engagement of endothelial junctions. In this review, mechanisms that contribute to endothelial regeneration and vascular repair are described. Targeting these mechanisms has the potential to improve outcome in diseases that are characterized by vascular injury, such as atherosclerosis, restenosis, peripheral vascular disease, sepsis, and acute respiratory distress syndrome. Future studies to further improve current understanding of the mechanisms that control endothelial regeneration and vascular repair are also highlighted.
Collapse
|
39
|
The Emerging Role of PPAR Beta/Delta in Tumor Angiogenesis. PPAR Res 2020; 2020:3608315. [PMID: 32855630 PMCID: PMC7443046 DOI: 10.1155/2020/3608315] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/24/2020] [Indexed: 12/31/2022] Open
Abstract
PPARs are ligand-activated transcriptional factors that belong to the nuclear receptor superfamily. Among them, PPAR alpha and PPAR gamma are prone to exert an antiangiogenic effect, whereas PPAR beta/delta has an opposite effect in physiological and pathological conditions. Angiogenesis has been known as a hallmark of cancer, and our recent works also demonstrate that vascular-specific PPAR beta/delta overexpression promotes tumor angiogenesis and progression in vivo. In this review, we will mainly focus on the role of PPAR beta/delta in tumor angiogenesis linked to the tumor microenvironment to further facilitate tumor progression and metastasis. Moreover, the crosstalk between PPAR beta/delta and its downstream key signal molecules involved in tumor angiogenesis will also be discussed, and the network of interplay between them will further be established in the review.
Collapse
|
40
|
Bhagwani A, Thompson AAR, Farkas L. When Innate Immunity Meets Angiogenesis-The Role of Toll-Like Receptors in Endothelial Cells and Pulmonary Hypertension. Front Med (Lausanne) 2020; 7:352. [PMID: 32850883 PMCID: PMC7410919 DOI: 10.3389/fmed.2020.00352] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/12/2020] [Indexed: 01/16/2023] Open
Abstract
Toll-like receptors serve a central role in innate immunity, but they can also modulate cell function in various non-immune cell types including endothelial cells. Endothelial cells are necessary for the organized function of the vascular system, and part of their fundamental role is also the regulation of immune function and inflammation. In this review, we summarize the current knowledge of how Toll-like receptors contribute to the immune and non-immune functions of the endothelial cells.
Collapse
Affiliation(s)
- Aneel Bhagwani
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH, United States
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, United States
| | - A. A. Roger Thompson
- Department of Infection, Immunity & Cardiovascular Disease, Faculty of Medicine, Dentistry & Health, University of Sheffield, Sheffield, United Kingdom
| | - Laszlo Farkas
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
41
|
Brulois K, Rajaraman A, Szade A, Nordling S, Bogoslowski A, Dermadi D, Rahman M, Kiefel H, O'Hara E, Koning JJ, Kawashima H, Zhou B, Vestweber D, Red-Horse K, Mebius RE, Adams RH, Kubes P, Pan J, Butcher EC. A molecular map of murine lymph node blood vascular endothelium at single cell resolution. Nat Commun 2020; 11:3798. [PMID: 32732867 PMCID: PMC7393069 DOI: 10.1038/s41467-020-17291-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/18/2020] [Indexed: 01/17/2023] Open
Abstract
Blood vascular endothelial cells (BECs) control the immune response by regulating blood flow and immune cell recruitment in lymphoid tissues. However, the diversity of BEC and their origins during immune angiogenesis remain unclear. Here we profile transcriptomes of BEC from peripheral lymph nodes and map phenotypes to the vasculature. We identify multiple subsets, including a medullary venous population whose gene signature predicts a selective role in myeloid cell (vs lymphocyte) recruitment to the medulla, confirmed by videomicroscopy. We define five capillary subsets, including a capillary resident precursor (CRP) that displays stem cell and migratory gene signatures, and contributes to homeostatic BEC turnover and to neogenesis of high endothelium after immunization. Cell alignments show retention of developmental programs along trajectories from CRP to mature venous and arterial populations. Our single cell atlas provides a molecular roadmap of the lymph node blood vasculature and defines subset specialization for leukocyte recruitment and vascular homeostasis.
Collapse
Affiliation(s)
- Kevin Brulois
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anusha Rajaraman
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Agata Szade
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Sofia Nordling
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ania Bogoslowski
- Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Denis Dermadi
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Milladur Rahman
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Helena Kiefel
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Edward O'Hara
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jasper J Koning
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Hiroto Kawashima
- Department of Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Tokyo, Japan
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 200031, Beijing, China
| | - Dietmar Vestweber
- Department Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | | | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, University of Münster, Faculty of Medicine, Münster, Germany
| | - Paul Kubes
- Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Junliang Pan
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Eugene C Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA.
- The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
42
|
Lukowski SW, Patel J, Andersen SB, Sim SL, Wong HY, Tay J, Winkler I, Powell JE, Khosrotehrani K. Single-Cell Transcriptional Profiling of Aortic Endothelium Identifies a Hierarchy from Endovascular Progenitors to Differentiated Cells. Cell Rep 2020; 27:2748-2758.e3. [PMID: 31141696 DOI: 10.1016/j.celrep.2019.04.102] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 03/07/2019] [Accepted: 04/22/2019] [Indexed: 12/21/2022] Open
Abstract
The cellular and molecular profiles that govern the endothelial heterogeneity of the circulatory system have yet to be elucidated. Using a data-driven approach to study the endothelial compartment via single-cell RNA sequencing, we characterized cell subpopulations within and assigned them to a defined endothelial hierarchy. We show that two transcriptionally distinct endothelial populations exist within the aorta and, using two independent trajectory analysis methods, confirm that they represent transitioning cells rather than discrete cell types. Gene co-expression analysis revealed crucial regulatory networks underlying each population, including significant metabolic gene networks in progenitor cells. Using mitochondrial activity assays and phenotyping, we confirm that endovascular progenitors display higher mitochondrial content compared to differentiated endothelial cells. The identities of these populations were further validated against bulk RNA sequencing (RNA-seq) data obtained from normal and tumor-derived vasculature. Our findings validate the heterogeneity of the aortic endothelium and previously suggested hierarchy between progenitor and differentiated cells.
Collapse
Affiliation(s)
- Samuel W Lukowski
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jatin Patel
- The University of Queensland Diamantina Institute, Brisbane, QLD 4102, Australia
| | - Stacey B Andersen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Seen-Ling Sim
- The University of Queensland Diamantina Institute, Brisbane, QLD 4102, Australia
| | - Ho Yi Wong
- The University of Queensland Diamantina Institute, Brisbane, QLD 4102, Australia
| | - Joshua Tay
- Faculty of Medicine, Translational Research Institute, Mater Research Institute-The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Ingrid Winkler
- Faculty of Medicine, Translational Research Institute, Mater Research Institute-The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Joseph E Powell
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales Sydney, Sydney, NSW 2010, Australia
| | | |
Collapse
|
43
|
Kim JO, Kim HN, Kim KH, Baek EJ, Park JY, Ha K, Heo DR, Seo MD, Park SG. Development and characterization of a fully human antibody targeting SCF/c-kit signaling. Int J Biol Macromol 2020; 159:66-78. [PMID: 32437800 DOI: 10.1016/j.ijbiomac.2020.05.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 12/23/2022]
Abstract
CD117/c-kit, a tyrosine kinase receptor, plays a critical role in hematopoiesis, pigmentation, and fertility. The overexpression and activation of c-kit are thought to promote tumor growth and have been reported in various cancers, including leukemia, glioblastoma and mastocytosis. To disrupt the SCF/c-kit signaling axis in cancer, we generated a c-kit antagonist human antibody (NN2101) that binds to domain 2/3 of c-kit. This completely blocked the SCF-mediated phosphorylation of c-kit and inhibited TF-1 cell proliferation, erythroleukemia. In addition, the examination of binding affinity using surface plasmon resonance (SPR) assay showed that NN2101 can bind to c-kit of monkeys (KD = 2.92 × 10-10 M), rats (KD = 1.68 × 10-6 M), mice (KD = 11.5 × 10-9 M), and humans (KD = 2.83 × 10-12 M). We showed that NN2101 does not cause antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity. The immunogenicity of NN2101 was similar to that of bevacizumab. Furthermore, the crystal structure of NN2101 Fab was determined and the structure of NN2101 Fab:c-kit complex was modeled. Structural information, as well as mutagenesis results, revealed that NN2101 can bind to the SCF-binding regions of c-kit. Collectively, we generated a c-kit neutralizing human antibody (NN2101) for the treatment of erythroleukemia and characterized its biophysical properties. NN2101 can potentially be used as a therapeutic antibody to treat different cancers.
Collapse
Affiliation(s)
- Jin-Ock Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea
| | - Ha-Neul Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea; Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea
| | - Kwang-Hyeok Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea
| | - Eun Ji Baek
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea
| | - Jeong-Yang Park
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea; Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea
| | - Kyungsoo Ha
- New Drug Development Center, Osong Medical Innovation Foundation, Osong 28160, Republic of Korea
| | - Deok Rim Heo
- New Drug Development Center, Osong Medical Innovation Foundation, Osong 28160, Republic of Korea; College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong 28160, Republic of Korea
| | - Min-Duk Seo
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea; Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea; Novelty Nobility, 227 Unjung-ro, Seongnam-si, Gyeonggi-do 13477, Republic of Korea.
| | - Sang Gyu Park
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea; Novelty Nobility, 227 Unjung-ro, Seongnam-si, Gyeonggi-do 13477, Republic of Korea.
| |
Collapse
|
44
|
Poh KK, Lee PSS, Djohan AH, Galupo MJ, Songco GG, Yeo TC, Tan HC, Richards AM, Ye L. Transplantation of Endothelial Progenitor Cells in Obese Diabetic Rats Following Myocardial Infarction: Role of Thymosin Beta-4. Cells 2020; 9:cells9040949. [PMID: 32290541 PMCID: PMC7226991 DOI: 10.3390/cells9040949] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/17/2020] [Accepted: 04/09/2020] [Indexed: 12/20/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are bone-marrow derived cells that are critical in the maintenance of endothelial wall integrity and protection of ischemic myocardium through the formation of new blood vessels (vasculogenesis) or proliferation of pre-existing vasculature (angiogenesis). Diabetes mellitus (DM) and the metabolic syndrome are commonly associated with ischemic heart disease through its pathological effects on the endothelium and consequent endothelial dysfunction. Thymosin-β4 (Tβ4) which expressed in the embryonic heart is critical in epicardial and coronary artery formation. In this study, we explored the effects of Tβ4 treatment on diabetic EPCs in vitro and intramyocardial injection of Tβ4-treated and non-Tβ4 treated EPCs following acute myocardial infarction (MI) of diabetic rats in vivo. It was found that 10 ng/mL Tβ4 increased migration, tubule formation, and angiogenic factor secretion of diabetic EPCs in vitro. In vivo, although implantation of Tβ4 treated diabetic EPCs significantly increased capillary density and attracted more c-Kit positive progenitor cells into the infarcted hearts as compared with implantation of non-Tβ4 treated diabetic EPCs, the significantly improved left ventricular ejection fraction was only found in the rats which received non-Tβ4 treated EPCs. The data suggests that a low dose Tβ4 increases diabetic EPC migration, tubule formation, and angiogenic factor secretion. However, it did not improve the effects of EPCs on left ventricular pump function in diabetic rats with MI.
Collapse
Affiliation(s)
- Kian Keong Poh
- Department of Cardiology, National University Heart Centre Singapore, National University Health System, Singapore 119074, Singapore; (K.K.P.); (P.S.S.L.); (A.H.D.); (M.J.G.); (G.G.S.); (T.C.Y.); (H.C.T.); (A.M.R.)
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Poay Sian Sabrina Lee
- Department of Cardiology, National University Heart Centre Singapore, National University Health System, Singapore 119074, Singapore; (K.K.P.); (P.S.S.L.); (A.H.D.); (M.J.G.); (G.G.S.); (T.C.Y.); (H.C.T.); (A.M.R.)
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Andie Hartanto Djohan
- Department of Cardiology, National University Heart Centre Singapore, National University Health System, Singapore 119074, Singapore; (K.K.P.); (P.S.S.L.); (A.H.D.); (M.J.G.); (G.G.S.); (T.C.Y.); (H.C.T.); (A.M.R.)
| | - Mary Joyce Galupo
- Department of Cardiology, National University Heart Centre Singapore, National University Health System, Singapore 119074, Singapore; (K.K.P.); (P.S.S.L.); (A.H.D.); (M.J.G.); (G.G.S.); (T.C.Y.); (H.C.T.); (A.M.R.)
| | - Geronica Gorospe Songco
- Department of Cardiology, National University Heart Centre Singapore, National University Health System, Singapore 119074, Singapore; (K.K.P.); (P.S.S.L.); (A.H.D.); (M.J.G.); (G.G.S.); (T.C.Y.); (H.C.T.); (A.M.R.)
| | - Tiong Cheng Yeo
- Department of Cardiology, National University Heart Centre Singapore, National University Health System, Singapore 119074, Singapore; (K.K.P.); (P.S.S.L.); (A.H.D.); (M.J.G.); (G.G.S.); (T.C.Y.); (H.C.T.); (A.M.R.)
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Huay Cheem Tan
- Department of Cardiology, National University Heart Centre Singapore, National University Health System, Singapore 119074, Singapore; (K.K.P.); (P.S.S.L.); (A.H.D.); (M.J.G.); (G.G.S.); (T.C.Y.); (H.C.T.); (A.M.R.)
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Arthur Mark Richards
- Department of Cardiology, National University Heart Centre Singapore, National University Health System, Singapore 119074, Singapore; (K.K.P.); (P.S.S.L.); (A.H.D.); (M.J.G.); (G.G.S.); (T.C.Y.); (H.C.T.); (A.M.R.)
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Lei Ye
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore 169609, Singapore
- Correspondence:
| |
Collapse
|
45
|
Isolation of tissue-resident vascular endothelial stem cells from mouse liver. Nat Protoc 2020; 15:1066-1081. [PMID: 32005982 DOI: 10.1038/s41596-019-0276-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 12/03/2019] [Indexed: 11/09/2022]
Abstract
Endothelial cells (ECs) are fundamental components of the blood vessels that comprise the vascular system; facilitate blood flow; and regulate permeability, angiogenesis, inflammatory responses and homeostatic tissue maintenance. Accumulating evidence suggests there is EC heterogeneity in vivo. However, isolation of fresh ECs from adult mice to investigate this further is challenging. Here, we describe an easy and reproducible protocol for isolation of different types of ECs and CD157+ vascular-resident endothelial stem cells (VESCs) by mechano-enzymatic tissue digestion followed by fluorescence-activated cell sorting. The procedure was established on liver tissue but can be used to isolate ECs from other organs with minimal modification. Preparation of single-cell suspensions can be completed in 2.5 h. We also describe assays for EC clonal and network formation, as well as transcriptomic analysis of isolated ECs. The protocol enables isolation of primary ECs and VESCs that can be used for a wide range of downstream analyses in vascular research.
Collapse
|
46
|
Naito H, Iba T, Takakura N. Mechanisms of new blood-vessel formation and proliferative heterogeneity of endothelial cells. Int Immunol 2020; 32:295-305. [DOI: 10.1093/intimm/dxaa008] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/27/2020] [Indexed: 12/26/2022] Open
Abstract
Abstract
The vast blood-vessel network of the circulatory system is crucial for maintaining bodily homeostasis, delivering essential molecules and blood cells, and removing waste products. Blood-vessel dysfunction and dysregulation of new blood-vessel formation are related to the onset and progression of many diseases including cancer, ischemic disease, inflammation and immune disorders. Endothelial cells (ECs) are fundamental components of blood vessels and their proliferation is essential for new vessel formation, making them good therapeutic targets for regulating the latter. New blood-vessel formation occurs by vasculogenesis and angiogenesis during development. Induction of ECs termed tip, stalk and phalanx cells by interactions between vascular endothelial growth factor A (VEGF-A) and its receptors (VEGFR1–3) and between Notch and Delta-like Notch ligands (DLLs) is crucial for regulation of angiogenesis. Although the importance of angiogenesis is unequivocal in the adult, vasculogenesis effected by endothelial progenitor cells (EPCs) may also contribute to post-natal vessel formation. However, the definition of these cells is ambiguous and they include several distinct cell types under the simple classification of ‘EPC’. Furthermore, recent evidence indicates that ECs within the intima show clonal expansion in some situations and that they may harbor vascular-resident endothelial stem cells. In this article, we summarize recent knowledge on vascular development and new blood-vessel formation in the adult. We also introduce concepts of EC heterogeneity and EC clonal expansion, referring to our own recent findings.
Collapse
Affiliation(s)
- Hisamichi Naito
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Tomohiro Iba
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Nobuyuki Takakura
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Signal Transduction, World Premier Institute Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
47
|
Sun Y, Chen S, Zhang X, Pei M. Significance of Cellular Cross-Talk in Stromal Vascular Fraction of Adipose Tissue in Neovascularization. Arterioscler Thromb Vasc Biol 2020; 39:1034-1044. [PMID: 31018663 DOI: 10.1161/atvbaha.119.312425] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Adult stem cell-based therapy has been regarded as a promising treatment for tissue ischemia because of its ability to promote new blood vessel formation. Bone marrow-derived mesenchymal stem cells are the most used angiogenic cells for therapeutic neovascularization, yet the side effects and low efficacy have limited their clinical application. Adipose stromal vascular fraction is an easily accessible, heterogeneous cell system comprised of endothelial, stromal, and hematopoietic cell lineages, which has been shown to spontaneously form robust, patent, and functional vasculatures in vivo. However, the characteristics of each cell population and their specific roles in neovascularization remain an area of ongoing investigation. In this review, we summarize the functional capabilities of various stromal vascular fraction constituents during the process of neovascularization and attempt to analyze whether the cross-talk between these constituents generates a synergetic effect, thus contributing to the development of new potential therapeutic strategies to promote neovascularization.
Collapse
Affiliation(s)
- Yuan Sun
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| | - Song Chen
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| | - Xicheng Zhang
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| | - Ming Pei
- From the Department of Vascular Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Jiangsu, China (Y.S., X.Z.); Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics (Y.S., M.P.), Exercise Physiology (M.P.), and WVU Cancer Institute, Robert C. Byrd Health Sciences Center (M.P.), West Virginia University, Morgantown; and Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China (S.C.)
| |
Collapse
|
48
|
Clonally selected primitive endothelial cells promote occlusive pulmonary arteriopathy and severe pulmonary hypertension in rats exposed to chronic hypoxia. Sci Rep 2020; 10:1136. [PMID: 31980720 PMCID: PMC6981224 DOI: 10.1038/s41598-020-58083-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 01/10/2020] [Indexed: 12/15/2022] Open
Abstract
One current concept suggests that unchecked proliferation of clonally selected precursors of endothelial cells (ECs) contribute to severe pulmonary arterial hypertension (PAH). We hypothesized that clonally selected ECs expressing the progenitor marker CD117 promote severe occlusive pulmonary hypertension (PH). The remodelled pulmonary arteries of PAH patients harboured CD117+ ECs. Rat lung CD117+ ECs underwent four generations of clonal expansion to enrich hyperproliferative ECs. The resulting clonally enriched ECs behaved like ECs, as measured by in vitro and in vivo angiogenesis assays. The same primitive ECs showed a limited ability for mesenchymal lineage differentiation. Endothelial differentiation and function were enhanced by blocking TGF-β signalling, promoting bone morphogenic protein (BMP) signalling. The transplantation of the EC clones caused arterio-occlusive PH in rats exposed to chronic hypoxia. These EC clones engrafted in the pulmonary arteries. Yet cessation of chronic hypoxia promoted lung cell apoptosis and resolution of vascular lesions. In conclusion, this is to the best of our knowledge, the first report that clonally enriched primitive ECs promote occlusive pulmonary arteriopathy and severe PH. These primitive EC clones further give rise to cells of endothelial and mesenchymal lineage as directed by BMP and TGF-β signaling.
Collapse
|
49
|
The c-kit Receptor Tyrosine Kinase Marks Sweet or Umami Sensing T1R3 Positive Adult Taste Cells in Mice. CHEMOSENS PERCEPT 2020. [DOI: 10.1007/s12078-019-09277-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
50
|
Gholobova D, Terrie L, Gerard M, Declercq H, Thorrez L. Vascularization of tissue-engineered skeletal muscle constructs. Biomaterials 2019; 235:119708. [PMID: 31999964 DOI: 10.1016/j.biomaterials.2019.119708] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 12/10/2019] [Accepted: 12/18/2019] [Indexed: 12/26/2022]
Abstract
Skeletal muscle tissue can be created in vitro by tissue engineering approaches, based on differentiation of muscle stem cells. Several approaches exist and generally result in three dimensional constructs composed of multinucleated myofibers to which we refer as myooids. Engineering methods date back to 3 decades ago and meanwhile a wide range of cell types and scaffold types have been evaluated. Nevertheless, in most approaches, myooids remain very small to allow for diffusion-mediated nutrient supply and waste product removal, typically less than 1 mm thick. One of the shortcomings of current in vitro skeletal muscle organoid development is the lack of a functional vascular structure, thus limiting the size of myooids. This is a challenge which is nowadays applicable to almost all organoid systems. Several approaches to obtain a vascular structure within myooids have been proposed. The purpose of this review is to give a concise overview of these approaches.
Collapse
Affiliation(s)
- D Gholobova
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven, E. Sabbelaan 53, 8500, Kortrijk, Belgium
| | - L Terrie
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven, E. Sabbelaan 53, 8500, Kortrijk, Belgium
| | - M Gerard
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven, E. Sabbelaan 53, 8500, Kortrijk, Belgium
| | - H Declercq
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven, E. Sabbelaan 53, 8500, Kortrijk, Belgium
| | - L Thorrez
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven, E. Sabbelaan 53, 8500, Kortrijk, Belgium.
| |
Collapse
|