1
|
Goeckel ME, Lee J, Levitas A, Colijn S, Mun G, Burton Z, Chintalapati B, Yin Y, Abello J, Stratman A. CXCR3-CXCL11 signaling restricts angiogenesis and promotes pericyte recruitment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.16.557842. [PMID: 37745440 PMCID: PMC10516035 DOI: 10.1101/2023.09.16.557842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Endothelial cell (EC)-pericyte interactions are known to remodel in response to hemodynamic forces, yet there is a lack of mechanistic understanding of the signaling pathways that underlie these events. Here, we have identified a novel signaling network regulated by blood flow in ECs-the chemokine receptor, CXCR3, and one of its ligands, CXCL11-that delimits EC angiogenic potential and suppresses pericyte recruitment during development through regulation of pdgfb expression in ECs. In vitro modeling of EC-pericyte interactions demonstrates that suppression of EC-specific CXCR3 signaling leads to loss of pericyte association with EC tubes. In vivo, phenotypic defects are particularly noted in the cranial vasculature, where we see a loss of pericyte association with and expansion of the vasculature in zebrafish treated with the Cxcr3 inhibitor AMG487. We also demonstrate using flow modeling platforms that CXCR3-deficient ECs are more elongated, move more slowly, and have impaired EC-EC junctions compared to their control counterparts. Together these data suggest that CXCR3 signaling in ECs drives vascular stabilization events during development.
Collapse
Affiliation(s)
- Megan E. Goeckel
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
- University of Nebraska Medical Center, Graduate Studies, Nebraska Medical Center, Omaha, NE 68198
| | - Jihui Lee
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Allison Levitas
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Sarah Colijn
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Geonyoung Mun
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Zarek Burton
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Bharadwaj Chintalapati
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Ying Yin
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Javier Abello
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Amber Stratman
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| |
Collapse
|
2
|
Muturi HT, Ghadieh HE, Abdolahipour R, Stankus HL, Belew GD, Liu JK, Jahromi MS, Lee AD, Singer BB, Angeli-Pahim I, Sehrawat TS, Malhi H, Verhulst S, van Grunsven LA, Zarrinpar A, Duarte S, Najjar SM. Loss of CEACAM1 in endothelial cells causes hepatic fibrosis. Metabolism 2023; 144:155562. [PMID: 37088122 PMCID: PMC10330196 DOI: 10.1016/j.metabol.2023.155562] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/29/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
OBJECTIVES Hepatocytic CEACAM1 plays a critical role in NASH pathogenesis, as bolstered by the development of insulin resistance, visceral obesity, steatohepatitis and fibrosis in mice with global Ceacam1 (Cc1) deletion. In contrast, VECadCre+Cc1fl/fl mice with endothelial loss of Cc1 manifested insulin sensitivity with no visceral obesity despite elevated NF-κB signaling and increased systemic inflammation. We herein investigated whether VECadCre+Cc1fl/fl male mice develop hepatic fibrosis and whether this is mediated by increased production of endothelin1 (ET1), a transcriptional NF-κB target. METHODS VECadCre+Et1.Cc1fl/fl mice with combined endothelial loss of Cc1/Et1 genes were generated. Histological and immunohistochemical analyses were conducted on their livers and on liver tissue biopsies from adult patients undergoing bariatric surgery or from patients with NASH diagnosis receiving liver transplant. RESULTS Hepatic fibrosis and inflammatory infiltration developed in VECadCre+Cc1fl/fl liver parenchyma. This was preceded by increased ET1 production and reversed with combined endothelial loss of Et1. Conditioned media from VECadCre+Cc1fl/fl, but not VECadCre+Et1.Cc1fl/fl primary liver endothelial cells activated wild-type hepatic stellate cells; a process inhibited by bosentan, an ETAR/ETBR dual antagonist. Consistently, immunohistochemical analysis of liver biopsies from patients with NASH showed a decline in endothelial CEACAM1 in parallel with increased plasma endothelin1 levels and progression of hepatic fibrosis stage. CONCLUSIONS The data demonstrated that endothelial CEACAM1 plays a key role in preventing hepatic fibrogenesis by reducing autocrine endothelin1 production.
Collapse
Affiliation(s)
- Harrison T Muturi
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Hilda E Ghadieh
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA; Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Balamand, Al-Koura, Lebanon
| | - Raziyeh Abdolahipour
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Hannah L Stankus
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Getachew Debas Belew
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - James K Liu
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Marziyeh Salehi Jahromi
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Abraham D Lee
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA; Department of Exercise and Rehabilitation Sciences, College of Health and Human Services, University of Toledo, Toledo, OH, USA
| | - Bernhard B Singer
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Isabella Angeli-Pahim
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Tejasav S Sehrawat
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Stefaan Verhulst
- Liver Cell Biology Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Leo A van Grunsven
- Liver Cell Biology Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ali Zarrinpar
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Sergio Duarte
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Sonia M Najjar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA; Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA; Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
| |
Collapse
|
3
|
Lu P, Wang P, Wu B, Wang Y, Liu Y, Cheng W, Feng X, Yuan X, Atteya MM, Ferro H, Sugi Y, Rydquist G, Esmaily M, Butcher JT, Chang CP, Lenz J, Zheng D, Zhou B. A SOX17-PDGFB signaling axis regulates aortic root development. Nat Commun 2022; 13:4065. [PMID: 35831318 PMCID: PMC9279414 DOI: 10.1038/s41467-022-31815-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 06/30/2022] [Indexed: 11/08/2022] Open
Abstract
Developmental etiologies causing complex congenital aortic root abnormalities are unknown. Here we show that deletion of Sox17 in aortic root endothelium in mice causes underdeveloped aortic root leading to a bicuspid aortic valve due to the absence of non-coronary leaflet and mispositioned left coronary ostium. The respective defects are associated with reduced proliferation of non-coronary leaflet mesenchyme and aortic root smooth muscle derived from the second heart field cardiomyocytes. Mechanistically, SOX17 occupies a Pdgfb transcriptional enhancer to promote its transcription and Sox17 deletion inhibits the endothelial Pdgfb transcription and PDGFB growth signaling to the non-coronary leaflet mesenchyme. Restoration of PDGFB in aortic root endothelium rescues the non-coronary leaflet and left coronary ostium defects in Sox17 nulls. These data support a SOX17-PDGFB axis underlying aortic root development that is critical for aortic valve and coronary ostium patterning, thereby informing a potential shared disease mechanism for concurrent anomalous aortic valve and coronary arteries.
Collapse
Affiliation(s)
- Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ping Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- School of Medical Imaging, Tianjin Medical University, Tianjin, China
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yidong Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Cardiovascular Research Center, School of Basic Medical Sciences, Jiaotong University, Xi'an, Shanxi, China
| | - Yang Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Wei Cheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xuhui Feng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xinchun Yuan
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Miriam M Atteya
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Haleigh Ferro
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Yukiko Sugi
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Grant Rydquist
- School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Mahdi Esmaily
- School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | | | - Ching-Pin Chang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jack Lenz
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
- Departments of Neurology and Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Bin Zhou
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
- Departments of Pediatrics and Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
4
|
Tao RH, Kobayashi M, Yang Y, Kleinerman ES. Exercise Inhibits Doxorubicin-Induced Damage to Cardiac Vessels and Activation of Hippo/YAP-Mediated Apoptosis. Cancers (Basel) 2021; 13:cancers13112740. [PMID: 34205942 PMCID: PMC8198139 DOI: 10.3390/cancers13112740] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/20/2021] [Accepted: 05/27/2021] [Indexed: 01/16/2023] Open
Abstract
Dose-related cardiomyopathy is a major side effect following doxorubicin (Dox). To investigate whether exercise (Ex)-induced vasculogenesis plays a role in reducing Dox-induced cardiotoxicity, GFP+ bone marrow (BM) cells from GFP transgenic mice were transplanted into wild-type mice. Transplanted mice were treated with Dox, Ex, Dox+Ex, or control. We found Dox therapy resulted in decreased systolic and diastolic blood flow, decreased ejection fraction and fractional shortening, and decreased vascular endothelial cells and pericytes. These abnormalities were not seen in Dox+Ex hearts. Heart tissues from control-, Ex-, or Dox-treated mice showed a small number of GFP+ cells. By contrast, the Dox+Ex-treated hearts had a significant increase in GFP+ cells. Further analyses demonstrated these GFP+ BM cells had differentiated into vascular endothelial cells (GFP+CD31+) and pericytes (GFP+NG2+). Decreased cardiomyocytes were also seen in Dox-treated but not Dox+Ex-treated hearts. Ex induced an increase in GFP+c-Kit+ cells. However, these c-Kit+ BM stem cells had not differentiated into cardiomyocytes. Dox therapy induced phosphorylation of MST1/2, LATS1, and YAP; a decrease in total YAP; and cleavage of caspase-3 and PARP in the heart tissues. Dox+Ex prevented these effects. Our data demonstrated Dox-induced cardiotoxicity is mediated by vascular damage resulting in decreased cardiac blood flow and through activation of Hippo-YAP signaling resulting in cardiomyocyte apoptosis. Furthermore, Ex inhibited these effects by promoting migration of BM stem cells into the heart to repair the cardiac vessels damaged by Dox and through inhibiting Dox-induced Hippo-YAP signaling-mediated apoptosis. These data support the concept of using exercise as an intervention to decrease Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Rong-Hua Tao
- Department of Pediatrics-Research, Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
- Correspondence: (R.-H.T.); (E.S.K.); Tel.: +1-(713)-563-7333 (R.-H.T.); +1-(713)-792-8110 (E.S.K.); Fax: +1-(713)-563-5407 (R.-H.T.); +1-(713)-563-5407 (E.S.K.)
| | - Masato Kobayashi
- School of Health Sciences, Institutes of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa 920-0942, Japan;
| | - Yuanzheng Yang
- Department of Pediatrics-Research, Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
| | - Eugenie S. Kleinerman
- Department of Pediatrics-Research, Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
- Correspondence: (R.-H.T.); (E.S.K.); Tel.: +1-(713)-563-7333 (R.-H.T.); +1-(713)-792-8110 (E.S.K.); Fax: +1-(713)-563-5407 (R.-H.T.); +1-(713)-563-5407 (E.S.K.)
| |
Collapse
|
5
|
Chemokine mediated signalling within arteries promotes vascular smooth muscle cell recruitment. Commun Biol 2020; 3:734. [PMID: 33277595 PMCID: PMC7719186 DOI: 10.1038/s42003-020-01462-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 11/05/2020] [Indexed: 01/13/2023] Open
Abstract
The preferential accumulation of vascular smooth muscle cells (vSMCs) on arteries versus veins during early development is a well-described phenomenon, but the molecular pathways underlying this polarization are not well understood. In zebrafish, the cxcr4a receptor (mammalian CXCR4) and its ligand cxcl12b (mammalian CXCL12) are both preferentially expressed on arteries at time points consistent with the arrival and differentiation of the first vSMCs during vascular development. We show that autocrine cxcl12b/cxcr4 activity leads to increased production of the vSMC chemoattractant ligand pdgfb by endothelial cells in vitro and increased expression of pdgfb by arteries of zebrafish and mice in vivo. Additionally, we demonstrate that expression of the blood flow-regulated transcription factor klf2a in primitive veins negatively regulates cxcr4/cxcl12 and pdgfb expression, restricting vSMC recruitment to the arterial vasculature. Together, this signalling axis leads to the differential acquisition of vSMCs at sites where klf2a expression is low and both cxcr4a and pdgfb are co-expressed, i.e. arteries during early development. Stratman et al. provide evidence linking the cxcl12b/cxcr4a signaling axis in endothelial cells to an increased release of platelet-derived growth factor b, leading to the recruitment of smooth muscle cells to developing arteries. This signalling axis is suppressed in the venous endothelium during early development by the high expression of blood flow-regulated transcription factor klf2a.
Collapse
|
6
|
Wang C, Liu Y, He D. Diverse effects of platelet-derived growth factor-BB on cell signaling pathways. Cytokine 2019; 113:13-20. [DOI: 10.1016/j.cyto.2018.10.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/17/2018] [Accepted: 10/19/2018] [Indexed: 12/12/2022]
|
7
|
Huang Y, Winklbauer R. Cell migration in the Xenopus gastrula. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 7:e325. [PMID: 29944210 DOI: 10.1002/wdev.325] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 05/22/2018] [Accepted: 05/30/2018] [Indexed: 12/17/2022]
Abstract
Xenopus gastrulation movements are in large part based on the rearrangement of cells by differential cell-on-cell migration within multilayered tissues. Different patterns of migration-based cell intercalation drive endoderm and mesoderm internalization and their positioning along their prospective body axes. C-cadherin, fibronectin, integrins, and focal contact components are expressed in all gastrula cells and play putative roles in cell-on-cell migration, but their actual functions in this respect are not yet understood. The gastrula can be subdivided into two motility domains, and in the vegetal, migratory domain, two modes of cell migration are discerned. Vegetal endoderm cells show ingression-type migration, a variant of amoeboid migration characterized by the lack of locomotory protrusions and by macropinocytosis as a mechanism of trailing edge resorption. Mesendoderm and prechordal mesoderm cells use lamellipodia in a mesenchymal mode of migration. Gastrula cell motility can be dissected into traits, such as cell polarity, adhesion, mobility, or protrusive activity, which are controlled separately yet in complex, combinatorial ways. Cells can instantaneously switch between different combinations of traits, showing plasticity as they respond to substratum properties. This article is categorized under: Early Embryonic Development > Gastrulation and Neurulation.
Collapse
Affiliation(s)
- Yunyun Huang
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Rudolf Winklbauer
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Berning P, Schaefer C, Clemens D, Korsching E, Dirksen U, Potratz J. The CXCR4 antagonist plerixafor (AMD3100) promotes proliferation of Ewing sarcoma cell lines in vitro and activates receptor tyrosine kinase signaling. Cell Commun Signal 2018; 16:21. [PMID: 29776413 PMCID: PMC5960216 DOI: 10.1186/s12964-018-0233-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/10/2018] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The CXCR4 receptor antagonist plerixafor (AMD3100) is raising interest as an anti-cancer agent that disrupts the CXCL12-CXCR4 chemokine - receptor interaction between neoplastic cells and their microenvironment in tumor progression and metastasis. Here, we investigated plerixafor for anti-cancer activity in Ewing sarcoma, a rare and aggressive cancer of bone and soft tissues. METHODS We used a variety of methods such as cell viability and migration assays, flow cytometry, phospho-tyrosine arrays and western blotting to determine plerixafor effects on five characterized Ewing sarcoma cell lines and a low-passage culture in vitro. RESULTS Unexpectedly, plerixafor led to an increase in cell viability and proliferation in standard cell growth conditions, and to chemotactic migration towards plerixafor. Exploring potential molecular mechanisms underlying this effect, we found that Ewing sarcoma cell lines divided into classes of high- and low-level CXCR4 surface expression. Proliferative plerixafor responses were observed in both groups, maintained despite significant CXCR4 down-regulation or inhibition of Gαi-protein signal transduction, and involved activation of multiple receptor tyrosine kinases (DDR2, MERTK, MST1R, NTRK1, RET), the most prominent being platelet-derived growth factor receptor beta (PDGFRB). PDGFRB was activated in response to inhibition of the CXCL12-CXCR4 axis by plerixafor and/or pertussis toxin (Gαi-protein inhibitor). Dasatinib, a multi-kinase inhibitor of both PDGFRB and the CXCR4 downstream kinase SRC, counteracted this activation in some but not all cell lines. CONCLUSION These data suggest a feedback interaction between the CXCR4 chemokine receptor and RTK signaling cascades that elicits compensatory cell survival signaling and can shift the net effect of plerixafor towards proliferation. PDGFRB was identified as a candidate driver RTK and potential therapeutic co-target for CXCR4 in Ewing sarcoma. Although as yet limited to in vitro studies, these findings call for further investigation in the cancer - microenvironment interplay in vivo.
Collapse
Affiliation(s)
- Philipp Berning
- Department of Pediatric Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.,Present address: Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Christiane Schaefer
- Department of Pediatric Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.,Division of Hematology and Oncology, Department of Pediatrics III, West German Cancer Centre, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Dagmar Clemens
- Department of Pediatric Hematology and Oncology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Eberhard Korsching
- Institute of Bioinformatics, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Uta Dirksen
- Division of Hematology and Oncology, Department of Pediatrics III, West German Cancer Centre, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Jenny Potratz
- Department of General Pediatrics, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.
| |
Collapse
|
9
|
The chemokines CXCL12 and CXCL14 differentially regulate connective tissue markers during limb development. Sci Rep 2017; 7:17279. [PMID: 29222527 PMCID: PMC5722906 DOI: 10.1038/s41598-017-17490-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 11/27/2017] [Indexed: 12/27/2022] Open
Abstract
Connective tissues (CT) support and connect organs together. Understanding the formation of CT is important, as CT deregulation leads to fibrosis. The identification of CT specific markers has contributed to a better understanding of CT function during development. In developing limbs, Osr1 transcription factor is involved in the differentiation of irregular CT while the transcription factor Scx labels tendon. In this study, we show that the CXCL12 and CXCL14 chemokines display distinct expression pattern in limb CT during chick development. CXCL12 positively regulates the expression of OSR1 and COL3A1, a collagen subtype of irregular CT, while CXCL14 activates the expression of the tendon marker SCX. We provide evidence that the CXCL12 effect on irregular CT involves CXCR4 receptor and vessels. In addition, the expression of CXCL12, CXCL14 and OSR genes is suppressed by the anti-fibrotic BMP signal. Finally, mechanical forces, known to be involved in adult fibrosis, control the expression of chemokines, CT-associated transcription factors and collagens during limb development. Such unexpected roles of CXCL12 and CXCL14 chemokines during CT differentiation can contribute to a better understanding of the fibrosis mechanisms in adult pathological conditions.
Collapse
|
10
|
Abstract
Fibrosis is part of a tissue repair response to injury, defined as increased deposition of extracellular matrix. In some instances, fibrosis is beneficial; however, in the majority of diseases fibrosis is detrimental. Virtually all chronic progressive diseases are associated with fibrosis, representing a huge number of patients worldwide. Fibrosis occurs in all organs and tissues, becomes irreversible with time and further drives loss of tissue function. Various cells types initiate and perpetuate pathological fibrosis by paracrine activation of the principal cellular executors of fibrosis, i.e. stromal mesenchymal cells like fibroblasts, pericytes and myofibroblasts. Multiple pathways are involved in fibrosis, platelet-derived growth factor (PDGF)-signaling being one of the central mediators. Stromal mesenchymal cells express both PDGF receptors (PDGFR) α and β, activation of which drives proliferation, migration and production of extracellular matrix, i.e. the principal processes of fibrosis. Here, we review the role of PDGF signaling in organ fibrosis, with particular focus on the more recently described ligands PDGF-C and -D. We discuss the potential challenges, opportunities and open questions in using PDGF as a potential target for anti-fibrotic therapies.
Collapse
Affiliation(s)
| | - Jürgen Floege
- Division of Nephrology, RWTH University of Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH University of Aachen, Germany; Division of Nephrology, RWTH University of Aachen, Germany.
| |
Collapse
|
11
|
Núñez-Gómez E, Pericacho M, Ollauri-Ibáñez C, Bernabéu C, López-Novoa JM. The role of endoglin in post-ischemic revascularization. Angiogenesis 2016; 20:1-24. [PMID: 27943030 DOI: 10.1007/s10456-016-9535-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
Abstract
Following arterial occlusion, blood vessels respond by forming a new network of functional capillaries (angiogenesis), by reorganizing preexisting capillaries through the recruitment of smooth muscle cells to generate new arteries (arteriogenesis) and by growing and remodeling preexisting collateral arterioles into physiologically relevant arteries (collateral development). All these processes result in the recovery of organ perfusion. The importance of endoglin in post-occlusion reperfusion is sustained by several observations: (1) endoglin expression is increased in vessels showing active angiogenesis/remodeling; (2) genetic endoglin haploinsufficiency in humans causes deficient angiogenesis; and (3) the reduction of endoglin expression by gene disruption or the administration of endoglin-neutralizing antibodies reduces angiogenesis and revascularization. However, the precise role of endoglin in the several processes associated with revascularization has not been completely elucidated and, in some cases, the function ascribed to endoglin by different authors is controversial. The purpose of this review is to organize in a critical way the information available for the role of endoglin in several phenomena (angiogenesis, arteriogenesis and collateral development) associated with post-ischemic revascularization.
Collapse
Affiliation(s)
- Elena Núñez-Gómez
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Miguel Pericacho
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Claudia Ollauri-Ibáñez
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Carmelo Bernabéu
- Centro de Investigaciones Biológicas, Spanish National Research Council (CIB, CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - José M López-Novoa
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain. .,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain.
| |
Collapse
|
12
|
Pinto MP, Sotomayor P, Carrasco-Avino G, Corvalan AH, Owen GI. Escaping Antiangiogenic Therapy: Strategies Employed by Cancer Cells. Int J Mol Sci 2016; 17:ijms17091489. [PMID: 27608016 PMCID: PMC5037767 DOI: 10.3390/ijms17091489] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 08/22/2016] [Accepted: 08/30/2016] [Indexed: 12/29/2022] Open
Abstract
Tumor angiogenesis is widely recognized as one of the "hallmarks of cancer". Consequently, during the last decades the development and testing of commercial angiogenic inhibitors has been a central focus for both basic and clinical cancer research. While antiangiogenic drugs are now incorporated into standard clinical practice, as with all cancer therapies, tumors can eventually become resistant by employing a variety of strategies to receive nutrients and oxygen in the event of therapeutic assault. Herein, we concentrate and review in detail three of the principal mechanisms of antiangiogenic therapy escape: (1) upregulation of compensatory/alternative pathways for angiogenesis; (2) vasculogenic mimicry; and (3) vessel co-option. We suggest that an understanding of how a cancer cell adapts to antiangiogenic therapy may also parallel the mechanisms employed in the bourgeoning tumor and isolated metastatic cells delivering responsible for residual disease. Finally, we speculate on strategies to adapt antiangiogenic therapy for future clinical uses.
Collapse
Affiliation(s)
- Mauricio P Pinto
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile.
| | - Paula Sotomayor
- Center for Integrative Medicine and Innovative Science, Facultad de Medicina, Universidad Andrés Bello, Santiago 8370071, Chile.
| | - Gonzalo Carrasco-Avino
- Department of Pathology, Faculty of Medicine, Universidad de Chile, Santiago 8380456, Chile.
| | - Alejandro H Corvalan
- Department of Hematology-Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330032, Chile.
- Center UC Investigation in Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago 8330023, Chile.
| | - Gareth I Owen
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile.
- Center UC Investigation in Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago 8330023, Chile.
- Biomedical Research Consortium of Chile, Santiago 8331150, Chile.
- Millennium Institute on Immunology & Immunotherapy, Santiago 8331150, Chile.
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago 8380492, Chile.
| |
Collapse
|
13
|
Mesenchymal stem cells generate pericytes to promote tumor recurrence via vasculogenesis after stereotactic body radiation therapy. Cancer Lett 2016; 375:349-359. [DOI: 10.1016/j.canlet.2016.02.033] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/30/2016] [Accepted: 02/17/2016] [Indexed: 01/16/2023]
|
14
|
Rossi E, Smadja DM, Boscolo E, Langa C, Arevalo MA, Pericacho M, Gamella-Pozuelo L, Kauskot A, Botella LM, Gaussem P, Bischoff J, Lopez-Novoa JM, Bernabeu C. Endoglin regulates mural cell adhesion in the circulatory system. Cell Mol Life Sci 2016; 73:1715-39. [PMID: 26646071 PMCID: PMC4805714 DOI: 10.1007/s00018-015-2099-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/19/2015] [Accepted: 11/23/2015] [Indexed: 02/06/2023]
Abstract
The circulatory system is walled off by different cell types, including vascular mural cells and podocytes. The interaction and interplay between endothelial cells (ECs) and mural cells, such as vascular smooth muscle cells or pericytes, play a pivotal role in vascular biology. Endoglin is an RGD-containing counter-receptor for β1 integrins and is highly expressed by ECs during angiogenesis. We find that the adhesion between vascular ECs and mural cells is enhanced by integrin activators and inhibited upon suppression of membrane endoglin or β1-integrin, as well as by addition of soluble endoglin (SolEng), anti-integrin α5β1 antibody or an RGD peptide. Analysis of different endoglin mutants, allowed the mapping of the endoglin RGD motif as involved in the adhesion process. In Eng (+/-) mice, a model for hereditary hemorrhagic telangectasia type 1, endoglin haploinsufficiency induces a pericyte-dependent increase in vascular permeability. Also, transgenic mice overexpressing SolEng, an animal model for preeclampsia, show podocyturia, suggesting that SolEng is responsible for podocytes detachment from glomerular capillaries. These results suggest a critical role for endoglin in integrin-mediated adhesion of mural cells and provide a better understanding on the mechanisms of vessel maturation in normal physiology as well as in pathologies such as preeclampsia or hereditary hemorrhagic telangiectasia.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Cell Adhesion/physiology
- Cell Line, Tumor
- Disease Models, Animal
- Endoglin
- Endothelium, Vascular/metabolism
- Female
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Integrin beta1/genetics
- Jurkat Cells
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Mice, Transgenic
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Neovascularization, Pathologic/metabolism
- Pericytes/metabolism
- Podocytes/metabolism
- Pre-Eclampsia/genetics
- Pre-Eclampsia/pathology
- Pregnancy
- Protein Binding
- RNA Interference
- RNA, Small Interfering
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Retina/metabolism
- Telangiectasia, Hereditary Hemorrhagic/genetics
- Telangiectasia, Hereditary Hemorrhagic/pathology
Collapse
Affiliation(s)
- Elisa Rossi
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), c/Ramiro de Maeztu 9, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain
- Paris Descartes University, Sorbonne Paris Cite, Paris, France
- Hematology Department, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - David M Smadja
- Hematology Department, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
- Faculté de Pharmacie, Inserm UMR-S1140, Paris, France
| | - Elisa Boscolo
- Department of Surgery, Harvard Medical School, Children's Hospital, Boston, MA, 02115, USA
| | - Carmen Langa
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), c/Ramiro de Maeztu 9, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain
| | - Miguel A Arevalo
- Departamento de Anatomía e Histología Humanas, Facultad de Medicina, Universidad de Salamanca, 37007, Salamanca, Spain
- Instituto de Investigaciones Biomédicas de Salamanca (IBSAL), 37007, Salamanca, Spain
| | - Miguel Pericacho
- Instituto de Investigaciones Biomédicas de Salamanca (IBSAL), 37007, Salamanca, Spain
- Departamento de Fisiología y Farmacología, Unidad de Fisiopatología Renal y Cardiovascular, Universidad de Salamanca, 37007, Salamanca, Spain
| | - Luis Gamella-Pozuelo
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), c/Ramiro de Maeztu 9, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain
- Departamento de Fisiología y Farmacología, Unidad de Fisiopatología Renal y Cardiovascular, Universidad de Salamanca, 37007, Salamanca, Spain
| | - Alexandre Kauskot
- Inserm UMR-S1176, Le Kremlin Bicêtre, Paris, France
- Université Paris Sud, Le Kremlin Bicêtre, Paris, France
| | - Luisa M Botella
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), c/Ramiro de Maeztu 9, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain
| | - Pascale Gaussem
- Hematology Department, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
- Faculté de Pharmacie, Inserm UMR-S1140, Paris, France
| | - Joyce Bischoff
- Department of Surgery, Harvard Medical School, Children's Hospital, Boston, MA, 02115, USA
| | - José M Lopez-Novoa
- Instituto de Investigaciones Biomédicas de Salamanca (IBSAL), 37007, Salamanca, Spain
- Departamento de Fisiología y Farmacología, Unidad de Fisiopatología Renal y Cardiovascular, Universidad de Salamanca, 37007, Salamanca, Spain
| | - Carmelo Bernabeu
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), c/Ramiro de Maeztu 9, 28040, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain.
| |
Collapse
|
15
|
Noskovičová N, Petřek M, Eickelberg O, Heinzelmann K. Platelet-Derived Growth Factor Signaling in the Lung. From Lung Development and Disease to Clinical Studies. Am J Respir Cell Mol Biol 2015; 52:263-84. [DOI: 10.1165/rcmb.2014-0294tr] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
16
|
Nakagawa Y, Chiba K. Role of microglial m1/m2 polarization in relapse and remission of psychiatric disorders and diseases. Pharmaceuticals (Basel) 2014; 7:1028-48. [PMID: 25429645 PMCID: PMC4276905 DOI: 10.3390/ph7121028] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/14/2014] [Accepted: 11/14/2014] [Indexed: 12/27/2022] Open
Abstract
Psychiatric disorders such as schizophrenia and major depressive disorder were thought to be caused by neurotransmitter abnormalities. Patients with these disorders often experience relapse and remission; however the underlying molecular mechanisms of relapse and remission still remain unclear. Recent advanced immunological analyses have revealed that M1/M2 polarization of macrophages plays an important role in controlling the balance between promotion and suppression in inflammation. Microglial cells share certain characteristics with macrophages and contribute to immune-surveillance in the central nervous system (CNS). In this review, we summarize immunoregulatory functions of microglia and discuss a possible role of microglial M1/M2 polarization in relapse and remission of psychiatric disorders and diseases. M1 polarized microglia can produce pro-inflammatory cytokines, reactive oxygen species, and nitric oxide, suggesting that these molecules contribute to dysfunction of neural network in the CNS. Alternatively, M2 polarized microglia express cytokines and receptors that are implicated in inhibiting inflammation and restoring homeostasis. Based on these aspects, we propose a possibility that M1 and M2 microglia are related to relapse and remission, respectively in psychiatric disorders and diseases. Consequently, a target molecule skewing M2 polarization of microglia may provide beneficial therapies for these disorders and diseases in the CNS.
Collapse
Affiliation(s)
- Yutaka Nakagawa
- Research Strategy and Planning Department, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama 227-0033, Japan.
| | - Kenji Chiba
- Advanced Medical Research Laboratories, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama 227-0033, Japan.
| |
Collapse
|
17
|
Seeger H, Braun N, Latus J, Alscher MD, Fritz P, Edenhofer I, Biegger D, Lindenmeier M, Wüthrich RP, Segerer S. Platelet-derived growth factor receptor-β expression in human peritoneum. Nephron Clin Pract 2014; 128:178-84. [PMID: 25376624 DOI: 10.1159/000368241] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 08/15/2014] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Simple peritoneal fibrosis and encapsulating peritoneal sclerosis (EPS) are important lesions in the peritoneum of patients on peritoneal dialysis (PD). We have previously described a population of podoplanin-positive myofibroblasts in peritoneal biopsies from patients with EPS. Platelet-derived growth factor receptor-β (PDGFRβ) is a marker of pericytes, and PDGFs might be involved in the fibrotic response of the peritoneum. This study aimed to describe PDGFRβ in the human peritoneum. METHODS In this retrospective analysis, we localized PDGFRβ in peritoneal biopsies from patients with EPS (n = 6) and patients on PD without signs of EPS (n = 5), and compared them with normal peritoneum (n = 4) and peritoneum from uremic patients (n = 5). Consecutive sections were stained for smooth-muscle actin (SMA) and podoplanin. Slides were scored semiquantitatively by 2 observers blinded to the diagnosis. RESULTS PDGFRβ was expressed by cells of arterial walls in all biopsies. A prominent population of PDGFRβ-positive cells was present in the normal peritoneum, which were SMA negative on consecutive sections. In patients on PD, a high number of PDGFRβ were also positive for SMA. In EPS, the majority of podoplanin-positive cells were positive for PDGFRβ. In peritoneal biopsies from normal and uremic patients, the expression of SMA was mainly restricted to cells of arterial walls. Podoplanin expression was restricted to lymphatic vessels in normal peritoneum, in uremic patients, and in patients on PD without EPS. CONCLUSIONS As podoplanin-positive myofibroblasts express PDGFRβ, these cells might be related to pericytes (rather than other sources of fibroblasts). PDGFRβ might turn out to be a therapeutic target in EPS.
Collapse
Affiliation(s)
- Harald Seeger
- Division of Nephrology, University Hospital, University of Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Boer JC, Walenkamp AME, den Dunnen WFA. Recruitment of bone marrow derived cells during anti-angiogenic therapy in GBM: the potential of combination strategies. Crit Rev Oncol Hematol 2014; 92:38-48. [PMID: 24933160 DOI: 10.1016/j.critrevonc.2014.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 04/10/2014] [Accepted: 05/02/2014] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma (GBM) is a highly vascular tumor characterized by rapid and invasive tumor growth, followed by oxygen depletion, hypoxia and neovascularization, which generate a network of disorganized, tortuous and permeable vessels. Recruitment of bone marrow derived cells (BMDC) is crucial for vasculogenesis. These cells may act as vascular progenitors by integrating into the newly formed blood vessels or as vascular modulators by releasing pro-angiogenic factors. In patients with recurrent GBM, anti-vascular endothelial growth factor (VEGF) therapy has been evaluated in combination with chemotherapy, yielding improvements in progression-free survival (PFS). However, benefits are temporary as vascular tumors acquire angiogenic pathways independently of VEGF. Specifically, acute hypoxia following prolonged VEGF depletion induces the recruitment of certain myeloid cell subpopulations, which highly contribute to treatment refractoriness. Here we review the molecular mechanisms of neovascularization in relation to bevacizumab therapy with special emphasis on the recruitment of BMDCs and possible combination therapies for GBM patients.
Collapse
Affiliation(s)
- Jennifer C Boer
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Annemiek M E Walenkamp
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wilfred F A den Dunnen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
19
|
Kovacic N, Croucher PI, McDonald MM. Signaling between tumor cells and the host bone marrow microenvironment. Calcif Tissue Int 2014; 94:125-39. [PMID: 24046000 DOI: 10.1007/s00223-013-9794-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 08/25/2013] [Indexed: 02/04/2023]
Abstract
Tumor cells with high skeletal homing affinity express numerous cell surface receptors that bind ligands produced in bone. Upon arrival, these cells survive in the host environment, encompassed in close proximity to bone marrow cells. Interactions between tumor cells and cells of the host microenvironment are essential to not only tumor cell survival but also their activation and proliferation into environment-modifying tumors. Through the production of RANKL, PTHrP, cytokines, and integrins, activated tumor cells stimulate osteoclastogenesis, enhance bone resorption, and subsequently release matrix-bound proteins that further promote tumor growth and bone resorption. In addition, alterations in the TGF-β/BMP and Wnt signaling pathways via tumor cell growth can either stimulate or suppress osteoblastic bone formation and function, leading to sclerotic or lytic bone disease, respectively. Hence, the presence of tumor cells in bone dysregulates bone remodeling, dramatically impairing skeletal integrity. Furthermore, through complex mechanisms, cells of the immune system interact with tumor cells to further impact bone remodeling. Lastly, with alterations in bone cell activity, the environment is permissive to promoting tumor growth further, suggesting an interdependence between tumor cells and bone cells in metastatic bone disease and multiple myeloma.
Collapse
Affiliation(s)
- Natasa Kovacic
- Bone Biology Group, Musculoskeletal Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia
| | | | | |
Collapse
|
20
|
Hamdan R, Zhou Z, Kleinerman ES. Blocking SDF-1α/CXCR4 downregulates PDGF-B and inhibits bone marrow-derived pericyte differentiation and tumor vascular expansion in Ewing tumors. Mol Cancer Ther 2013; 13:483-91. [PMID: 24282276 DOI: 10.1158/1535-7163.mct-13-0447] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Bone marrow cells (BMC) are critical to the expansion of the tumor vessel network that supports Ewing sarcoma growth. BMCs migrate to the tumor and differentiate into endothelial cells and pericytes. We recently demonstrated that stromal-derived growth factor 1α (SDF-1α) regulates platelet-derived growth factor B (PDGF-B) and that this pathway plays a critical role in bone marrow-derived pericyte differentiation in vitro. We investigated the role of SDF-1α/PDGF-B in the tumor microenvironment in vivo in promoting bone marrow-derived pericyte differentiation in Ewing tumors. The CXCR4 antagonist AMD 3100 was used to disrupt the SDF-1α/CXCR4 axis in vivo in two xenograft Ewing tumor models. BMCs from GFP(+) transgenic mice were transplanted into lethally irradiated nude mice to track BMC migration to the tumor site. Following BMC engraftment, tumor-bearing mice received daily subcutaneous injections of either PBS or AMD 3100 for 3 weeks. Tumors were resected and tumor sections were analyzed by immunohistochemistry. AMD 3100 inhibited BMC differentiation into desmin(+) and NG2(+) pericytes, affected the morphology of the tumor vasculature, decreased perfusion, and increased tumor cell apoptosis. We observed smaller vessels with tiny lumens and a decrease in the microvessel density. AMD 3100 also inhibited PDGF-B protein expression in vitro and in vivo. SDF-1α in the tumor microenvironment plays a critical role in promoting pericyte formation and Ewing sarcoma tumor neovascularization by regulating PDGF-B expression. Interfering with this pathway affects tumor vascular morphology and expansion.
Collapse
MESH Headings
- Animals
- Benzylamines
- Blotting, Western
- Bone Marrow Cells/metabolism
- Bone Marrow Transplantation/methods
- Cell Differentiation/drug effects
- Cell Line, Tumor
- Cell Survival/drug effects
- Chemokine CXCL12/antagonists & inhibitors
- Chemokine CXCL12/metabolism
- Cyclams
- Down-Regulation
- Female
- Heterocyclic Compounds/pharmacology
- Humans
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Microscopy, Fluorescence
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/prevention & control
- Pericytes/metabolism
- Proto-Oncogene Proteins c-sis/metabolism
- Receptors, CXCR4/antagonists & inhibitors
- Receptors, CXCR4/metabolism
- Sarcoma, Ewing/genetics
- Sarcoma, Ewing/metabolism
- Sarcoma, Ewing/prevention & control
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Randala Hamdan
- Corresponding Author: Eugenie S. Kleinerman, Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center, Houston TX 77030.
| | | | | |
Collapse
|
21
|
Nenasheva VV, Kovaleva GV, Khaidarova NV, Novosadova EV, Manuilova ES, Antonov SA, Tarantul VZ. Trim14 overexpression causes the same transcriptional changes in mouse embryonic stem cells and human HEK293 cells. In Vitro Cell Dev Biol Anim 2013; 50:121-8. [PMID: 24092016 DOI: 10.1007/s11626-013-9683-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 08/05/2013] [Indexed: 12/11/2022]
Abstract
The trim14 (pub, KIAA0129) gene encodes the TRIM14 protein which is a member of the tripartite motif (TRIM) family. Previously, we revealed high expression levels of trim14 in HIV- or SIV-associated lymphomas and demonstrated the influence of trim14 on mesodermal differentiation of mouse embryonic stem cells (mESC). In the present work, to elucidate the role of trim14 in normal and pathological processes in the cell, we used two different types of cells transfected with trim14: mESC and human HEK293. Using subtractive hybridization and real-time PCR, we found a number of genes which expression was elevated in trim14-transfected mESC: hsp90ab1, prr13, pu.1, tnfrsf13c (baff-r), tnfrsf13b (taci), hlx1, hbp1, junb, and pdgfrb. A further analysis of the trim14-transfected mESC at the initial stage of differentiation (embryoid bodies (EB) formation) showed essential changes in the expression of these upregulated genes. The transfection of trim14 into HEK293 also induced an enhanced expression of the several genes upregulated in trim14-transfected mESC (hsp90ab1, prr13, pu.1, tnfrsf13c (baff-r), tnfrsf13b (taci), and hlx1). Summarizing, we found similar genes that participated in trim14-directed processes both in mESC and in HEK293. These results demonstrate the presence of the similar mechanism of trim14 gene action in different types of mammalian cells.
Collapse
Affiliation(s)
- Valentina V Nenasheva
- Department of viral and cellular molecular genetics, Institute of Molecular Genetics, Russian Academy of Sciences, 2 Kurchatov Square, Moscow, 123182, Russia,
| | | | | | | | | | | | | |
Collapse
|
22
|
McKenzie KP, Mayer DC, Aubin JE. Osteogenesis and expression of the bone marrow niche in endothelial cell-depleted HipOPs. J Cell Biochem 2013; 114:1066-73. [PMID: 23161750 DOI: 10.1002/jcb.24446] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 10/30/2012] [Indexed: 12/11/2022]
Abstract
The identification and purification of murine multipotent mesenchymal stem cells (MSCs) have been difficult due to their low frequency, the presence of contaminating cell types and lack of unambiguous markers. Using a magnetic micro-beads negative selection technique to remove hematopoietic cells from mouse bone marrow stromal cells (BMSCs), our lab recently isolated a highly purified osteoprogenitor (HipOP) population that was also enriched for other mesenchymal precursors, including MSCs [Itoh and Aubin, 2009]. We now report that HipOPs are also highly enriched in vascular endothelial cells (VECs), which we hypothesized were an accessory cell type regulating osteogenesis. However, when VECs were immunodepleted from HipOPs with anti-CD31 antibodies, the resulting CD31(-) HipOP population had equal osteogenic capacity to the HipOPs in vitro and in vivo. Analysis of gene expression of Ncad, Pth1r, Ang1, Cxcl12, Jag1, Pdgfr-β, α-sma, Desmin, and Ng2 suggested that both HipOPs and CD31(-) HipOPs are hemopoietic stem cell (HSC) niche populations. However, the data support the view that osteoblast differentiation and depletion of VECs modulate the HSC niche.
Collapse
Affiliation(s)
- Kristen P McKenzie
- Department of Dentistry, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 4245, Toronto, Ontario, Canada M5S 1A8
| | | | | |
Collapse
|
23
|
Kelly KJ, Liu Y, Zhang J, Goswami C, Lin H, Dominguez JH. Comprehensive genomic profiling in diabetic nephropathy reveals the predominance of proinflammatory pathways. Physiol Genomics 2013; 45:710-9. [PMID: 23757392 DOI: 10.1152/physiolgenomics.00028.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Despite advances in the treatment of diabetic nephropathy (DN), currently available therapies have not prevented the epidemic of progressive chronic kidney disease (CKD). The morbidity of CKD, and the inexorable increase in the prevalence of end-stage renal disease, demands more effective approaches to prevent and treat progressive CKD. We undertook next-generation sequencing in a rat model of diabetic nephropathy to study in depth the pathogenic alterations involved in DN with progressive CKD. We employed the obese, diabetic ZS rat, a model that develops diabetic nephropathy, characterized by progressive CKD, inflammation, and fibrosis, the hallmarks of human disease. We then used RNA-seq to examine the combined effects of renal cells and infiltrating inflammatory cells acting as a pathophysiological unit. The comprehensive systems biology analysis of progressive CKD revealed multiple interactions of altered genes that were integrated into morbid networks. These pathological gene assemblies lead to renal inflammation and promote apoptosis and cell cycle arrest in progressive CKD. Moreover, in what is clearly a major therapeutic challenge, multiple and redundant pathways were found to be linked to renal fibrosis, a major cause of kidney loss. We conclude that systems biology applied to progressive CKD in DN can be used to develop novel therapeutic strategies directed to restore critical anomalies in affected gene networks.
Collapse
Affiliation(s)
- K J Kelly
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | | | | | | |
Collapse
|
24
|
Dar A, Itskovitz-Eldor J. Therapeutic potential of perivascular cells from human pluripotent stem cells. J Tissue Eng Regen Med 2013; 9:977-87. [PMID: 23365073 DOI: 10.1002/term.1698] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Revised: 10/17/2012] [Accepted: 12/20/2012] [Indexed: 12/20/2022]
Abstract
Vascularization of injured tissues or artificial grafts is a major challenge in tissue engineering, stimulating a continued search for alternative sources for vasculogenic cells and the development of therapeutic strategies. Human pluripotent stem cells (hPSCs), either embryonic or induced, offer a plentiful platform for the derivation of large numbers of vasculogenic cells, as required for clinical transplantations. Various protocols for generation of vasculogenic smooth muscle cells (SMCs) from hPSCs have been described with considerably different SMC derivatives. In addition, we recently identified hPSC-derived pericytes, which are similar to their physiological counterparts, exhibiting unique features of blood vessel-residing perivascular cells, as well as multipotent mesenchymal precursors with therapeutic angiogenic potential. In this review we refer to methodologies for the development of a variety of perivascular cells from hPSCs with respect to developmental induction, differentiation capabilities, potency and their dual function as mesenchymal precursors. The therapeutic effect of hPSC-derived perivascular cells in experimental models of tissue engineering and regenerative medicine are described and compared to those of their native physiological counterparts.
Collapse
Affiliation(s)
- Ayelet Dar
- Sohnis and Forman Families Stem Cell Centre, the Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Joseph Itskovitz-Eldor
- Sohnis and Forman Families Stem Cell Centre, the Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Department of Obstetrics and Gynaecology, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
25
|
|
26
|
Abstract
BMP9 signaling has been implicated in hereditary hemorrhagic telangiectasia (HHT) and vascular remodeling, acting via the HHT target genes, endoglin and ALK1. This study sought to identify endothelial BMP9-regulated proteins that could affect the HHT phenotype. Gene ontology analysis of cDNA microarray data obtained after BMP9 treatment of primary human endothelial cells indicated regulation of chemokine, adhesion, and inflammation pathways. These responses included the up-regulation of the chemokine CXCL12/SDF1 and down-regulation of its receptor CXCR4. Quantitative mass spectrometry identified additional secreted proteins, including the chemokine CXCL10/IP10. RNA knockdown of endoglin and ALK1 impaired SDF1/CXCR4 regulation by BMP9. Because of the association of SDF1 with ischemia, we analyzed its expression under hypoxia in response to BMP9 in vitro, and during the response to hindlimb ischemia, in endoglin-deficient mice. BMP9 and hypoxia were additive inducers of SDF1 expression. Moreover, the data suggest that endoglin deficiency impaired SDF1 expression in endothelial cells in vivo. Our data implicate BMP9 in regulation of the SDF1/CXCR4 chemokine axis in endothelial cells and point to a role for BMP9 signaling via endoglin in a switch from an SDF1-responsive autocrine phenotype to an SDF1 nonresponsive paracrine state that represses endothelial cell migration and may promote vessel maturation.
Collapse
|
27
|
Bottsford-Miller JN, Coleman RL, Sood AK. Resistance and escape from antiangiogenesis therapy: clinical implications and future strategies. J Clin Oncol 2012; 30:4026-34. [PMID: 23008289 DOI: 10.1200/jco.2012.41.9242] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Angiogenesis has long been considered an important target for cancer therapy. Initial efforts have primarily focused on targeting of endothelial and tumor-derived vascular endothelial growth factor signaling. As evidence emerges that angiogenesis has significant mechanistic complexity, therapeutic resistance and escape have become practical limitations to drug development. Here, we review the mechanisms by which dynamic changes occur in the tumor microenvironment in response to antiangiogenic therapy, leading to drug resistance. These mechanisms include direct selection of clonal cell populations with the capacity to rapidly upregulate alternative proangiogenic pathways, increased invasive capacity, and intrinsic resistance to hypoxia. The implications of normalization of vasculature with subsequently improved vascular function as a result of antiangiogenic therapy are explored, as are the implications of the ability to incorporate and co-opt otherwise normal vasculature. Finally, we consider the extent to which a better understanding of the biology of hypoxia and reoxygenation, as well as the depth and breadth of systems invested in angiogenesis, may offer putative biomarkers and novel therapeutic targets. Insights gained through this work may offer solutions for personalizing antiangiogenesis approaches and improving the outcome of patients with cancer.
Collapse
Affiliation(s)
- Justin N Bottsford-Miller
- Departments of Gynecologic Oncology and Cancer Biology, University of Texas MD Anderson Cancer Center, Unit 1362, PO Box 301439, Houston, TX 77230-1439, USA
| | | | | |
Collapse
|
28
|
Zhu H, Shao Q, Sun X, Deng Z, Yuan X, Zhou X, Ding Y. Bone marrow cells: Important role on neovascularization of hepatocellular carcinoma. J Gastroenterol Hepatol 2012; 27:1241-51. [PMID: 22142567 DOI: 10.1111/j.1440-1746.2011.07044.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIM Present antivascular therapies including embolization to hepatocellular carcinoma (HCC) were not as satisfying as expected. The aim was to explore whether or not bone marrow cells (BMCs) played an important role on neovascularization in HCC. METHODS Bone marrow-GFP(+) orthotropic HCC mice model was used. In controls and HCC mice, the dynamic change of circulating BMCs and serum vascular endothelial growth factor (VEGF), platelet derived growth factor (PDGF) were measured by flow cytometry and enzyme linked immunosorbent assay, respectively. Intrahepatic distribution of BMCs was evaluated using immunofluorescent and realtime polymerase chain reaction protocols. BMCs' intrahepatic differentiation and proportion in vessels was investigated by immunofluorescent methods. Immunohistochemistry and western blotting were performed to examine the expression of adhesion molecule in tumor tissues and tumor free tissues. RESULTS Compared with controls, the frequency of circulating BMCs and serum VEGF, PDGF were much higher in HCC mice. The number of BMCs and the level of CD133 gene in tumor increased significantly relative to the tumor free zone. Since the early stage of HCC, BMCs have been mobilized, recruited into tumor and incorporated into different types of vessels of the liver. Besides into endothelial cells, BMCs also differentiated into vascular fibroblast and hepatic stellate cells. Moreover with tumor growth, the proportion of BMCs in vessels increased gradually. CONCLUSION Mobilized BMCs played an important role in tumor vasculogenesis of HCC. Combined blockading of bone marrow-mediated vasculogenesis may improve the efficacy of current therapy to HCC patients.
Collapse
Affiliation(s)
- Haitao Zhu
- Institute of Hepatobiliary Surgery and Department of Hepatobiliary Surgery, the Affiliated DrumTower Hospital, School of Medicine, Nanjing University, NanJing, JiangSu Province, China
| | | | | | | | | | | | | |
Collapse
|