1
|
Hilage P, Damle MN, Sharma RK, Joshi MG. Melanoma Cell Adhesion Molecule (CD 146) in Endometrial Physiology and Disorder. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024. [PMID: 39400880 DOI: 10.1007/5584_2024_826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
The human endometrium, the innermost lining of the uterus, is the anatomic prerequisite for pregnancy. It is the only dynamic tissue that undergoes more than 400 cycles of regeneration throughout the reproductive life of women. Key to this function are endometrial stem cells as well as cell adhesion molecules. Melanoma cell adhesion molecule (MCAM/CD146/MUC18) is a membrane glycoprotein of the mucin family and a key cell adhesion protein, highly expressed by endometrial cells. CD146 is a significant molecule pivotal in endometrial physiology, assisting tissue regeneration and angiogenesis. Endometrium also acts as a culprit in causing several endometrial dysfunctions, such as endometriosis, endometrial hyperplasia, and endometrial carcinoma, due to interrupted molecular and functional mechanisms. Though most of the endometrial dysfunctions arise as a result of endocrine disturbance, it has a major pathological role associated with angiogenesis. It has already been proven that CD146 is a potential marker for the diagnosis of angiogenic dysfunctions and malignancy, including endometrial cancer. However, its mechanistic role in causing the pathology is a mystery. This chapter explores the role of CD146 in normal and pathological endometrial conditions and the therapeutic implications of CD146.
Collapse
Affiliation(s)
- Priyanka Hilage
- Department of Stem Cells & Regenerative Medicine, D.Y. Patil Education Society (Deemed to be University), Kolhapur, Maharashtra, India
| | - Mrunal N Damle
- Department of Stem Cells & Regenerative Medicine, D.Y. Patil Education Society (Deemed to be University), Kolhapur, Maharashtra, India
| | - Rakesh Kumar Sharma
- Department of Obstetrics and Gynecology, D.Y. Patil Medical College, Kolhapur, Maharashtra, India.
| | - Meghnad G Joshi
- Department of Stem Cells & Regenerative Medicine, D.Y. Patil Education Society (Deemed to be University), Kolhapur, Maharashtra, India
- Stem Plus Biotech Pvt. Ltd, Sangli, Maharashtra, India
| |
Collapse
|
2
|
Guo X, Hu M, Zhang Q, Liu J, Shi J, Tang Y, Liu S, Guo J, Kong Y, Zhu H, Yang Z. Preclinical ImmunoPET Imaging Using a Zr-89-Labeled Anti-CD146 Monoclonal Antibody for Diagnosis of Melanoma. Mol Pharm 2024; 21:4490-4497. [PMID: 39077827 DOI: 10.1021/acs.molpharmaceut.4c00348] [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] [Indexed: 07/31/2024]
Abstract
The aim of this study was to evaluate the preclinical efficacy of [89Zr]Zr-DFO-Ab253 as a novel positron emission tomography (PET) tracer for CD146-positive malignant melanoma imaging. Considering the high expression of CD146 in malignant melanoma, this study investigated the effect of different CD146 expression levels on the tumor uptake of [89Zr]Zr-DFO-Ab253. CD146 selectivity was investigated by using the CD146-positive human melanoma cell A375 and the CD146-negative human alveolar epithelial cell A549. The cell uptake of [89Zr]Zr-DFO-Ab253 tracers was investigated, and receptor-binding affinities were measured by radioactive enzyme-linked immunosorbent assay. Biodistribution studies and micro-PET imaging of the radiotracers were performed on mice bearing A375 and A549 xenografts under baseline and blocking conditions. An immunohistochemical test was performed using A375 and A549 tissue sections for CD146 expression level analysis. [89Zr]Zr-DFO-Ab253 was obtained with a high radiochemical yield (87.86 ± 4.66%) and a satisfactory radiochemical purity (>98.0%). The specificity and affinity of [89Zr]Zr-DFO-Ab253 were confirmed in melanoma A375 cells and in vivo PET imaging of A375 tumor models. [89Zr]Zr-DFO-IgG and A549 lung tumors were prepared as control radiotracers and negative models to verify the specificity of [89Zr]Zr-DFO-Ab253 on CD146. [89Zr]Zr-DFO-Ab253 has a Kd of 4.01 ± 0.50 nM. PET imaging and biodistribution showed a higher uptake of [89Zr]Zr-DFO-Ab253 in A375 melanomas than that in A549 tumors (42.1 ± 4.04% vs 7.87 ± 1.30% ID/g at 120 h, P < 0.05). A low tumor uptake of [89Zr]Zr-DFO-IgG was observed with uptakes of 1.91 ± 0.41 and 2.80 ± 0.14 ID%/g when blocked at 120 h. The radiation-absorbed dose was calculated to be 0.13 mSv/MBq. This study demonstrates the synthesis and preclinical evaluation of [89Zr]Zr-DFO-Ab253 and indicates that the novel tracer has promising applications in malignant melanoma-specific PET imaging because of its high uptake and long-time retention in malignant melanoma. It also provides feasibility for the development of integrated molecular probes for diagnosis and treatment based on the CD146 target.
Collapse
Affiliation(s)
- Xiaoyi Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Muye Hu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Qian Zhang
- Guizhou University Medicine College, Guiyang 550025, Guizhou, China
| | - Jiayue Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jing Shi
- Multitude Therapeutics, 159 Tianzhou Road, Xuhui District, Shanghai 200030, China
| | - Yanfang Tang
- Multitude Therapeutics, 159 Tianzhou Road, Xuhui District, Shanghai 200030, China
| | - ShuHui Liu
- Multitude Therapeutics, 159 Tianzhou Road, Xuhui District, Shanghai 200030, China
| | - Jun Guo
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Yan Kong
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Hua Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhi Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
3
|
Guo Y, Kasai Y, Tanaka Y, Ohashi‐Kumagai Y, Sakamoto T, Ito T, Murakami Y. IGSF3 is a homophilic cell adhesion molecule that drives lung metastasis of melanoma by promoting adhesion to vascular endothelium. Cancer Sci 2024; 115:1936-1947. [PMID: 38590281 PMCID: PMC11145127 DOI: 10.1111/cas.16166] [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: 11/09/2023] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
The immunoglobulin superfamily (IgSF) is one of the largest families of cell-surface molecules involved in various cell-cell interactions, including cancer-stromal interactions. In this study, we undertook a comprehensive RT-PCR-based screening for IgSF molecules that promote experimental lung metastasis in mice. By comparing the expression of 325 genes encoding cell-surface IgSF molecules between mouse melanoma B16 cells and its highly metastatic subline, B16F10 cells, we found that expression of the immunoglobulin superfamily member 3 gene (Igsf3) was significantly enhanced in B16F10 cells than in B16 cells. Knockdown of Igsf3 in B16F10 cells significantly reduced lung metastasis following intravenous injection into C57BL/6 mice. IGSF3 promoted adhesion of B16F10 cells to vascular endothelial cells and functioned as a homophilic cell adhesion molecule between B16F10 cells and vascular endothelial cells. Notably, the knockdown of IGSF3 in either B16F10 cells or vascular endothelial cells suppressed the transendothelial migration of B16F10 cells. Moreover, IGSF3 knockdown suppressed the extravasation of B16F10 cells into the lungs after intravenous injection. These results suggest that IGSF3 promotes the metastatic potential of B16F10 cells in the lungs by facilitating their adhesion to vascular endothelial cells.
Collapse
Affiliation(s)
- Yue Guo
- Division of Molecular PathologyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Yutaka Kasai
- Division of Molecular PathologyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Yuto Tanaka
- Division of Molecular PathologyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Yuki Ohashi‐Kumagai
- Division of Molecular PathologyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Takeharu Sakamoto
- Department of Cancer BiologyInstitute of Biomedical Science, Kansai Medical UniversityHirakataJapan
| | - Takeshi Ito
- Division of Molecular PathologyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Yoshinori Murakami
- Division of Molecular PathologyThe Institute of Medical Science, The University of TokyoTokyoJapan
| |
Collapse
|
4
|
Yang Q, Huang W, Hsu JC, Song L, Sun X, Li C, Cai W, Kang L. CD146-targeted nuclear medicine imaging in cancer: state of the art. VIEW 2023; 4:20220085. [PMID: 38076327 PMCID: PMC10703309 DOI: 10.1002/viw.20220085] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/02/2023] [Indexed: 01/02/2024] Open
Abstract
The transmembrane glycoprotein adhesion molecule CD146 is overexpressed in a wide variety of cancers. Through molecular imaging, a specific biomarker's expression and distribution can be viewed in vivo non-invasively. Radionuclide-labeled monoclonal antibodies or relevant fragments that target CD146 may find potential applications in cancer imaging, thereby offering tremendous value in cancer diagnosis, staging, prognosis evaluation, and prediction of drug resistance. This review discusses the recent developments of CD146-targeted molecular imaging via nuclear medicine, especially in malignant melanoma, brain tumor, lung cancer, liver cancer, breast cancer, and pancreatic cancer. Many studies have proved that CD146 targeting may present a promising strategy for cancer theranostics.
Collapse
Affiliation(s)
- Qi Yang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Wenpeng Huang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Jessica C. Hsu
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States of America
| | - Lele Song
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Xinyao Sun
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Cuicui Li
- Department of Nuclear Medicine, Beijing Friendship Hospital of Capital Medical University, Beijing 100050, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States of America
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
5
|
Lahooti B, Akwii RG, Zahra FT, Sajib MS, Lamprou M, Alobaida A, Lionakis MS, Mattheolabakis G, Mikelis CM. Targeting endothelial permeability in the EPR effect. J Control Release 2023; 361:212-235. [PMID: 37517543 DOI: 10.1016/j.jconrel.2023.07.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/19/2023] [Accepted: 07/23/2023] [Indexed: 08/01/2023]
Abstract
The characteristics of the primary tumor blood vessels and the tumor microenvironment drive the enhanced permeability and retention (EPR) effect, which confers an advantage towards enhanced delivery of anti-cancer nanomedicine and has shown beneficial effects in preclinical models. Increased vascular permeability is a landmark feature of the tumor vessels and an important driver of the EPR. The main focus of this review is the endothelial regulation of vascular permeability. We discuss current challenges of targeting vascular permeability towards clinical translation and summarize the structural components and mechanisms of endothelial permeability, the principal mediators and signaling players, the targeted approaches that have been used and their outcomes to date. We also critically discuss the effects of the tumor-infiltrating immune cells, their interplay with the tumor vessels and the impact of immune responses on nanomedicine delivery, the impact of anti-angiogenic and tumor-stroma targeting approaches, and desirable nanoparticle design approaches for greater translational benefit.
Collapse
Affiliation(s)
- Behnaz Lahooti
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Racheal G Akwii
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Fatema Tuz Zahra
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Md Sanaullah Sajib
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Margarita Lamprou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece
| | - Ahmed Alobaida
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Ha'il 81442, Saudi Arabia
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA.
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece.
| |
Collapse
|
6
|
Long Non-Coding RNAs as Novel Targets for Phytochemicals to Cease Cancer Metastasis. Molecules 2023; 28:molecules28030987. [PMID: 36770654 PMCID: PMC9921150 DOI: 10.3390/molecules28030987] [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: 11/23/2022] [Revised: 12/31/2022] [Accepted: 01/11/2023] [Indexed: 01/21/2023] Open
Abstract
Metastasis is a multi-step phenomenon during cancer development leading to the propagation of cancer cells to distant organ(s). According to estimations, metastasis results in over 90% of cancer-associated death around the globe. Long non-coding RNAs (LncRNAs) are a group of regulatory RNA molecules more than 200 base pairs in length. The main regulatory activity of these molecules is the modulation of gene expression. They have been reported to affect different stages of cancer development including proliferation, apoptosis, migration, invasion, and metastasis. An increasing number of medical data reports indicate the probable function of LncRNAs in the metastatic spread of different cancers. Phytochemical compounds, as the bioactive agents of plants, show several health benefits with a variety of biological activities. Several phytochemicals have been demonstrated to target LncRNAs to defeat cancer. This review article briefly describes the metastasis steps, summarizes data on some well-established LncRNAs with a role in metastasis, and identifies the phytochemicals with an ability to suppress cancer metastasis by targeting LncRNAs.
Collapse
|
7
|
Tsurudome Y, Morita N, Horiguchi M, Ushijima K. Decreased ZO1 expression causes loss of time-dependent tight junction function in the liver of ob/ob mice. Mol Biol Rep 2022; 49:11881-11890. [PMID: 36224445 DOI: 10.1007/s11033-022-07940-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/18/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022]
Abstract
Diabetes patients are at a high risk of developing complications related to angiopathy and disruption of the signal transduction system. The liver is one of the multiple organs damaged during diabetes. Few studies have evaluated the morphological effects of adhesion factors in diabetic liver. The influence of diurnal variation has been observed in the expression and functioning of adhesion molecules to maintain tissue homeostasis associated with nutrient uptake. The present study demonstrated that the rhythm-influenced functioning of tight junction was impaired in the liver of ob/ob mice. The tight junctions of hepatocytes were loosened during the dark period in control mice compared to those in ob/ob mice, where the hepatocyte gaps remained open throughout the day. The time-dependent expression of zonula occludens 1 (ZO1, encoded by Tjp1 gene) in the liver plays a vital role in the functioning of the tight junction. The time-dependent expression of ZO1 was nullified and its expression was attenuated in the liver of ob/ob mice. ZO1 expression was inhibited at the mRNA and protein levels. The expression rhythm of ZO1 was found to be regulated by heat shock factor (HSF)1/2, the expression of which was reduced in the liver of ob/ob mice. The DNA-binding ability of HSF1/2 was decreased in the liver of ob/ob mice compared to that in control mice. These findings suggest the involvement of impaired expression and functioning of adhesion factors in diabetic liver complications.
Collapse
Affiliation(s)
- Yuya Tsurudome
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 756-0884, Yamaguchi, Japan
| | - Nao Morita
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 756-0884, Yamaguchi, Japan
| | - Michiko Horiguchi
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 756-0884, Yamaguchi, Japan
| | - Kentaro Ushijima
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 756-0884, Yamaguchi, Japan. .,Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Tochigi, Japan.
| |
Collapse
|
8
|
Heim X, Bermudez J, Joshkon A, Kaspi E, Bachelier R, Nollet M, Vélier M, Dou L, Brodovitch A, Foucault-Bertaud A, Leroyer AS, Benyamine A, Daumas A, Granel B, Sabatier F, Dignat-George F, Blot-Chabaud M, Bardin N. CD146 at the Interface between Oxidative Stress and the Wnt Signaling Pathway in Systemic Sclerosis. J Invest Dermatol 2022; 142:3200-3210.e5. [PMID: 35690141 DOI: 10.1016/j.jid.2022.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 03/01/2022] [Accepted: 03/12/2022] [Indexed: 01/05/2023]
Abstract
CD146 involvement was recently described in skin fibrosis of systemic sclerosis through its regulation of the Wnt pathway. Because the interaction between Wnt and ROS signaling plays a major role in fibrosis, we hypothesized that in systemic sclerosis, CD146 may regulate Wnt/ROS crosstalk. Using a transcriptomic and western blot analysis performed on CD146 wild-type or knockout mouse embryonic fibroblasts, we showed a procanonical Wnt hallmark in the absence of CD146 that is reversed when CD146 expression is restored. We found an elevated ROS content in knockout cells and an increase in DNA oxidative damage in the skin sections of knockout mice compared with those of wild-type mice. We also showed that ROS increased CD146 and its noncanonical Wnt ligand, WNT5A, only in wild-type cells. In humans, fibroblasts from patients with systemic sclerosis presented higher ROS content and expressed CD146, whereas control fibroblasts did not. Moreover, CD146 and its ligand were upregulated by ROS in both human fibroblasts. The increase in bleomycin-induced WNT5A expression was abrogated when CD146 was silenced. We showed an interplay between Wnt and ROS signaling in systemic sclerosis, regulated by CD146, which promotes the noncanonical Wnt pathway and prevents ROS signaling, opening the way for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Xavier Heim
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France; Service d'immunologie, Biogénopôle, Hôpital de la Timone, Assistance Publique des Hôpitaux de Marseille (AP-HM), Marseille, France.
| | | | - Ahmad Joshkon
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
| | - Elise Kaspi
- Aix Marseille University, APHM, INSERM, MMG, Hôpital la Timone, Service de Biologie Cellulaire, Marseille, France
| | | | - Marie Nollet
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
| | - Mélanie Vélier
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France; Hematology and Vascular Biology Department, Hopital de la Conception, Assistance Publique des Hôpitaux de Marseille (AP-HM), Marseille, France
| | - Laetitia Dou
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
| | - Alexandre Brodovitch
- Service d'immunologie, Biogénopôle, Hôpital de la Timone, Assistance Publique des Hôpitaux de Marseille (AP-HM), Marseille, France
| | | | | | - Audrey Benyamine
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France; Internal Medicine Department, Hopital Nord, Assistance Publique des Hôpitaux de Marseille (AP-HM), Marseille, France
| | - Aurélie Daumas
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France; Internal Medicine, Geriatric and Therapeutic Department, Hopital de la Timone, Assistance Publique des Hôpitaux de Marseille (AP-HM), Marseille, France
| | - Brigitte Granel
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France; Internal Medicine Department, Hopital Nord, Assistance Publique des Hôpitaux de Marseille (AP-HM), Marseille, France
| | - Florence Sabatier
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France; Cell Therapy Laboratory, INSERM CIC BT 1409, Hôpital de la Conception, Assistance Publique des Hôpitaux de Marseille (AP-HM), Marseille, France
| | - Françoise Dignat-George
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France; Hematology and Vascular Biology Department, Hopital de la Conception, Assistance Publique des Hôpitaux de Marseille (AP-HM), Marseille, France
| | | | - Nathalie Bardin
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France; Service d'immunologie, Biogénopôle, Hôpital de la Timone, Assistance Publique des Hôpitaux de Marseille (AP-HM), Marseille, France
| |
Collapse
|
9
|
Yang Y, Cao Y. The impact of VEGF on cancer metastasis and systemic disease. Semin Cancer Biol 2022; 86:251-261. [PMID: 35307547 DOI: 10.1016/j.semcancer.2022.03.011] [Citation(s) in RCA: 105] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 01/27/2023]
Abstract
Metastasis is the leading cause of cancer-associated mortality and the underlying mechanisms of cancer metastasis remain elusive. Both blood and lymphatic vasculatures are essential structures for mediating distal metastasis. The vasculature plays multiple functions, including accelerating tumor growth, sustaining the tumor microenvironment, supplying growth and invasive signals, promoting metastasis, and causing cancer-associated systemic disease. VEGF is one of the key angiogenic factors in tumors and participates in the initial stage of tumor development, progression and metastasis. Consequently, VEGF and its receptor-mediated signaling pathways have become one of the most important therapeutic targets for treating various cancers. Today, anti-VEGF-based antiangiogenic drugs (AADs) are widely used in the clinic for treating different types of cancer in human patients. Despite nearly 20-year clinical experience with AADs, the impact of these drugs on cancer metastasis and systemic disease remains largely unknown. In this review article, we focus our discussion on tumor VEGF in cancer metastasis and systemic disease and mechanisms underlying AADs in clinical benefits.
Collapse
Affiliation(s)
- Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, Karolinska Institute, 171 77 Stockholm, Sweden.
| |
Collapse
|
10
|
Galectin-3 Is a Natural Binding Ligand of MCAM (CD146, MUC18) in Melanoma Cells and Their Interaction Promotes Melanoma Progression. Biomolecules 2022; 12:biom12101451. [PMID: 36291660 PMCID: PMC9599063 DOI: 10.3390/biom12101451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Melanoma cell adhesion molecule (MCAM, CD146, MUC18) is a heavily glycosylated transmembrane protein and a marker of melanoma metastasis. It is expressed in advanced primary melanoma and metastasis but rarely in benign naevi or normal melanocytes. More and more evidence has shown that activation of the MCAM on cell surface plays a vital role in melanoma progression and metastasis. However, the natural MCAM binding ligand that initiates MCAM activation in melanoma so far remains elusive. This study revealed that galectin-3, a galactoside-binding protein that is commonly overexpressed in many cancers including melanoma, is naturally associated with MCAM on the surface of both skin and uveal melanoma cells. Binding of galectin-3 to MCAM, via O-linked glycans on the MCAM, induces MCAM dimerization and clustering on cell surface and subsequent activation of downstream AKT signalling. This leads to the increases of a number of important steps in melanoma progression of cell proliferation, adhesion, migration, and invasion. Thus, galectin-3 is a natural binding ligand of MCAM in melanoma, and their interaction activates MCAM and promotes MCAM-mediated melanoma progression. Targeting the galectin-3–MCAM interaction may potentially be a useful therapeutic strategy for melanoma treatment.
Collapse
|
11
|
Zhang ZY, Zhai C, Yang XY, Li HB, Wu LL, Li L. Knockdown of CD146 promotes endothelial-to-mesenchymal transition via Wnt/β-catenin pathway. PLoS One 2022; 17:e0273542. [PMID: 36001597 PMCID: PMC9401105 DOI: 10.1371/journal.pone.0273542] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 08/10/2022] [Indexed: 11/27/2022] Open
Abstract
Purpose Cardiac fibrosis is characterized by the excessive deposition of extracellular matrix (ECM) proteins and leads to the maladaptive changes in myocardium. Endothelial cells (ECs) undergoing mesenchymal transition contributes to the occurrence and development of cardiac fibrosis. CD146 is an adhesion molecule highly expressed in ECs. The present study was performed to explore the role of CD146 in modulating endothelial to mesenchymal transition (EndMT). Methods C57BL/6 mice were subjected to subcutaneous implantation of osmotic minipump infused with angiotensin II (Ang Ⅱ). Adenovirus carrying CD146 short hairpin RNA (shRNA) or CD146 encoding sequence were infected into cultured human umbilical vein endothelial cells (HUVECs) followed by stimulation with Ang II or transforming growth factor-β1 (TGF-β1). Differentially expressed genes were revealed by RNA-sequencing (RNA-Seq) analysis. Gene expression was measured by quantitative real-time PCR, and protein expression and distribution were determined by Western blot and immunofluorescence staining, respectively. Results CD146 was predominantly expressed by ECs in normal mouse hearts. CD146 was upregulated in ECs but not fibroblasts and myocytes in hearts of Ang II-infused mice and in HUVECs stimulated with Ang Ⅱ. RNA-Seq analysis revealed the differentially expressed genes related to EndMT and Wnt/β-catenin signaling pathway. CD146 knockdown and overexpression facilitated and attenuated, respectively, EndMT induced by Ang II or TGF-β1. CD146 knockdown upregulated Wnt pathway-related genes including Wnt4, LEF1, HNF4A, FOXA1, SOX6, and CCND3, and increased the protein level and nuclear translocation of β-catenin. Conclusions Knockdown of CD146 exerts promotional effects on EndMT via activating Wnt/β-catenin pathway and the upregulation of CD146 might play a protective role against EndMT and cardiac fibrosis.
Collapse
Affiliation(s)
- Zhao-Yu Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Chao Zhai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Xue-Yuan Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Hai-Bing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Li-Ling Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Li Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
- * E-mail:
| |
Collapse
|
12
|
Lin J, Cui K, Xu Y, Tang X, Shi Y, Lu X, Yang B, He Q, Yu S, Liang X. Inhibition of CD146 attenuates retinal neovascularization via vascular endothelial growth factor receptor 2 signalling pathway in proliferative diabetic retinopathy. Acta Ophthalmol 2022; 100:e899-e911. [PMID: 34477295 DOI: 10.1111/aos.15007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 06/24/2021] [Accepted: 08/04/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE To investigate the expression of CD146 and its role in proliferative diabetic retinopathy (PDR). METHODS Enzyme linked immunosorbent assay was performed to analyse the expression and relationship of sCD146, vascular endothelial growth factor (VEGF), sVEGFR1 and sVEGFR2 in vitreous specimens from PDR and idiopathic epiretinal membranes (IERM) or idiopathic macular hole patients. The location of CD146 in ERMs was detected by immunofluorescence. The oxygen-induced retinopathy (OIR) mice model was established and the adeno-associated virus expressing a shRNA of CD146 (AAV1-shCD146-GFP) was administered via intravitreal injection. The effect of AAV1-shCD146-GFP was explored by immunofluorescence, Western blot and quantitative real-time PCR. RESULTS The levels of sCD146 in vitreous specimens from PDR patients and CD146 in retinas from OIR mice were significantly increased. Immunofluorescence showed that CD146 was co-located with CD31, VEGF, VEGFR1 and VEGFR2, respectively. Intravitreal injection of AAV1-shCD146-GFP could dramatically reduce the formation of neovascularization and non-perfusion area by inhibiting VEGFR2 phosphorylation. CONCLUSION Our results indicated that CD146 was involved in the development of retinal neovascularization via VEGFR2 pathway. Anti-CD146 may be an innovative or adjuvant therapy, which provides a new direction for the treatment of PDR and other ocular neovascular diseases.
Collapse
Affiliation(s)
- Jianqiang Lin
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Kaixuan Cui
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Yue Xu
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Xiaoyu Tang
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Yuxun Shi
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Xi Lu
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Boyu Yang
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Qingjing He
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Shanshan Yu
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| |
Collapse
|
13
|
Novel treatment strategy for NRAS-mutated melanoma through a selective inhibitor of CD147/VEGFR-2 interaction. Oncogene 2022; 41:2254-2264. [PMID: 35217792 DOI: 10.1038/s41388-022-02244-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 01/28/2022] [Accepted: 02/11/2022] [Indexed: 11/09/2022]
Abstract
More than 70% of human NRASmut melanomas are resistant to MEK inhibitors highlighting the crucial need for efficient therapeutic strategies for these tumors. CD147, a membrane receptor, is overexpressed in most cancers including melanoma and is associated with poor prognosis. We show here that CD147i, a specific inhibitor of CD147/VEGFR-2 interaction represents a potential therapeutic strategy for NRASmut melanoma cells. It significantly inhibited the malignant properties of NRASmut melanomas ex vivo and in vivo. Importantly, NRASmut patient's-derived xenografts, which were resistant to MEKi, became sensitive when combined with CD147i leading to decreased proliferation ex vivo and tumor regression in vivo. Mechanistic studies revealed that CD147i effects were mediated through STAT3 pathway. These data bring a proof of concept on the impact of the inhibition of CD147/VEGFR-2 interaction on melanoma progression and represents a new therapeutic opportunity for NRASmut melanoma when combined with MEKi.
Collapse
|
14
|
Abstract
Melanoma is the most lethal skin cancer that originates from the malignant transformation of melanocytes. Although melanoma has long been regarded as a cancerous malignancy with few therapeutic options, increased biological understanding and unprecedented innovations in therapies targeting mutated driver genes and immune checkpoints have substantially improved the prognosis of patients. However, the low response rate and inevitable occurrence of resistance to currently available targeted therapies have posed the obstacle in the path of melanoma management to obtain further amelioration. Therefore, it is necessary to understand the mechanisms underlying melanoma pathogenesis more comprehensively, which might lead to more substantial progress in therapeutic approaches and expand clinical options for melanoma therapy. In this review, we firstly make a brief introduction to melanoma epidemiology, clinical subtypes, risk factors, and current therapies. Then, the signal pathways orchestrating melanoma pathogenesis, including genetic mutations, key transcriptional regulators, epigenetic dysregulations, metabolic reprogramming, crucial metastasis-related signals, tumor-promoting inflammatory pathways, and pro-angiogenic factors, have been systemically reviewed and discussed. Subsequently, we outline current progresses in therapies targeting mutated driver genes and immune checkpoints, as well as the mechanisms underlying the treatment resistance. Finally, the prospects and challenges in the development of melanoma therapy, especially immunotherapy and related ongoing clinical trials, are summarized and discussed.
Collapse
Affiliation(s)
- Weinan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 of West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Huina Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 of West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 of West Changle Road, 710032, Xi'an, Shaanxi, China.
| |
Collapse
|
15
|
Xiao ST, Kuang CY. Endothelial progenitor cells and coronary artery disease: Current concepts and future research directions. World J Clin Cases 2021; 9:8953-8966. [PMID: 34786379 PMCID: PMC8567528 DOI: 10.12998/wjcc.v9.i30.8953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/12/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023] Open
Abstract
Vascular injury is a frequent pathology in coronary artery disease. To repair the vasculature, scientists have found that endothelial progenitor cells (EPCs) have excellent properties associated with angiogenesis. Over time, research on EPCs has made encouraging progress regardless of pathology or clinical technology. This review focuses on the origins and cell markers of EPCs, and the connection between EPCs and coronary artery disease. In addition, we summarized various studies of EPC-capturing stents and EPC infusion therapy, and aim to learn from past technology to predict the future.
Collapse
Affiliation(s)
- Sen-Tong Xiao
- Department of Cardiovascular Diseases, People’s Hospital Affiliated to Guizhou Medical University, Guiyang 550003, Guizhou Province, China
| | - Chun-Yan Kuang
- Department of Cardiovascular Diseases, Guizhou Provincial People's Hospital, Guiyang 550003, Guizhou Province, China
| |
Collapse
|
16
|
Obu S, Umeda K, Ueno H, Sonoda M, Tasaka K, Ogata H, Kouzuki K, Nodomi S, Saida S, Kato I, Hiramatsu H, Okamoto T, Ogawa E, Okajima H, Morita K, Kamikubo Y, Kawaguchi K, Watanabe K, Iwafuchi H, Yagyu S, Iehara T, Hosoi H, Nakahata T, Adachi S, Uemoto S, Heike T, Takita J. CD146 is a potential immunotarget for neuroblastoma. Cancer Sci 2021; 112:4617-4626. [PMID: 34464480 PMCID: PMC8586675 DOI: 10.1111/cas.15124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/13/2021] [Accepted: 08/26/2021] [Indexed: 11/28/2022] Open
Abstract
Neuroblastoma, the most common extracranial solid tumor of childhood, is thought to arise from neural crest‐derived immature cells. The prognosis of patients with high‐risk or recurrent/refractory neuroblastoma remains quite poor despite intensive multimodality therapy; therefore, novel therapeutic interventions are required. We examined the expression of a cell adhesion molecule CD146 (melanoma cell adhesion molecule [MCAM]) by neuroblastoma cell lines and in clinical samples and investigated the anti‐tumor effects of CD146‐targeting treatment for neuroblastoma cells both in vitro and in vivo. CD146 is expressed by 4 cell lines and by most of primary tumors at any stage. Short hairpin RNA‐mediated knockdown of CD146, or treatment with an anti‐CD146 polyclonal antibody, effectively inhibited growth of neuroblastoma cells both in vitro and in vivo, principally due to increased apoptosis via the focal adhesion kinase and/or nuclear factor‐kappa B signaling pathway. Furthermore, the anti‐CD146 polyclonal antibody markedly inhibited tumor growth in immunodeficient mice inoculated with primary neuroblastoma cells. In conclusion, CD146 represents a promising therapeutic target for neuroblastoma.
Collapse
Affiliation(s)
- Satoshi Obu
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Katsutsugu Umeda
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroo Ueno
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mari Sonoda
- Department of Pediatric Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keiji Tasaka
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideto Ogata
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kagehiro Kouzuki
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Seishiro Nodomi
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Saida
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Itaru Kato
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidefumi Hiramatsu
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tatsuya Okamoto
- Department of Pediatric Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Eri Ogawa
- Department of Pediatric Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideaki Okajima
- Department of Pediatric Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Pediatric Surgery, Kanazawa Medical University, Ishikawa, Japan
| | - Ken Morita
- Department of Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuhiko Kamikubo
- Department of Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koji Kawaguchi
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Kenichiro Watanabe
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Hideto Iwafuchi
- Department of Pathology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Shigeki Yagyu
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomoko Iehara
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hajime Hosoi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tatsutoshi Nakahata
- Drug Discovery Technology Development Office, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Souichi Adachi
- Department of Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinji Uemoto
- Department of Pediatric Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshio Heike
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junko Takita
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
17
|
Abu El-Asrar AM, Nawaz MI, Ahmad A, Siddiquei MM, Allegaert E, Gikandi PW, De Hertogh G, Opdenakker G. CD146/Soluble CD146 Pathway Is a Novel Biomarker of Angiogenesis and Inflammation in Proliferative Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2021; 62:32. [PMID: 34293080 PMCID: PMC8300056 DOI: 10.1167/iovs.62.9.32] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose Inflammation, angiogenesis and fibrosis are pathological hallmarks of proliferative diabetic retinopathy (PDR). The CD146/sCD146 pathway displays proinflammatory and proangiogenic properties. We investigated the role of this pathway in the pathophysiology of PDR. Methods Vitreous samples from 41 PDR and 27 nondiabetic patients, epiretinal fibrovascular membranes from 18 PDR patients, rat retinas, human retinal microvascular endothelial cells (HRMECs) and human retinal Müller glial cells were studied by ELISA, Western blot analysis, immunohistochemistry and immunofluorescence microscopy analysis. Blood-retinal barrier breakdown was assessed with fluorescein isothiocyanate-conjugated dextran. Results sCD146 and VEGF levels were significantly higher in vitreous samples from PDR patients than in nondiabetic patients. In epiretinal membranes, immunohistochemical analysis revealed CD146 expression in leukocytes, vascular endothelial cells and myofibroblasts. Significant positive correlations were detected between numbers of blood vessels expressing CD31, reflecting angiogenic activity of PDR, and numbers of blood vessels and stromal cells expressing CD146. Western blot analysis showed significant increase of CD146 in diabetic rat retinas. sCD146 induced upregulation of phospho-ERK1/2, NF-κB, VEGF and MMP-9 in Müller cells. The hypoxia mimetic agent cobalt chloride, VEGF and TNF-α induced upregulation of sCD146 in HRMECs. The MMP inhibitor ONO-4817 attenuated TNF-α-induced upregulation of sCD146 in HRMECs. Intravitreal administration of sCD146 in normal rats significantly increased retinal vascular permeability and induced significant upregulation of phospho-ERK1/2, intercellular adhesion molecule-1 and VEGF in the retina. sCD146 induced migration of HRMECs. Conclusions These results suggest that the CD146/sCD146 pathway is involved in the initiation and progression of PDR.
Collapse
Affiliation(s)
- Ahmed M Abu El-Asrar
- Department of Ophthalmology, College of Medicine and Medical City, King Saud University, Riyadh, Saudi Arabia.,Dr. Nasser Al-Rashid Research Chair in Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mohd Imtiaz Nawaz
- Department of Ophthalmology, College of Medicine and Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Ajmal Ahmad
- Department of Ophthalmology, College of Medicine and Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad Mairaj Siddiquei
- Department of Ophthalmology, College of Medicine and Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Eef Allegaert
- Laboratory of Histochemistry and Cytochemistry, University of Leuven (KU Leuven), Leuven, Belgium.,University Hospitals, UZ Gasthuisberg, Leuven, Belgium
| | - Priscilla W Gikandi
- Department of Ophthalmology, College of Medicine and Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Gert De Hertogh
- Laboratory of Histochemistry and Cytochemistry, University of Leuven (KU Leuven), Leuven, Belgium.,University Hospitals, UZ Gasthuisberg, Leuven, Belgium
| | - Ghislain Opdenakker
- University Hospitals, UZ Gasthuisberg, Leuven, Belgium.,Department of Microbiology and Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven (KU Leuven), Leuven, Belgium
| |
Collapse
|
18
|
Abed A, Leroyer AS, Kavvadas P, Authier F, Bachelier R, Foucault-Bertaud A, Bardin N, Cohen CD, Lindenmeyer MT, Genest M, Joshkon A, Jourde-Chiche N, Burtey S, Blot-Chabaud M, Dignat-George F, Chadjichristos CE. Endothelial-Specific Deletion of CD146 Protects Against Experimental Glomerulonephritis in Mice. Hypertension 2021; 77:1260-1272. [PMID: 33689459 DOI: 10.1161/hypertensionaha.119.14176] [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] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Ahmed Abed
- From the INSERM UMR-S1155, Tenon Hospital, Paris, France (A.A., P.K., F.A., M.G., C.E.C.).,Sorbonne Université, Paris, France (A.A., C.E.C.)
| | - Aurélie S Leroyer
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Panagiotis Kavvadas
- From the INSERM UMR-S1155, Tenon Hospital, Paris, France (A.A., P.K., F.A., M.G., C.E.C.)
| | - Florence Authier
- From the INSERM UMR-S1155, Tenon Hospital, Paris, France (A.A., P.K., F.A., M.G., C.E.C.)
| | - Richard Bachelier
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Alexandrine Foucault-Bertaud
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Nathalie Bardin
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Clemens D Cohen
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Germany (C.D.C.)
| | - Maja T Lindenmeyer
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany (M.T.L.)
| | - Magali Genest
- From the INSERM UMR-S1155, Tenon Hospital, Paris, France (A.A., P.K., F.A., M.G., C.E.C.)
| | - Ahmad Joshkon
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Noémie Jourde-Chiche
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.).,Department of Nephrology, Aix-Marseille University, AP-HM Hôpital de la Conception, Marseille, France (N.J.-C., S.B.)
| | - Stéphane Burtey
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.).,Department of Nephrology, Aix-Marseille University, AP-HM Hôpital de la Conception, Marseille, France (N.J.-C., S.B.)
| | - Marcel Blot-Chabaud
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Françoise Dignat-George
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Christos E Chadjichristos
- From the INSERM UMR-S1155, Tenon Hospital, Paris, France (A.A., P.K., F.A., M.G., C.E.C.).,Sorbonne Université, Paris, France (A.A., C.E.C.)
| |
Collapse
|
19
|
Stock C. Circulating Tumor Cells: Does Ion Transport Contribute to Intravascular Survival, Adhesion, Extravasation, and Metastatic Organotropism? Rev Physiol Biochem Pharmacol 2021; 182:139-175. [DOI: 10.1007/112_2021_68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
20
|
Feng R, Wang Y, Ramachandran V, Ma Q, May MM, Li M, Zhou JX, Xu X, Xu K, Fang S, Xia W, Sui D, Liu H, Gao X, Prieto V, Blacklow SC, Lu M, Lee JE. Characterization of novel neutralizing mouse monoclonal antibody JM1-24-3 developed against MUC18 in metastatic melanoma. J Exp Clin Cancer Res 2020; 39:273. [PMID: 33278894 PMCID: PMC7718695 DOI: 10.1186/s13046-020-01722-8] [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: 05/08/2020] [Accepted: 09/28/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND MUC18 is a glycoprotein highly expressed on the surface of melanoma and other cancers which promotes tumor progression and metastasis. However, its mechanism of action and suitability as a therapeutic target are unknown. METHODS A monoclonal antibody (mAb) (JM1-24-3) was generated from metastatic melanoma tumor live cell immunization, and high-throughput screening identified MUC18 as the target. RESULTS Analysis of molecular interactions between MUC18 and JM1-24-3 revealed that the downstream signaling events depended on binding of the mAb to a conformational epitope on the extracellular domain of MUC18. JM1-24-3 inhibited melanoma cell proliferation, migration and invasion in vitro and reduced tumor growth and metastasis in vivo. CONCLUSION These results confirm that MUC18 is mechanistically important in melanoma growth and metastasis, suggest that the MUC18 epitope identified is a promising therapeutic target, and that the JM1-24-3 mAb may serve as the basis for a potential therapeutic agent.
Collapse
Affiliation(s)
- Runhua Feng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuling Wang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Vijaya Ramachandran
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | | | - Matthew M May
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,Department of Otolaryngology, Mayo School of Graduate Medical Education, Rochester, MN, 55905, USA
| | - Ming Li
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,Department of Pathology, Anhui Province Hospital, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Joe X Zhou
- LC Sciences, LLC, Houston, TX, 77054, USA
| | - Xiang Xu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard University School of Medicine, Boston, MA, 02115, USA.,Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kejing Xu
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Shenying Fang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Weiya Xia
- Department of Molecular & Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Dawen Sui
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Huey Liu
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xiaolian Gao
- Department of Biology and Biochemistry, The University of Houston, Houston, TX, 77204, USA
| | - Victor Prieto
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Harvard University School of Medicine, Boston, MA, 02115, USA
| | - Mason Lu
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA. .,MedAbome, Inc., Fremont, CA, 94538, USA.
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
21
|
Choi S, Ferrari G, Tedesco FS. Cellular dynamics of myogenic cell migration: molecular mechanisms and implications for skeletal muscle cell therapies. EMBO Mol Med 2020; 12:e12357. [PMID: 33210465 PMCID: PMC7721365 DOI: 10.15252/emmm.202012357] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/02/2020] [Accepted: 08/28/2020] [Indexed: 12/14/2022] Open
Abstract
Directional cell migration is a critical process underlying morphogenesis and post-natal tissue regeneration. During embryonic myogenesis, migration of skeletal myogenic progenitors is essential to generate the anlagen of limbs, diaphragm and tongue, whereas in post-natal skeletal muscles, migration of muscle satellite (stem) cells towards regions of injury is necessary for repair and regeneration of muscle fibres. Additionally, safe and efficient migration of transplanted cells is critical in cell therapies, both allogeneic and autologous. Although various myogenic cell types have been administered intramuscularly or intravascularly, functional restoration has not been achieved yet in patients with degenerative diseases affecting multiple large muscles. One of the key reasons for this negative outcome is the limited migration of donor cells, which hinders the overall cell engraftment potential. Here, we review mechanisms of myogenic stem/progenitor cell migration during skeletal muscle development and post-natal regeneration. Furthermore, strategies utilised to improve migratory capacity of myogenic cells are examined in order to identify potential treatments that may be applied to future transplantation protocols.
Collapse
Affiliation(s)
- SungWoo Choi
- Department of Cell and Developmental Biology, University College London, London, UK.,The Francis Crick Institute, London, UK
| | - Giulia Ferrari
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Francesco Saverio Tedesco
- Department of Cell and Developmental Biology, University College London, London, UK.,The Francis Crick Institute, London, UK.,Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
22
|
Prevention of Melanoma Extravasation as a New Treatment Option Exemplified by p38/MK2 Inhibition. Int J Mol Sci 2020; 21:ijms21218344. [PMID: 33172202 PMCID: PMC7664432 DOI: 10.3390/ijms21218344] [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: 10/19/2020] [Revised: 11/02/2020] [Accepted: 11/02/2020] [Indexed: 01/01/2023] Open
Abstract
Melanoma releases numerous tumor cells into the circulation; however, only a very small fraction of these cells is able to establish distant metastasis. Intravascular survival of circulating tumor cells is limited through hemodynamic forces and by the lack of matrix interactions. The extravasation step is, thus, of unique importance to establish metastasis. Similar to leukocyte extravasation, this process is under the control of adhesion molecule pairs expressed on melanoma and endothelial cells, and as for leukocytes, ligands need to be adequately presented on cell surfaces. Based on melanoma plasticity, there is considerable heterogeneity even within one tumor and one patient resulting in a mixture of invasive or proliferative cells. The molecular control for this switch is still ill-defined. Recently, the balance between two kinase pathways, p38 and JNK, has been shown to determine growth characteristics of melanoma. While an active JNK pathway induces a proliferative phenotype with reduced invasive features, an active p38/MK2 pathway results in an invasive phenotype and supports the extravasation step via the expression of molecules capable of binding to endothelial integrins. Therapeutic targeting of MK2 to prevent extravasation might reduce metastatic spread.
Collapse
|
23
|
Kodet O, Kučera J, Strnadová K, Dvořánková B, Štork J, Lacina L, Smetana K. Cutaneous melanoma dissemination is dependent on the malignant cell properties and factors of intercellular crosstalk in the cancer microenvironment (Review). Int J Oncol 2020; 57:619-630. [PMID: 32705148 PMCID: PMC7384852 DOI: 10.3892/ijo.2020.5090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 06/15/2020] [Indexed: 12/17/2022] Open
Abstract
The incidence of cutaneous malignant melanoma has been steadily increasing worldwide for several decades. This phenomenon seems to follow the trend observed in many types of malignancies caused by multiple significant factors, including ageing. Despite the progress in cutaneous malignant melanoma therapeutic options, the curability of advanced disease after metastasis represents a serious challenge for further research. In this review, we summarise data on the microenvironment of cutaneous malignant melanoma with emphasis on intercellular signalling during the disease progression. Malignant melanocytes with features of neural crest stem cells interact with non‑malignant populations within this microenvironment. We focus on representative bioactive factors regulating this intercellular crosstalk. We describe the possible key factors and signalling cascades responsible for the high complexity of the melanoma microenvironment and its premetastatic niches. Furthermore, we present the concept of melanoma early becoming a systemic disease. This systemic effect is presented as a background for the new horizons in the therapy of cutaneous melanoma.
Collapse
Affiliation(s)
- Ondřej Kodet
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague 2
- Department of Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague
- Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec (BIOCEV), First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
| | - Jan Kučera
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague 2
- Department of Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague
| | - Karolína Strnadová
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague 2
- Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec (BIOCEV), First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
| | - Barbora Dvořánková
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague 2
- Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec (BIOCEV), First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
| | - Jiří Štork
- Department of Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague
| | - Lukáš Lacina
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague 2
- Department of Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague
- Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec (BIOCEV), First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
| | - Karel Smetana
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague 2
- Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec (BIOCEV), First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
| |
Collapse
|
24
|
Wang Z, Xu Q, Zhang N, Du X, Xu G, Yan X. CD146, from a melanoma cell adhesion molecule to a signaling receptor. Signal Transduct Target Ther 2020; 5:148. [PMID: 32782280 PMCID: PMC7421905 DOI: 10.1038/s41392-020-00259-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 06/14/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022] Open
Abstract
CD146 was originally identified as a melanoma cell adhesion molecule (MCAM) and highly expressed in many tumors and endothelial cells. However, the evidence that CD146 acts as an adhesion molecule to mediate a homophilic adhesion through the direct interactions between CD146 and itself is still lacking. Recent evidence revealed that CD146 is not merely an adhesion molecule, but also a cellular surface receptor of miscellaneous ligands, including some growth factors and extracellular matrixes. Through the bidirectional interactions with its ligands, CD146 is actively involved in numerous physiological and pathological processes of cells. Overexpression of CD146 can be observed in most of malignancies and is implicated in nearly every step of the development and progression of cancers, especially vascular and lymphatic metastasis. Thus, immunotherapy against CD146 would provide a promising strategy to inhibit metastasis, which accounts for the majority of cancer-associated deaths. Therefore, to deepen the understanding of CD146, we review the reports describing the newly identified ligands of CD146 and discuss the implications of these findings in establishing novel strategies for cancer therapy.
Collapse
Affiliation(s)
- Zhaoqing Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China.
| | - Qingji Xu
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
- College of Life Science, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Nengwei Zhang
- Department of Gastrointestinal Hepatobiliary Tumor Surgery, Beijing Shijitan Hospital, Capital Medical University, 100038, Beijing, China
| | - Xuemei Du
- Departments of Pathology, Beijing Shijitan Hospital, Capital Medical University, 100038, Beijing, China
| | - Guangzhong Xu
- Department of Gastrointestinal Hepatobiliary Tumor Surgery, Beijing Shijitan Hospital, Capital Medical University, 100038, Beijing, China
| | - Xiyun Yan
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China.
- College of Life Science, University of Chinese Academy of Sciences, 100049, Beijing, China.
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
25
|
Leroyer AS, Blin MG, Bachelier R, Bardin N, Blot-Chabaud M, Dignat-George F. CD146 (Cluster of Differentiation 146). Arterioscler Thromb Vasc Biol 2020; 39:1026-1033. [PMID: 31070478 DOI: 10.1161/atvbaha.119.312653] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
CD146 (cluster of differentiation 146) is an adhesion molecule that is expressed by different cells constituting vessels, particularly endothelial cells. The last 30 years of research in this field have shown that CD146 plays a key role in the control of several vessel functions. Three forms of CD146 have been described, including 2 transmembrane isoforms and a soluble protein that is detectable in the plasma. These CD146 forms mediate pleiotropic functions through homophilic and heterophilic interactions with proteins present on surrounding partners. Several studies used neutralizing antibodies, siRNA, or genetically modified mice to demonstrate the involvement of CD146 in the regulation of angiogenesis, vascular permeability, and leukocyte transmigration. In this review, we will focus on the current knowledge of the roles of CD146 in vascular homeostasis and diseases associated with endothelial dysfunction.
Collapse
Affiliation(s)
- Aurélie S Leroyer
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.)
| | - Muriel G Blin
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.)
| | - Richard Bachelier
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.)
| | - Nathalie Bardin
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.).,Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, France (N.B., F.D.-G.)
| | - Marcel Blot-Chabaud
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.)
| | - Françoise Dignat-George
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.).,Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, France (N.B., F.D.-G.)
| |
Collapse
|
26
|
Fibronectin in Cancer: Friend or Foe. Cells 2019; 9:cells9010027. [PMID: 31861892 PMCID: PMC7016990 DOI: 10.3390/cells9010027] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/13/2019] [Accepted: 12/18/2019] [Indexed: 01/10/2023] Open
Abstract
The role of fibronectin (FN) in tumorigenesis and malignant progression has been highly controversial. Cancerous FN plays a tumor-suppressive role, whereas it is pro-metastatic and associated with poor prognosis. Interestingly, FN matrix deposited in the tumor microenvironments (TMEs) promotes tumor progression but is paradoxically related to a better prognosis. Here, we justify how FN impacts tumor transformation and subsequently metastatic progression. Next, we try to reconcile and rationalize the seemingly conflicting roles of FN in cancer and TMEs. Finally, we propose future perspectives for potential FN-based therapeutic strategies.
Collapse
|
27
|
Wettschureck N, Strilic B, Offermanns S. Passing the Vascular Barrier: Endothelial Signaling Processes Controlling Extravasation. Physiol Rev 2019; 99:1467-1525. [PMID: 31140373 DOI: 10.1152/physrev.00037.2018] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A central function of the vascular endothelium is to serve as a barrier between the blood and the surrounding tissue of the body. At the same time, solutes and cells have to pass the endothelium to leave or to enter the bloodstream to maintain homeostasis. Under pathological conditions, for example, inflammation, permeability for fluid and cells is largely increased in the affected area, thereby facilitating host defense. To appropriately function as a regulated permeability filter, the endothelium uses various mechanisms to allow solutes and cells to pass the endothelial layer. These include transcellular and paracellular pathways of which the latter requires remodeling of intercellular junctions for its regulation. This review provides an overview on endothelial barrier regulation and focuses on the endothelial signaling mechanisms controlling the opening and closing of paracellular pathways for solutes and cells such as leukocytes and metastasizing tumor cells.
Collapse
Affiliation(s)
- Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| |
Collapse
|
28
|
Kircher DA, Trombetti KA, Silvis MR, Parkman GL, Fischer GM, Angel SN, Stehn CM, Strain SC, Grossmann AH, Duffy KL, Boucher KM, McMahon M, Davies MA, Mendoza MC, VanBrocklin MW, Holmen SL. AKT1 E17K Activates Focal Adhesion Kinase and Promotes Melanoma Brain Metastasis. Mol Cancer Res 2019; 17:1787-1800. [PMID: 31138602 PMCID: PMC6726552 DOI: 10.1158/1541-7786.mcr-18-1372] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/18/2019] [Accepted: 05/22/2019] [Indexed: 02/03/2023]
Abstract
Alterations in the PI3K/AKT pathway occur in up to 70% of melanomas and are associated with disease progression. The three AKT paralogs are highly conserved but data suggest they have distinct functions. Activating mutations of AKT1 and AKT3 occur in human melanoma but their role in melanoma formation and metastasis remains unclear. Using an established melanoma mouse model, we evaluated E17K, E40K, and Q79K mutations in AKT1, AKT2, and AKT3 and show that mice harboring tumors expressing AKT1E17K had the highest incidence of brain metastasis and lowest mean survival. Tumors expressing AKT1E17K displayed elevated levels of focal adhesion factors and enhanced phosphorylation of focal adhesion kinase (FAK). AKT1E17K expression in melanoma cells increased invasion and this was reduced by pharmacologic inhibition of either AKT or FAK. These data suggest that the different AKT paralogs have distinct roles in melanoma brain metastasis and that AKT and FAK may be promising therapeutic targets. IMPLICATIONS: This study suggests that AKT1E17K promotes melanoma brain metastasis through activation of FAK and provides a rationale for the therapeutic targeting of AKT and/or FAK to reduce melanoma metastasis.
Collapse
Affiliation(s)
- David A Kircher
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Kirby A Trombetti
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Mark R Silvis
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Gennie L Parkman
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Grant M Fischer
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephanie N Angel
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Christopher M Stehn
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Sean C Strain
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Allie H Grossmann
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- ARUP Institute for Clinical and Experimental Pathology, Salt Lake City, Utah
- Department of Pathology, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Keith L Duffy
- Department of Dermatology, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Kenneth M Boucher
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Martin McMahon
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Dermatology, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Michael A Davies
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michelle C Mendoza
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Matthew W VanBrocklin
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Surgery, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Sheri L Holmen
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah.
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Surgery, University of Utah Health Sciences Center, Salt Lake City, Utah
| |
Collapse
|
29
|
Blin MG, Bachelier R, Fallague K, Moussouni K, Aurrand-Lions M, Fernandez S, Guillet B, Robert S, Foucault-Bertaud A, Bardin N, Blot-Chabaud M, Dignat-George F, Leroyer AS. CD146 deficiency promotes plaque formation in a mouse model of atherosclerosis by enhancing RANTES secretion and leukocyte recruitment. J Mol Cell Cardiol 2019; 130:76-87. [PMID: 30928429 DOI: 10.1016/j.yjmcc.2019.03.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 02/26/2019] [Accepted: 03/25/2019] [Indexed: 11/18/2022]
Abstract
AIMS The progression of atherosclerosis is based on the continued recruitment of leukocytes in the vessel wall. The previously described role of CD146 in leukocyte infiltration suggests an involvement for this adhesion molecule in the inflammatory response. In this study, we investigated the role of CD146 in leukocyte recruitment by using an experimental model of atherogenesis. METHODS AND RESULTS The role of CD146 was explored in atherosclerosis by crossing CD146-/- mice with ApoE-/- mice. CD146 -/-/ApoE -/- and ApoE -/- mice were fed a Western diet for 24 weeks and were monitored for aortic wall thickness using high frequency ultrasound. The arterial wall was significantly thicker in CD146-deficient mice. After 24 weeks of Western diet, a significant increase of atheroma in both total aortic lesion and aortic sinus of CD146-null mice was observed. In addition, atherosclerotic lesions were more inflammatory since plaques from CD146-deficient mice contained more neutrophils and macrophages. This was due to up-regulation of RANTES secretion by macrophages in CD146-deficient atherosclerotic arteries. This prompted us to further address the function of CD146 in leukocyte recruitment during acute inflammation by using a second experimental model of peritonitis induced by thioglycollate. Neutrophil recruitment was significantly increased in CD146-deficient mice 12 h after peritonitis induction and associated with higher RANTES levels in the peritoneal cavity. In CD146-null macrophages, we also showed that increased RANTES production was dependent on constitutive inhibition of the p38-MAPK signaling pathway. Finally, Maraviroc, a RANTES receptor antagonist, was able to reduce atherosclerotic lesions and neutrophilia in CD146-deficient mice to the same level as that found in ApoE -/- mice. CONCLUSIONS Our data indicate that CD146 deficiency is associated with the upregulation of RANTES production and increased inflammation of atheroma, which could influence the atherosclerotic plaque fate. Thus, these data identify CD146 agonists as potential new therapeutic candidates for atherosclerosis treatment.
Collapse
Affiliation(s)
- Muriel G Blin
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France
| | - Richard Bachelier
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France
| | - Karim Fallague
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France
| | - Karima Moussouni
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France
| | - Michel Aurrand-Lions
- Aix Marseille Univ., CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Samantha Fernandez
- Aix-Marseille Univ., CERIMED, Secteur Nucléaire Pré-clinique, Timone, 13005 Marseille, France
| | - Benjamin Guillet
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France; Aix-Marseille Univ., CERIMED, Secteur Nucléaire Pré-clinique, Timone, 13005 Marseille, France; Assistance Publique-Hôpitaux de Marseille, Hôpital Nord, Marseille, France
| | - Stéphane Robert
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France
| | | | - Nathalie Bardin
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France; Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, 13385 Marseille, France
| | | | - Françoise Dignat-George
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France; Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, 13385 Marseille, France
| | - Aurélie S Leroyer
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France.
| |
Collapse
|
30
|
Zhou P, Xiong T, Chen J, Li F, Qi T, Yuan J. Clinical significance of melanoma cell adhesion molecule CD146 and VEGFA expression in epithelial ovarian cancer. Oncol Lett 2018; 17:2418-2424. [PMID: 30675307 DOI: 10.3892/ol.2018.9840] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 10/26/2018] [Indexed: 12/19/2022] Open
Abstract
Ovarian cancer is the fifth most common type of cancer in females; however, its asymptomatic progression and the lack of an efficient screening strategy leads to late diagnosis. The present study aimed to investigate the expression levels of cluster of differentiation (CD)146 and vascular endothelial growth factor A (VEGFA) in epithelial ovarian cancer, and their clinical significance. A total of 52 ovarian samples were tested, of which 22 were from patients with epithelial ovarian cancer and 30 were from non-cancer patients. The relative gene expression of CD146 and VEGFA was quantified using reverse transcription-quantitative polymerase chain reaction analysis. Western blotting was used to determine the protein expression levels. The relative gene expression levels of CD146 and VEGFA in tumor tissues were significantly increased compared with the control (4.92±0.44 vs. 1.05±0.06 and 3.08±0.17 vs. 1.06±0.07, P<0.01). The protein expression levels of CD146 and VEGFA in tumor tissue were also significantly increased compared with the control (0.70±0.02 vs. 0.41±0.07 and 0.54±0.01 vs. 0.26±0.01, P<0.01). There was a positive correlation between the expression levels of CD146 and VEGFA genes (r=0.78) and between the two proteins (r=0.69). Dot density frequency analysis indicated that CD146 and VEGFA were specifically present in tumor tissues. In conclusion, CD146 and VEGFA are co-overexpressed in ovarian cancer; their potential as tumor biomarkers or therapeutic targets for the treatment of ovarian cancer requires further investigation.
Collapse
Affiliation(s)
- Ping Zhou
- Department of Gynecology and Obstetrics, Cancer Hospital Affiliated to Xinjiang Medical University, Wulumuqi, Xinjiang 830011, P.R. China
| | - Tingchuan Xiong
- Department of Gynecology and Obstetrics, Cancer Hospital Affiliated to Xinjiang Medical University, Wulumuqi, Xinjiang 830011, P.R. China
| | - Jingxin Chen
- Department of Gynaecology and Obstetrics, Dongfang Hospital Affiliated to Tongji University, Shanghai 200120, P.R. China
| | - Fen Li
- Department of Gynecology and Obstetrics, Cancer Hospital Affiliated to Xinjiang Medical University, Wulumuqi, Xinjiang 830011, P.R. China
| | - Tingting Qi
- Department of Gynecology and Obstetrics, Cancer Hospital Affiliated to Xinjiang Medical University, Wulumuqi, Xinjiang 830011, P.R. China
| | - Jianlin Yuan
- Department of Gynecology and Obstetrics, Cancer Hospital Affiliated to Xinjiang Medical University, Wulumuqi, Xinjiang 830011, P.R. China
| |
Collapse
|
31
|
Long Noncoding RNA uc001pwg.1 Is Downregulated in Neointima in Arteriovenous Fistulas and Mediates the Function of Endothelial Cells Derived from Pluripotent Stem Cells. Stem Cells Int 2017; 2017:4252974. [PMID: 29387090 PMCID: PMC5745761 DOI: 10.1155/2017/4252974] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 09/19/2017] [Accepted: 10/02/2017] [Indexed: 11/17/2022] Open
Abstract
Recent studies indicate important roles for long noncoding RNAs (lncRNAs) as essential regulators of gene expression. However, the specific roles of lncRNAs in stenotic lesions of arteriovenous fistula (AVF) failure are still largely unknown. We first analyzed the expression profiles of lncRNAs in human stenosed and nonstenotic uremic veins using RNA-sequencing methodology. A total of 19 lncRNAs were found to be differentially expressed in stenotic lesions. Among these, uc001pwg.1 was one of the most significantly downregulated lncRNAs and enriched in both control vein segments and human umbilical vein endothelial cells (HUVECs). Further studies revealed that uc001pwg.1 overexpression could increase nitric oxide synthase (eNOS) phosphorylation and nitric oxide (NO) production in endothelial cells (ECs) derived from human-induced pluripotent stem cells (HiPSCs). Mechanistically, uc001pwg.1 improves endothelial function via mediating MCAM expression. This study represents the first effort of identifying a novel candidate lncRNA for modulating the function of iPSC-ECs, which may facilitate the improvement of stem cell-based therapies for AVF failure.
Collapse
|
32
|
Moreno-Fortuny A, Bragg L, Cossu G, Roostalu U. MCAM contributes to the establishment of cell autonomous polarity in myogenic and chondrogenic differentiation. Biol Open 2017; 6:1592-1601. [PMID: 28923978 PMCID: PMC5703611 DOI: 10.1242/bio.027771] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cell polarity has a fundamental role in shaping the morphology of cells and growing tissues. Polarity is commonly thought to be established in response to extracellular signals. Here we used a minimal in vitro assay that enabled us to monitor the determination of cell polarity in myogenic and chondrogenic differentiation in the absence of external signalling gradients. We demonstrate that the initiation of cell polarity is regulated by melanoma cell adhesion molecule (MCAM). We found highly polarized localization of MCAM, Moesin (MSN), Scribble (SCRIB) and Van-Gogh-like 2 (VANGL2) at the distal end of elongating myotubes. Knockout of MCAM or elimination of its endocytosis motif does not impair the initiation of myogenesis or myoblast fusion, but prevents myotube elongation. MSN, SCRIB and VANGL2 remain uniformly distributed in MCAM knockout cells. We show that MCAM is also required at early stages of chondrogenic differentiation. In both myogenic and chondrogenic differentiation MCAM knockout leads to transcriptional downregulation of Scrib and enhanced MAP kinase activity. Our data demonstrates the importance of cell autonomous polarity in differentiation. Summary: CD146/MCAM regulates cell autonomous polarization and asymmetric localization of Scribble, Van-Gogh-like 2 and Moesin, which is required in skeletal muscle myotube elongation and chondrocyte differentiation.
Collapse
Affiliation(s)
- Artal Moreno-Fortuny
- Manchester Academic Health Science Centre, Division of Extracellular Matrix and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Laricia Bragg
- Manchester Academic Health Science Centre, Division of Extracellular Matrix and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Giulio Cossu
- Manchester Academic Health Science Centre, Division of Extracellular Matrix and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Urmas Roostalu
- Manchester Academic Health Science Centre, Division of Extracellular Matrix and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| |
Collapse
|
33
|
Strilic B, Offermanns S. Intravascular Survival and Extravasation of Tumor Cells. Cancer Cell 2017; 32:282-293. [PMID: 28898694 DOI: 10.1016/j.ccell.2017.07.001] [Citation(s) in RCA: 262] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/24/2017] [Accepted: 07/05/2017] [Indexed: 12/17/2022]
Abstract
Most metastasizing tumor cells reach distant sites by entering the circulatory system. Within the bloodstream, they are exposed to severe stress due to loss of adhesion to extracellular matrix, hemodynamic shear forces, and attacks of the immune system, and only a few cells manage to extravasate and to form metastases. We review the current understanding of the cellular and molecular mechanisms that allow tumor cells to survive in the intravascular environment and that mediate and promote tumor cell extravasation. As these processes are critical for the metastatic spread of tumor cells, we discuss implications for potential therapeutic approaches and future research.
Collapse
Affiliation(s)
- Boris Strilic
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Ludwigstr. 43, 61231 Bad Nauheim, Germany
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Ludwigstr. 43, 61231 Bad Nauheim, Germany; J.W. Goethe University Frankfurt, Center for Molecular Medicine, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| |
Collapse
|
34
|
Integrative analysis of DNA methylation and mRNA expression during differentiation of umbilical cord blood derived mononuclear cells to endothelial cells. Gene 2017; 635:48-60. [PMID: 28887159 DOI: 10.1016/j.gene.2017.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 08/29/2017] [Accepted: 09/04/2017] [Indexed: 01/03/2023]
Abstract
Differentiation of umbilical cord blood derived mononuclear cells to endothelial cells is accompanied by massive changes in gene expression. Although methylation and demethylation of DNA likely play crucial roles in regulating gene expression, their interplay during differentiation remains elusive. To address this question, we performed deep sequencing of DNA methylation and mRNA expression to profile global changes in promoter methylation and gene expression during differentiation from mononuclear cells to outgrowing cells. We identified 61 downregulated genes with hypermethylation, including CD74, VAV1, TLR8, and NCF4, as well as 21 upregulated genes with hypomethylation, including ECSCR, MCAM, PGF, and ARHGEF15. Interestingly, gene ontology analysis showed that downregulated genes with hypermethylation were enriched in immune-related functions, and upregulated genes with hypomethylation were enriched in the developmental process and angiogenesis, indicating the important roles of DNA methylation in regulating differentiation. We performed polymerase chain reaction analyses and bisulfite sequencing of representative genes (CD74, VAV1, ECSCR, and MCAM) to verify the negative correlation between DNA methylation and gene expression. Further, inhibition of DNA methyltransferase and demethylase activities using 5'-aza-dc and shRNAs, specific for TET1 and TET2 mRNAs, respectively, revealed that DNA methylation was the main regulator of the reversible expression of functionally important genes. Collectively, our findings implicate DNA methylation as a critical regulator of gene expression during umbilical cord blood derived mononuclear cells to endothelial cell differentiation.
Collapse
|
35
|
Sechler M, Parrish JK, Birks DK, Jedlicka P. The histone demethylase KDM3A, and its downstream target MCAM, promote Ewing Sarcoma cell migration and metastasis. Oncogene 2017; 36:4150-4160. [PMID: 28319067 PMCID: PMC5519422 DOI: 10.1038/onc.2017.44] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 01/27/2017] [Accepted: 02/02/2017] [Indexed: 12/15/2022]
Abstract
Ewing Sarcoma is the second most common solid pediatric malignant neoplasm of bone and soft tissue. Driven by EWS/Ets, or rarely variant, oncogenic fusions, Ewing Sarcoma is a biologically and clinically aggressive disease with a high propensity for metastasis. However, the mechanisms underpinning Ewing Sarcoma metastasis are currently not well understood. In the present study, we identify and characterize a novel metastasis-promotional pathway in Ewing Sarcoma, involving the histone demethylase KDM3A, previously identified by our laboratory as a new cancer-promoting gene in this disease. Using global gene expression profiling, we show that KDM3A positively regulates genes and pathways implicated in cell migration and metastasis, and demonstrate, using functional assays, that KDM3A promotes migration in vitro and experimental, post-intravasation, metastasis in vivo. We further identify the melanoma cell adhesion molecule (MCAM) as a novel KDM3A target gene in Ewing Sarcoma, and an important effector of KDM3A pro-metastatic action. Specifically, we demonstrate that MCAM depletion, like KDM3A depletion, inhibits cell migration in vitro and experimental metastasis in vivo, and that MCAM partially rescues impaired migration due to KDM3A knock-down. Mechanistically, we show that KDM3A regulates MCAM expression both through a direct mechanism, involving modulation of H3K9 methylation at the MCAM promoter, and an indirect mechanism, via the Ets1 transcription factor. Finally, we identify an association between high MCAM levels in patient tumors and poor survival, in two different Ewing Sarcoma clinical cohorts. Taken together, our studies uncover a new metastasis-promoting pathway in Ewing Sarcoma, with therapeutically targetable components.
Collapse
Affiliation(s)
- Marybeth Sechler
- Cancer Biology Graduate Training Program
- University of Colorado Denver, Anschutz Medical Campus, Aurora CO
| | - Janet K. Parrish
- Department of Pathology
- University of Colorado Denver, Anschutz Medical Campus, Aurora CO
| | - Diane K. Birks
- Department of Neurosurgery
- University of Colorado Denver, Anschutz Medical Campus, Aurora CO
| | - Paul Jedlicka
- Cancer Biology Graduate Training Program
- Department of Pathology
- University of Colorado Denver, Anschutz Medical Campus, Aurora CO
| |
Collapse
|
36
|
Kaspi E, Heim X, Granel B, Guillet B, Stalin J, Nollet M, Bertaud-Foucault A, Robaglia-Schlupp A, Roll P, Cau P, Leroyer A, Bachelier R, Benyamine A, Dignat-George F, Blot-Chabaud M, Bardin N. Identification of CD146 as a novel molecular actor involved in systemic sclerosis. J Allergy Clin Immunol 2017; 140:1448-1451.e6. [PMID: 28606586 DOI: 10.1016/j.jaci.2017.04.046] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 03/23/2017] [Accepted: 04/07/2017] [Indexed: 11/26/2022]
Affiliation(s)
- Elise Kaspi
- Aix Marseille Univ, INSERM, GMGF, Marseille, France; APHM, Hôpital la Timone, Service de Biologie Cellulaire, Marseille, France
| | - Xavier Heim
- Aix Marseille Univ, INSERM, VRCM, Marseille, France
| | - Brigitte Granel
- Aix Marseille Univ, INSERM, VRCM, Marseille, France; APHM, Hôpital Nord, Médecine Interne, Marseille, France
| | - Benjamin Guillet
- Aix Marseille Univ, INSERM, VRCM, Marseille, France; APHM, Hôpital Nord, Service de Radio-Pharmacie, Marseille, France; Aix Marseille Univ, CERIMED, Marseille, France
| | - Jimmy Stalin
- Aix Marseille Univ, INSERM, VRCM, Marseille, France
| | - Marie Nollet
- Aix Marseille Univ, INSERM, VRCM, Marseille, France
| | | | - Andrée Robaglia-Schlupp
- Aix Marseille Univ, INSERM, GMGF, Marseille, France; APHM, Hôpital la Timone, Service de Biologie Cellulaire, Marseille, France
| | - Patrice Roll
- Aix Marseille Univ, INSERM, GMGF, Marseille, France; APHM, Hôpital la Timone, Service de Biologie Cellulaire, Marseille, France
| | - Pierre Cau
- Aix Marseille Univ, INSERM, GMGF, Marseille, France
| | | | | | | | - Françoise Dignat-George
- Aix Marseille Univ, INSERM, VRCM, Marseille, France; APHM, Hôpital La Conception, Laboratoire d'Hématologie, Marseille, France
| | | | - Nathalie Bardin
- Aix Marseille Univ, INSERM, VRCM, Marseille, France; APHM, Hôpital La Conception, Laboratoire d'Hématologie, Marseille, France.
| |
Collapse
|
37
|
Colomb F, Wang W, Simpson D, Zafar M, Beynon R, Rhodes JM, Yu LG. Galectin-3 interacts with the cell-surface glycoprotein CD146 (MCAM, MUC18) and induces secretion of metastasis-promoting cytokines from vascular endothelial cells. J Biol Chem 2017; 292:8381-8389. [PMID: 28364041 DOI: 10.1074/jbc.m117.783431] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/29/2017] [Indexed: 12/12/2022] Open
Abstract
The galactoside-binding protein galectin-3 is increasingly recognized as an important player in cancer development, progression, and metastasis via its interactions with various galactoside-terminated glycans. We have shown previously that circulating galectin-3, which is increased up to 30-fold in cancer patients, promotes blood-borne metastasis in an animal cancer model. This effect is partly attributable to the interaction of galectin-3 with unknown receptor(s) on vascular endothelial cells and causes endothelial secretion of several metastasis-promoting cytokines. Here we sought to identify the galectin-3-binding molecule(s) on the endothelial cell surface responsible for the galectin-3-mediated cytokine secretion. Using two different galectin-3 affinity purification processes, we extracted four cell membrane glycoproteins, CD146/melanoma cell adhesion molecule (MCAM)/MUC18, CD31/platelet endothelial cell adhesion molecule-1 (PECAM-1), CD144/VE-cadherin, and CD106/Endoglin, from vascular endothelial cells. CD146 was the major galectin-3-binding ligand and strongly co-localized with galectin-3 on endothelial cell surfaces treated with exogenous galectin-3. Moreover, galectin-3 bound to N-linked glycans on CD146 and induced CD146 dimerization and subsequent activation of AKT signaling. siRNA-mediated suppression of CD146 expression completely abolished the galectin-3-induced secretion of IL-6 and G-CSF cytokines from the endothelial cells. Thus, CD146/MCAM is the functional galectin-3-binding ligand on endothelial cell surfaces responsible for galectin-3-induced secretion of metastasis-promoting cytokines. We conclude that CD146/MCAM interactions with circulating galectin-3 may have an important influence on cancer progression and metastasis.
Collapse
Affiliation(s)
- Florent Colomb
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE
| | - Weikun Wang
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE
| | - Deborah Simpson
- Centre for Proteome Research, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, United Kingdom
| | - Mudaser Zafar
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE
| | - Robert Beynon
- Centre for Proteome Research, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, United Kingdom
| | - Jonathan M Rhodes
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE
| | - Lu-Gang Yu
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE.
| |
Collapse
|
38
|
Hofschröer V, Koch KA, Ludwig FT, Friedl P, Oberleithner H, Stock C, Schwab A. Extracellular protonation modulates cell-cell interaction mechanics and tissue invasion in human melanoma cells. Sci Rep 2017; 7:42369. [PMID: 28205573 PMCID: PMC5304230 DOI: 10.1038/srep42369] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 01/10/2017] [Indexed: 12/25/2022] Open
Abstract
Detachment of cells from the primary tumour precedes metastatic progression by
facilitating cell release into the tissue. Solid tumours exhibit altered pH
homeostasis with extracellular acidification. In human melanoma, the
Na+/H+ exchanger NHE1 is an important modifier of
the tumour nanoenvironment. Here we tested the modulation of cell-cell-adhesion by
extracellular pH and NHE1. MV3 tumour spheroids embedded in a collagen matrix
unravelled the efficacy of cell-cell contact loosening and 3D emigration into an
environment mimicking physiological confinement. Adhesive interaction strength
between individual MV3 cells was quantified using atomic force microscopy and
validated by multicellular aggregation assays. Extracellular acidification from
pHe7.4 to 6.4 decreases cell migration and invasion but increases
single cell detachment from the spheroids. Acidification and NHE1 overexpression
both reduce cell-cell adhesion strength, indicated by reduced maximum pulling forces
and adhesion energies. Multicellular aggregation and spheroid formation are strongly
impaired under acidification or NHE1 overexpression. We show a clear dependence of
melanoma cell-cell adhesion on pHe and NHE1 as a modulator. These effects
are opposite to cell-matrix interactions that are strengthened by protons extruded
via NHE1. We conclude that these opposite effects of NHE1 act synergistically during
the metastatic cascade.
Collapse
Affiliation(s)
| | | | | | - Peter Friedl
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Department of Cell Biology, Nijmegen, The Netherlands.,David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States.,Cancer Genomics Center, CG Utrecht, The Netherlands
| | | | - Christian Stock
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, Münster, Germany
| |
Collapse
|
39
|
Liang YK, Zeng D, Xiao YS, Wu Y, Ouyang YX, Chen M, Li YC, Lin HY, Wei XL, Zhang YQ, Kruyt FAE, Zhang GJ. MCAM/CD146 promotes tamoxifen resistance in breast cancer cells through induction of epithelial-mesenchymal transition, decreased ERα expression and AKT activation. Cancer Lett 2017; 386:65-76. [PMID: 27838413 DOI: 10.1016/j.canlet.2016.11.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 02/05/2023]
Abstract
Tamoxifen resistance presents a prominent clinical challenge in endocrine therapy for hormone sensitive breast cancer. However, the underlying mechanisms that contribute to tamoxifen resistance are not fully understood. In this study, we established a tamoxifen resistant MCF-7 cell line (MCF-7-Tam-R) by continuously incubating MCF-7 cells with 4-OH-tamoxifen. We found that melanoma cell adhesion molecule (MCAM/CD146), a unique epithelial-to-mesenchymal transition (EMT) inducer, was significantly up-regulated at both mRNA and protein levels in MCF-7-Tam-R cells compared to parental MCF-7 cells. Mechanistic research demonstrated that MCAM promotes tamoxifen resistance by transcriptionally suppressing ERα expression and activating the AKT pathway, followed by induction of EMT. Elevated MCAM expression was inversely correlated with recurrence-free and distant metastasis-free survival in a cohort of 4142 patients with breast cancer derived from a public database, particularly in the subgroup only treated with tamoxifen. These results demonstrate a novel function of MCAM in conferring tamoxifen resistance in breast cancer. Targeting MCAM might be a promising therapeutic strategy to overcome tamoxifen resistance in breast cancer patients.
Collapse
Affiliation(s)
- Yuan-Ke Liang
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China; Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - De Zeng
- ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China; Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China
| | - Ying-Sheng Xiao
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Yang Wu
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Yan-Xiu Ouyang
- ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Min Chen
- ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Yao-Chen Li
- ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Hao-Yu Lin
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Shantou University Medical College, 57 Changping Road, Shantou, China
| | - Xiao-Long Wei
- Department of Pathology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China
| | - Yong-Qu Zhang
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Frank A E Kruyt
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| | - Guo-Jun Zhang
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China.
| |
Collapse
|
40
|
Sá da Bandeira D, Casamitjana J, Crisan M. Pericytes, integral components of adult hematopoietic stem cell niches. Pharmacol Ther 2016; 171:104-113. [PMID: 27908803 DOI: 10.1016/j.pharmthera.2016.11.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The interest in perivascular cells as a niche for adult hematopoietic stem cells (HSCs) is significantly growing. In the adult bone marrow (BM), perivascular cells and HSCs cohabit. Among perivascular cells, pericytes are precursors of mesenchymal stem/stromal cells (MSCs) that are capable of differentiating into osteoblasts, adipocytes and chondrocytes. In situ, pericytes are recognised by their localisation to the abluminal side of the blood vessel wall and closely associated with endothelial cells, in combination with the expression of markers such as CD146, neural glial 2 (NG2), platelet derived growth factor receptor β (PDGFRβ), α-smooth muscle actin (α-SMA), nestin (Nes) and/or leptin receptor (LepR). However, not all pericytes share a common phenotype: different immunophenotypes can be associated with distinct mesenchymal features, including hematopoietic support. In adult BM, arteriolar and sinusoidal pericytes control HSC behaviour, maintenance, quiescence and trafficking through paracrine effects. Different groups identified and characterized hematopoietic supportive pericyte subpopulations using various markers and mouse models. In this review, we summarize recent work performed by others to understand the role of the perivascular niche in the biology of HSCs in adults, as well as their importance in the development of therapies.
Collapse
Affiliation(s)
- D Sá da Bandeira
- BHF Centre for Cardiovascular Science, MRC Scottish Centre for Regenerative Medicine, The Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - J Casamitjana
- BHF Centre for Cardiovascular Science, MRC Scottish Centre for Regenerative Medicine, The Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - M Crisan
- BHF Centre for Cardiovascular Science, MRC Scottish Centre for Regenerative Medicine, The Edinburgh Medical School, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
41
|
EphrinB2/EphB4 pathway in postnatal angiogenesis: a potential therapeutic target for ischemic cardiovascular disease. Angiogenesis 2016; 19:297-309. [PMID: 27216867 DOI: 10.1007/s10456-016-9514-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 05/13/2016] [Indexed: 01/12/2023]
Abstract
Ischemic cardiovascular disease remains one of the leading causes of morbidity and mortality in the world. Proangiogenic therapy appears to be a promising and feasible strategy for the patients with ischemic cardiovascular disease, but the results of preclinical and clinical trials are limited due to the complicated mechanisms of angiogenesis. Facilitating the formation of functional vessels is important in rescuing the ischemic cardiomyocytes. EphrinB2/EphB4, a novel pathway in angiogenesis, plays a critical role in both microvascular growth and neovascular maturation. Hence, investigating the mechanisms of EphrinB2/EphB4 pathway in angiogenesis may contribute to the development of novel therapeutics for ischemic cardiovascular disease. Previous reviews mainly focused on the role of EphrinB2/EphB4 pathway in embryo vascular development, but their role in postnatal angiogenesis in ischemic heart disease has not been fully illustrated. Here, we summarized the current knowledge of EphrinB2/EphB4 in angiogenesis and their interaction with other angiogenic pathways in ischemic cardiovascular disease.
Collapse
|
42
|
Hordijk PL. Recent insights into endothelial control of leukocyte extravasation. Cell Mol Life Sci 2016; 73:1591-608. [PMID: 26794844 PMCID: PMC11108429 DOI: 10.1007/s00018-016-2136-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 12/30/2022]
Abstract
In the process of leukocyte migration from the circulation across the vascular wall, the crosstalk with endothelial cells that line the blood vessels is essential. It is now firmly established that in endothelial cells important signaling events are initiated upon leukocyte adhesion that impinge on the regulation of cell-cell contact and control the efficiency of transendothelial migration. In addition, several external factors such as shear force and vascular stiffness were recently identified as important regulators of endothelial signaling and, consequently, leukocyte transmigration. Here, I review recent insights into endothelial signaling events that are linked to leukocyte migration across the vessel wall. In this field, protein phosphorylation and Rho-mediated cytoskeletal dynamics are still widely studied using increasingly sophisticated mouse models. In addition, activation of tyrosine phosphatases, changes in endothelial cell stiffness as well as different vascular beds have all been established as important factors in endothelial signaling and leukocyte transmigration. Finally, I address less-well-studied but interesting components in the endothelium that also control transendothelial migration, such as the ephrins and their Eph receptors, that provide novel insights in the complexity associated with this process.
Collapse
Affiliation(s)
- Peter L Hordijk
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Swammerdam Institute for Life Sciences, University of Amsterdam, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands.
- Department of Physiology, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
43
|
Correa D, Somoza RA, Lin P, Schiemann WP, Caplan AI. Mesenchymal stem cells regulate melanoma cancer cells extravasation to bone and liver at their perivascular niche. Int J Cancer 2015; 138:417-27. [PMID: 26235173 DOI: 10.1002/ijc.29709] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 07/08/2015] [Accepted: 07/20/2015] [Indexed: 12/25/2022]
Abstract
Skeleton and liver are preferred organs for cancer dissemination in metastatic melanoma negatively impacting quality of life, therapeutic success and overall survival rates. At the target organ, the local microenvironment and cell-to-cell interactions between invading and resident stromal cells constitute critical components during the establishment and progression of metastasis. Mesenchymal stem cells (MSCs) possess, in addition to their cell progenitor function, a secretory capacity based on cooperativity with other cell types in injury sites including primary tumors (PT). However, their role at the target organ microenvironment during cancer dissemination is not known. We report that local MSCs, acting as pericytes, regulate the extravasation of melanoma cancer cells (MCC) specifically to murine bone marrow (BM) and liver. Intra-arterially injected wild-type MCC fail to invade those selective organs in a genetic model of perturbed pericyte coverage of the vasculature (PDGF-B(ret/ret)), similar to CD146-deficient MCC injected into wild type mice. Invading MCC interact with resident MSCs/pericytes at the perivascular space through co-expressed CD146 and Sdf-1/CXCL12-CXCR4 signaling. Implanted engineered bone structures with MSCs/pericytes deficient of either Sdf-1/CXCL12 or CD146 become resistant to invasion by circulating MCC. Collectively, the presence of MSCs/pericytes surrounding the target organ vasculature is required for efficient melanoma metastasis to BM and liver.
Collapse
Affiliation(s)
- Diego Correa
- Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, OH
| | - Rodrigo A Somoza
- Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, OH
| | - Paul Lin
- Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, OH
| | - William P Schiemann
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Arnold I Caplan
- Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, OH
| |
Collapse
|