1
|
Degroote RL, Schmalen A, Renner S, Wolf E, Hauck SM, Deeg CA. Diabetic retinopathy from the vitreous proteome perspective: The INS C94Y transgenic pig model study. Proteomics 2024; 24:e2300591. [PMID: 39126128 DOI: 10.1002/pmic.202300591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024]
Abstract
INSC94Y transgenic pigs represent a model for mutant insulin gene-induced diabetes of youth, with impaired insulin secretion and beta cell loss, leading to elevated fasting blood glucose levels. A key complication of diabetes mellitus is diabetic retinopathy (DR), characterized by hyperglycemia-induced abnormalities in the retina. Adjacent to the retina lies the vitreous, a gelatinous matrix vital for ocular function. It harbors proteins and signaling molecules, offering insights into vitreous biology and ocular health. Moreover, as a reservoir for secreted molecules, the vitreous illuminates molecular processes within intraocular structures, especially under pathological conditions. To uncover the proteomic profile of porcine vitreous and explore its relevance to DR, we employed discovery proteomics to compare vitreous samples from INSC94Y transgenic pigs and wild-type controls. Our analysis identified 1404 proteins, with 266 showing differential abundance in INSC94Y vitreous. Notably, the abundances of ITGB1, COX2, and GRIFIN were significantly elevated in INSC94Y vitreous. Gene Set Enrichment Analysis unveiled heightened MYC and mTORC1 signaling in INSC94Y vitreous, shedding light on its biological significance in diabetes-associated ocular pathophysiology. These findings deepen our understanding of vitreous involvement in DR and provide valuable insights into potential therapeutic targets. Raw data are accessible via ProteomeXchange (PXD038198).
Collapse
Affiliation(s)
- Roxane L Degroote
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Adrian Schmalen
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Simone Renner
- Chair for Molecular Animal Breeding and Biotechnology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
| | - Cornelia A Deeg
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| |
Collapse
|
2
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [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: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
3
|
Hayderi A, Zegeye MM, Meydan S, Sirsjö A, Kumawat AK, Ljungberg LU. TNF Induces Laminin-332-Encoding Genes in Endothelial Cells and Laminin-332 Promotes an Atherogenic Endothelial Phenotype. Int J Mol Sci 2024; 25:8699. [PMID: 39201392 PMCID: PMC11354388 DOI: 10.3390/ijms25168699] [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: 07/09/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
Laminins are essential components of the basement membranes, expressed in a tissue- and cell-specific manner under physiological conditions. During inflammatory circumstances, such as atherosclerosis, alterations in laminin composition within vessels have been observed. Our study aimed to assess the influence of tumor necrosis factor-alpha (TNF), a proinflammatory cytokine abundantly found in atherosclerotic lesions, on endothelial laminin gene expression and the effects of laminin-332 (LN332) on endothelial cells' behavior. We also evaluated the expression of LN332-encoding genes in human carotid atherosclerotic plaques. Our findings demonstrate that TNF induces upregulation of LAMB3 and LAMC2, which, along with LAMA3, encode the LN332 isoform. Endothelial cells cultured on recombinant LN332 exhibit decreased claudin-5 expression and display a loosely connected phenotype, with an elevated expression of chemokines and leukocyte adhesion molecules, enhancing their attractiveness and adhesion to leukocytes in vitro. Furthermore, LAMB3 and LAMC2 are upregulated in human carotid plaques and show a positive correlation with TNF expression. In summary, TNF stimulates the expression of LN332-encoding genes in human endothelial cells and LN332 promotes an endothelial phenotype characterized by compromised junctional integrity and increased leukocyte interaction. These findings highlight the importance of basement membrane proteins for endothelial integrity and the potential role of LN332 in atherosclerosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Liza U. Ljungberg
- Cardiovascular Research Centre, Department of Medical Sciences, School of Medicine, Örebro University, 70362 Örebro, Sweden; (A.H.); (S.M.); (A.S.); (A.K.K.)
| |
Collapse
|
4
|
Casali BC, Baptista MP, Pachane BC, Cortez AA, Altei WF, Selistre-de-Araújo HS. Blockage of αvβ3 integrin in 3D culture of triple-negative breast cancer and endothelial cells inhibits migration and discourages endothelial-to-mesenchymal plasticity. Biochem Biophys Rep 2024; 38:101686. [PMID: 38524278 PMCID: PMC10957371 DOI: 10.1016/j.bbrep.2024.101686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 03/26/2024] Open
Abstract
Breast cancer is a relevant cause of mortality in women and its triple-negative subtype (TNBC) is usually associated with poor prognosis. During tumor progression to metastasis, angiogenesis is triggered by the sprouting of endothelial cells from pre-existing vessels by a dynamic chain of events including VE-cadherin downregulation, actin protrusion, and integrin-mediated adhesion, allowing for migration and proliferation. The binding of tumoral and tumor-associated stromal cells with the extracellular matrix through integrins mediates angiogenic processes and certain integrin subtypes, such as the αvβ3 integrin, are upregulated in hypoxic TNBC models. Integrin αvβ3 inhibition by the high-affinity binding disintegrin DisBa-01 was previously demonstrated to induce anti-tumoral and anti-angiogenic responses in traditional 2D cell assays. Here, we investigate the effects of integrin αvβ3 blockage in endothelial and TNBC cells by DisBa-01 in 3D cultures under two oxygen conditions (1% and 20%). 3D cultures created using non-adhesive micromolds with Matrigel were submitted to migration assay in Boyden chambers and fluorescence analysis. DisBa-01 inhibited cell migration in normoxia and hypoxia in both MDA-MB-231 and HUVEC spheroids. Protein levels of integrin αvβ3 were overexpressed in HUVEC spheroids compared to MDA-MB-231 spheroids. In HUVEC 3D cultures, sprouting assays in collagen type I were decreased in normoxia upon DisBa-01 treatment, and VE-cadherin levels were diminished in HUVEC spheroids in hypoxia and upon DisBa-01 treatment. In conclusion, the blockage of integrin αvβ3 by DisBa-01 inhibits cell migration in 3D culture and interferes with tumor-derived responses in different oxygen settings, implicating its crucial role in angiogenesis and tumor progression.
Collapse
Affiliation(s)
- Bruna Carla Casali
- Biochemistry and Molecular Biology Laboratory, Universidade Federal de São Carlos - UFSCar, São Carlos, SP, Brazil
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
| | - Matheus Pintor Baptista
- Biochemistry and Molecular Biology Laboratory, Universidade Federal de São Carlos - UFSCar, São Carlos, SP, Brazil
- Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
| | - Bianca Cruz Pachane
- Biochemistry and Molecular Biology Laboratory, Universidade Federal de São Carlos - UFSCar, São Carlos, SP, Brazil
| | - Anelise Abreu Cortez
- Biochemistry and Molecular Biology Laboratory, Universidade Federal de São Carlos - UFSCar, São Carlos, SP, Brazil
| | - Wanessa Fernanda Altei
- Biochemistry and Molecular Biology Laboratory, Universidade Federal de São Carlos - UFSCar, São Carlos, SP, Brazil
- Radiation Oncology Department, Barretos Cancer Hospital, Barretos, SP, Brazil
- Center of Molecular Oncology Research, Barretos Cancer Hospital, Barretos, SP, Brazil
| | | |
Collapse
|
5
|
Njegić A, Laid L, Zi M, Maniati E, Wang J, Chelu A, Wisniewski L, Hunter J, Prehar S, Stafford N, Gilon C, Hoffman A, Weinmüller M, Kessler H, Cartwright EJ, Hodivala-Dilke K. Treatment with αvβ3-integrin-specific 29P attenuates pressure-overload induced cardiac remodelling after transverse aortic constriction in mice. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 8:100069. [PMID: 38933087 PMCID: PMC11196926 DOI: 10.1016/j.jmccpl.2024.100069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/28/2024] [Accepted: 03/11/2024] [Indexed: 06/28/2024]
Abstract
Heart failure remains one of the largest clinical burdens globally, with little to no improvement in the development of disease-eradicating therapeutics. Integrin targeting has been used in the treatment of ocular disease and cancer, but little is known about its utility in the treatment of heart failure. Here we sought to determine whether the second generation orally available, αvβ3-specific RGD-mimetic, 29P , was cardioprotective. Male mice were subjected to transverse aortic constriction (TAC) and treated with 50 μg/kg 29P or volume-matched saline as Vehicle control. At 3 weeks post-TAC, echocardiography showed that 29P treatment significantly restored cardiac function and structure indicating the protective effect of 29P treatment in this model of heart failure. Importantly, 29P treatment improved cardiac function giving improved fractional shortening, ejection fraction, heart weight and lung weight to tibia length fractions, together with partial restoration of Ace and Mme levels, as markers of the TAC insult. At a tissue level, 29P reduced cardiomyocyte hypertrophy and interstitial fibrosis, both of which are major clinical features of heart failure. RNA sequencing identified that, mechanistically, this occurred with concomitant alterations to genes involved molecular pathways associated with these processes such as metabolism, hypertrophy and basement membrane formation. Overall, targeting αvβ3 with 29P provides a novel strategy to attenuate pressure-overload induced cardiac hypertrophy and fibrosis, providing a possible new approach to heart failure treatment.
Collapse
Affiliation(s)
- Alexandra Njegić
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Lina Laid
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Min Zi
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Eleni Maniati
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Jun Wang
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Alexandru Chelu
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Laura Wisniewski
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Jenna Hunter
- Division of Diabetes, Endocrinology & Gastroenterology, School of Medical Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Sukhpal Prehar
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Nicholas Stafford
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Chaim Gilon
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Amnon Hoffman
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, P.O. Box 12065, Jerusalem 91120, Israel
| | - Michael Weinmüller
- Institute for Advanced Study, TUM School of Natural Science, Technische Universität München, Lichtenbergstr. 4, 85747 Garching, Germany
| | - Horst Kessler
- Institute for Advanced Study, TUM School of Natural Science, Technische Universität München, Lichtenbergstr. 4, 85747 Garching, Germany
| | - Elizabeth J. Cartwright
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Kairbaan Hodivala-Dilke
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| |
Collapse
|
6
|
Tripathy DK, Panda LP, Biswal S, Barhwal K. Insights into the glioblastoma tumor microenvironment: current and emerging therapeutic approaches. Front Pharmacol 2024; 15:1355242. [PMID: 38523646 PMCID: PMC10957596 DOI: 10.3389/fphar.2024.1355242] [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: 12/13/2023] [Accepted: 02/07/2024] [Indexed: 03/26/2024] Open
Abstract
Glioblastoma (GB) is an intrusive and recurrent primary brain tumor with low survivability. The heterogeneity of the tumor microenvironment plays a crucial role in the stemness and proliferation of GB. The tumor microenvironment induces tumor heterogeneity of cancer cells by facilitating clonal evolution and promoting multidrug resistance, leading to cancer cell progression and metastasis. It also plays an important role in angiogenesis to nourish the hypoxic tumor environment. There is a strong interaction of neoplastic cells with their surrounding microenvironment that comprise several immune and non-immune cellular components. The tumor microenvironment is a complex network of immune components like microglia, macrophages, T cells, B cells, natural killer (NK) cells, dendritic cells and myeloid-derived suppressor cells, and non-immune components such as extracellular matrix, endothelial cells, astrocytes and neurons. The prognosis of GB is thus challenging, making it a difficult target for therapeutic interventions. The current therapeutic approaches target these regulators of tumor micro-environment through both generalized and personalized approaches. The review provides a summary of important milestones in GB research, factors regulating tumor microenvironment and promoting angiogenesis and potential therapeutic agents widely used for the treatment of GB patients.
Collapse
Affiliation(s)
- Dev Kumar Tripathy
- Department of Physiology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Lakshmi Priya Panda
- Department of Physiology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Suryanarayan Biswal
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Kalpana Barhwal
- Department of Physiology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| |
Collapse
|
7
|
Liang JY, Wei HJ, Tang YY. Isthmin: A multifunctional secretion protein. Cytokine 2024; 173:156423. [PMID: 37979212 DOI: 10.1016/j.cyto.2023.156423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 11/20/2023]
Abstract
Isthmin is a polypeptide secreted by adipocytes that was first detected in Xenopus gastrula embryos. Recent studies have focused on the biological functions of isthmin in growth and development, angiogenesis, and metabolism. Distinct spatiotemporal expression of isthmin-1 (ISM-1) was observed during growth and development. ISM-1 plays an important role in the occurrence and development of cancer by regulating cell proliferation, migration, angiogenesis, and immune microenvironments. Moreover, ISM-1, as a newly identified insulin-like adipokine, increases adipocyte glucose uptake and inhibits hepatic lipid synthesis. However, the biological function of ISM-1 remains largely unknown. In this review, we highlight the structure and physiological functions of isthmin and explore its application potential, contributing to a better understanding of its function and providing prevention and treatment strategies for various diseases.
Collapse
Affiliation(s)
- Jin-Yu Liang
- Department of Physiology, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, PR China; Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China
| | - Hai-Jun Wei
- Department of Physiology, Hunan Polytechnic of Environment and Biology, Hengyang 421001, Hunan, PR China
| | - Yi-Yun Tang
- Department of Physiology, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, PR China; Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China.
| |
Collapse
|
8
|
Halder SK, Sapkota A, Milner R. The importance of laminin at the blood-brain barrier. Neural Regen Res 2023; 18:2557-2563. [PMID: 37449589 DOI: 10.4103/1673-5374.373677] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
The blood-brain barrier is a unique property of central nervous system blood vessels that protects sensitive central nervous system cells from potentially harmful blood components. The mechanistic basis of this barrier is found at multiple levels, including the adherens and tight junction proteins that tightly bind adjacent endothelial cells and the influence of neighboring pericytes, microglia, and astrocyte endfeet. In addition, extracellular matrix components of the vascular basement membrane play a critical role in establishing and maintaining blood-brain barrier integrity, not only by providing an adhesive substrate for blood-brain barrier cells to adhere to, but also by providing guidance cues that strongly influence vascular cell behavior. The extracellular matrix protein laminin is one of the most abundant components of the basement membrane, and several lines of evidence suggest that it plays a key role in directing blood-brain barrier behavior. In this review, we describe the basic structure of laminin and its receptors, the expression patterns of these molecules in central nervous system blood vessels and how they are altered in disease states, and most importantly, how genetic deletion of different laminin isoforms or their receptors reveals the contribution of these molecules to blood-brain barrier function and integrity. Finally, we discuss some of the important unanswered questions in the field and provide a "to-do" list of some of the critical outstanding experiments.
Collapse
Affiliation(s)
- Sebok K Halder
- San Diego Biomedical Research Institute, San Diego, CA, USA
| | - Arjun Sapkota
- San Diego Biomedical Research Institute, San Diego, CA, USA
| | - Richard Milner
- San Diego Biomedical Research Institute, San Diego, CA, USA
| |
Collapse
|
9
|
Chen X, Zhao Y, Huang Y, Zhu K, Zeng F, Zhao J, Zhang H, Zhu X, Kettenmann H, Xiang X. TREM2 promotes glioma progression and angiogenesis mediated by microglia/brain macrophages. Glia 2023; 71:2679-2695. [PMID: 37641212 DOI: 10.1002/glia.24456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/23/2023] [Accepted: 07/31/2023] [Indexed: 08/31/2023]
Abstract
Triggering receptor expressed on myeloid cell 2 (TREM2), a myeloid cell-specific signaling molecule, controls essential functions of microglia and impacts on the pathogenesis of Alzheimer's disease and other neurodegenerative disorders. TREM2 is also highly expressed in tumor-associated macrophages in different types of cancer. Here, we studied whether TREM2 influences glioma progression. We found a gender-dependent effect of glioma growth in wild-type (WT) animals injected with GL261-EGFP glioma cells. Most importantly, TREM2 promotes glioma progression in male but not female animals. The accumulation of glioma-associated microglia/macrophages (GAMs) and CD31+ blood vessel density is reduced in male TREM2-deficient mice. A transcriptomic analysis of glioma tissue revealed that TREM2 deficiency suppresses immune-related genes. In an organotypic slice model devoid of functional vascularization and immune components from periphery, the tumor size was not affected by TREM2-deficiency. In human resection samples from glioblastoma, TREM2 is upregulated in GAMs. Based on the Cancer Genome Atlas Program (TCGA) and the Chinese Glioma Genome Atlas (CGGA) databases, the TREM2 expression levels were negatively correlated with survival. Thus, the TREM2-dependent crosstalk between GAMs and the vasculature formation promotes glioma growth.
Collapse
Affiliation(s)
- Xuezhen Chen
- Shenzhen Key Laboratory of Immunomodulation for Neurological Diseases, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yue Zhao
- Shenzhen Key Laboratory of Immunomodulation for Neurological Diseases, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yimin Huang
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Kaichuan Zhu
- Shenzhen Key Laboratory of Immunomodulation for Neurological Diseases, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fan Zeng
- Shenzhen Key Laboratory of Immunomodulation for Neurological Diseases, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Junyi Zhao
- Shenzhen Key Laboratory of Immunomodulation for Neurological Diseases, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Huaqiu Zhang
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xinzhou Zhu
- Shenzhen Key Laboratory of Immunomodulation for Neurological Diseases, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Helmut Kettenmann
- Shenzhen Key Laboratory of Immunomodulation for Neurological Diseases, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Xianyuan Xiang
- Shenzhen Key Laboratory of Immunomodulation for Neurological Diseases, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
10
|
Fejza A, Camicia L, Carobolante G, Poletto E, Paulitti A, Schinello G, Di Siena E, Cannizzaro R, Iozzo RV, Baldassarre G, Andreuzzi E, Spessotto P, Mongiat M. Emilin2 fosters vascular stability by promoting pericyte recruitment. Matrix Biol 2023; 122:18-32. [PMID: 37579864 DOI: 10.1016/j.matbio.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/20/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
Angiogenesis, the formation of the new blood vessels from pre-existing vasculature, is an essential process occurring under both normal and pathological conditions, such as inflammation and cancer. This complex process is regulated by several cytokines, growth factors and extracellular matrix components modulating endothelial cell and pericyte function. In this study, we discovered that the extracellular matrix glycoprotein Elastin Microfibril Interfacer 2 (Emilin2) plays a prominent role in pericyte physiology. This work was originally prompted by the observations that tumor-associated vessels from Emilin2-/- mice display less pericyte coverage, impaired vascular perfusion, and reduced drug efficacy, suggesting that Emilin2 could promote vessel maturation and stabilization affecting pericyte recruitment. We found that Emilin2 affects different mechanisms engaged in pericyte recruitment and vascular stabilization. First, human primary endothelial cells challenged with recombinant Emilin2 synthesized and released ∼ 2.1 and 1.2 folds more PDGF-BB and HB-EGF, two cytokines known to promote pericyte recruitment. We also discovered that Emilin2, by directly engaging α5β1 and α6β1 integrins, highly expressed in pericytes, served as an adhesion substrate and haptotactic stimulus for pericytes. Moreover, Emilin2 evoked increased NCadherin expression via the sphingosine-1-phosphate receptor, leading to enhanced vascular stability by fostering interconnection between endothelial cells and pericytes. Finally, restoring pericyte coverage in melanoma and ovarian tumor vessels developed in Emilin2-/- mice improved drug delivery to the tumors. Collectively, our results implicate Emilin2 as a prominent regulator of pericyte function and suggest that Emilin2 expression could represent a promising maker to predict the clinical outcome of patients with melanoma, ovarian, and potentially other forms of cancer.
Collapse
Affiliation(s)
- Albina Fejza
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy; UBT-Higher Education Institution, Kalabria, Street Rexhep Krasniqi Nr. 56, Prishtina 10000, Kosovo
| | - Lucrezia Camicia
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Greta Carobolante
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Evelina Poletto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Alice Paulitti
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy; VivaBioCell S.P.A., Udine, Italy
| | - Giorgia Schinello
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Emanuele Di Siena
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Renato Cannizzaro
- Department of Clinical Oncology, Oncological Gastroenterology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste 34127, Italy
| | - Renato V Iozzo
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Gustavo Baldassarre
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Eva Andreuzzi
- Obstetrics and Gynecology, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste 34137, Italy
| | - Paola Spessotto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Maurizio Mongiat
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy.
| |
Collapse
|
11
|
Zhang L, Wang P, Zhou XQ, Bretin L, Zeng X, Husiev Y, Polanco EA, Zhao G, Wijaya LS, Biver T, Le Dévédec SE, Sun W, Bonnet S. Cyclic Ruthenium-Peptide Conjugates as Integrin-Targeting Phototherapeutic Prodrugs for the Treatment of Brain Tumors. J Am Chem Soc 2023. [PMID: 37379365 DOI: 10.1021/jacs.3c04855] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
To investigate the potential of tumor-targeting photoactivated chemotherapy, a chiral ruthenium-based anticancer warhead, Λ/Δ-[Ru(Ph2phen)2(OH2)2]2+, was conjugated to the RGD-containing Ac-MRGDH-NH2 peptide by direct coordination of the M and H residues to the metal. This design afforded two diastereoisomers of a cyclic metallopeptide, Λ-[1]Cl2 and Δ-[1]Cl2. In the dark, the ruthenium-chelating peptide had a triple action. First, it prevented other biomolecules from coordinating with the metal center. Second, its hydrophilicity made [1]Cl2 amphiphilic so that it self-assembled in culture medium into nanoparticles. Third, it acted as a tumor-targeting motif by strongly binding to the integrin (Kd = 0.061 μM for the binding of Λ-[1]Cl2 to αIIbβ3), which resulted in the receptor-mediated uptake of the conjugate in vitro. Phototoxicity studies in two-dimensional (2D) monolayers of A549, U87MG, and PC-3 human cancer cell lines and U87MG three-dimensional (3D) tumor spheroids showed that the two isomers of [1]Cl2 were strongly phototoxic, with photoindexes up to 17. Mechanistic studies indicated that such phototoxicity was due to a combination of photodynamic therapy (PDT) and photoactivated chemotherapy (PACT) effects, resulting from both reactive oxygen species generation and peptide photosubstitution. Finally, in vivo studies in a subcutaneous U87MG glioblastoma mice model showed that [1]Cl2 efficiently accumulated in the tumor 12 h after injection, where green light irradiation generated a stronger tumoricidal effect than a nontargeted analogue ruthenium complex [2]Cl2. Considering the absence of systemic toxicity for the treated mice, these results demonstrate the high potential of light-sensitive integrin-targeted ruthenium-based anticancer compounds for the treatment of brain cancer in vivo.
Collapse
Affiliation(s)
- Liyan Zhang
- Leiden Institute of Chemistry, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Peiyuan Wang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China
| | - Xue-Quan Zhou
- Leiden Institute of Chemistry, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Ludovic Bretin
- Leiden Institute of Chemistry, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Xiaolong Zeng
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Yurii Husiev
- Leiden Institute of Chemistry, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Ehider A Polanco
- Leiden Institute of Chemistry, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Gangyin Zhao
- Leiden Institute of Biology, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Lukas S Wijaya
- Leiden Academic Centre for Drug Research, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Tarita Biver
- Department of Chemistry and Industrial Chemistry, University of Pisa, 56124 Pisa, Italy
| | - Sylvia E Le Dévédec
- Leiden Academic Centre for Drug Research, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Sylvestre Bonnet
- Leiden Institute of Chemistry, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| |
Collapse
|
12
|
The Active Role of Pericytes During Neuroinflammation in the Adult Brain. Cell Mol Neurobiol 2023; 43:525-541. [PMID: 35195811 DOI: 10.1007/s10571-022-01208-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/13/2022] [Indexed: 12/11/2022]
Abstract
Microvessels in the central nervous system (CNS) have one of the highest populations of pericytes, indicating their crucial role in maintaining homeostasis. Pericytes are heterogeneous cells located around brain microvessels; they present three different morphologies along the CNS vascular tree: ensheathing, mesh, and thin-strand pericytes. At the arteriole-capillary transition ensheathing pericytes are found, while mesh and thin-strand pericytes are located at capillary beds. Brain pericytes are essential for the establishment and maintenance of the blood-brain barrier, which restricts the passage of soluble and potentially toxic molecules from the circulatory system to the brain parenchyma. Pericytes play a key role in regulating local inflammation at the CNS. Pericytes can respond differentially, depending on the degree of inflammation, by secreting a set of neurotrophic factors to promote cell survival and regeneration, or by potentiating inflammation through the release of inflammatory mediators (e.g., cytokines and chemokines), and the overexpression of cell adhesion molecules. Under inflammatory conditions, pericytes may regulate immune cell trafficking to the CNS and play a role in perpetuating local inflammation. In this review, we describe pericyte responses during acute and chronic neuroinflammation.
Collapse
|
13
|
Moreira HR, Rodrigues DB, Freitas-Ribeiro S, da Silva LP, Morais ADS, Jarnalo M, Horta R, Reis RL, Pirraco RP, Marques AP. Spongy-like hydrogels prevascularization with the adipose tissue vascular fraction delays cutaneous wound healing by sustaining inflammatory cell influx. Mater Today Bio 2022; 17:100496. [PMID: 36420053 PMCID: PMC9677215 DOI: 10.1016/j.mtbio.2022.100496] [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: 07/28/2022] [Revised: 11/03/2022] [Accepted: 11/13/2022] [Indexed: 11/16/2022] Open
Abstract
In vitro prevascularization is one of the most explored approaches to foster engineered tissue vascularization. We previously demonstrated a benefit in tissue neovascularization by using integrin-specific biomaterials prevascularized by stromal vascular fraction (SVF) cells, which triggered vasculogenesis in the absence of extrinsic growth factors. SVF cells are also associated to biological processes important in cutaneous wound healing. Thus, we aimed to investigate whether in vitro construct prevascularization with SVF accelerates the healing cascade by fostering early vascularization vis-à-vis SVF seeding prior to implantation. Prevascularized constructs delayed re-epithelization of full-thickness mice wounds compared to both non-prevascularized and control (no SVF) groups. Our results suggest this delay is due to a persistent inflammation as indicated by a significantly lower M2(CD163+)/M1(CD86+) macrophage subtype ratio. Moreover, a slower transition from the inflammatory to the proliferative phase of the healing was confirmed by reduced extracellular matrix deposition and increased presence of thick collagen fibers from early time-points, suggesting the prevalence of fiber crosslinking in relation to neodeposition. Overall, while prevascularization potentiates inflammatory cell influx, which negatively impacts the cutaneous wound healing cascade, an effective wound healing was guaranteed in non-prevascularized SVF cell-containing spongy-like hydrogels confirming that the SVF can have enhanced efficacy.
Collapse
Affiliation(s)
- Helena R. Moreira
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, Portugal
| | - Daniel B. Rodrigues
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, Portugal
| | - Sara Freitas-Ribeiro
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, Portugal
| | - Lucília P. da Silva
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, Portugal
| | - Alain da S. Morais
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, Portugal
| | - Mariana Jarnalo
- Department of Plastic and Reconstructive Surgery, And Burn Unity, Centro Hospitalar de São João, Porto, Portugal
- Faculty of Medicine - University of Porto, Portugal
| | - Ricardo Horta
- Department of Plastic and Reconstructive Surgery, And Burn Unity, Centro Hospitalar de São João, Porto, Portugal
- Faculty of Medicine - University of Porto, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, Portugal
| | - Rogério P. Pirraco
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, Portugal
| | - Alexandra P. Marques
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, Portugal
| |
Collapse
|
14
|
Parfenova LV, Galimshina ZR, Gil’fanova GU, Alibaeva EI, Danilko KV, Aubakirova VR, Farrakhov RG, Parfenov EV, Valiev RZ. Modeling of Biological Activity of PEO-Coated Titanium Implants with Conjugates of Cyclic RGD Peptide with Amino Acid Bisphosphonates. MATERIALS (BASEL, SWITZERLAND) 2022; 15:8120. [PMID: 36431607 PMCID: PMC9699121 DOI: 10.3390/ma15228120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/07/2022] [Accepted: 11/12/2022] [Indexed: 06/16/2023]
Abstract
Titanium is considered to be the most essential metal in the field of implantology. The main factors determining metal biocompatibility, among others, include the morphology and chemical composition of the titanium surface. Therefore, the aim of this work was to develop approaches to control the biological activity of the titanium surface by creating coatings that combine both an inorganic phase with a given morphology and organic molecules containing an integrin-selective peptide that regulate cell adhesion and proliferation. As such, we synthesized new c(RGDfC) derivatives of amino acid bisphosphonates (four examples) with different bisphosphonate anchors and maleimide linkers. These molecules were deposited on a highly developed porous surface obtained via the plasma electrolytic oxidation (PEO) of coarse-grained and nanostructured titanium. In vitro studies demonstrated the increase in the viability degree of mesenchymal stem cells and fibroblasts on the surface of coarse-grained or nanostructured titanium modified with PEO and a c(RGDfC) derivative of ε-aminocaproic acid bisphophonate with an SMCC linker. As a result, the use of conjugates of amino acid bisphosphonates with a cyclic RGD peptide for the modification of PEO-coated titanium opens the ways for the effective control of the biological activity of the metal implant surface.
Collapse
Affiliation(s)
- Lyudmila V. Parfenova
- Institute of Petrochemistry and Catalysis, Ufa Federal Research Center, Russian Academy of Sciences, 141, Prospekt Oktyabrya, 450075 Ufa, Russia
| | - Zulfiya R. Galimshina
- Institute of Petrochemistry and Catalysis, Ufa Federal Research Center, Russian Academy of Sciences, 141, Prospekt Oktyabrya, 450075 Ufa, Russia
| | - Guzel U. Gil’fanova
- Institute of Petrochemistry and Catalysis, Ufa Federal Research Center, Russian Academy of Sciences, 141, Prospekt Oktyabrya, 450075 Ufa, Russia
| | - Eliza I. Alibaeva
- Institute of Petrochemistry and Catalysis, Ufa Federal Research Center, Russian Academy of Sciences, 141, Prospekt Oktyabrya, 450075 Ufa, Russia
| | - Ksenia V. Danilko
- Central Research Laboratory, Bashkir State Medical University, 3 Lenin Street, 450000 Ufa, Russia
| | - Veta R. Aubakirova
- Department of Electronic Engineering, Ufa State Aviation Technical University, 12 Karl Marx Street, 450008 Ufa, Russia
| | - Ruzil G. Farrakhov
- Department of Electronic Engineering, Ufa State Aviation Technical University, 12 Karl Marx Street, 450008 Ufa, Russia
| | - Evgeny V. Parfenov
- Department of Materials Science and Physics of Metals, Ufa State Aviation Technical University, 12 Karl Marx Street, 450008 Ufa, Russia
| | - Ruslan Z. Valiev
- Department of Materials Science and Physics of Metals, Ufa State Aviation Technical University, 12 Karl Marx Street, 450008 Ufa, Russia
| |
Collapse
|
15
|
Chen JR, Zhao JT, Xie ZZ. Integrin-mediated cancer progression as a specific target in clinical therapy. Biomed Pharmacother 2022; 155:113745. [DOI: 10.1016/j.biopha.2022.113745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 11/15/2022] Open
|
16
|
Blanchard N, Link PA, Farkas D, Harmon B, Hudson J, Bogamuwa S, Piper B, Authelet K, Cool CD, Heise RL, Freishtat R, Farkas L. Dichotomous role of integrin-β5 in lung endothelial cells. Pulm Circ 2022; 12:e12156. [PMID: 36438452 PMCID: PMC9684688 DOI: 10.1002/pul2.12156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 10/17/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive, devastating disease, and its main histological manifestation is an occlusive pulmonary arteriopathy. One important functional component of PAH is aberrant endothelial cell (EC) function including apoptosis-resistance, unchecked proliferation, and impaired migration. The mechanisms leading to and maintaining physiologic and aberrant EC function are not fully understood. Here, we tested the hypothesis that in PAH, ECs have increased expression of the transmembrane protein integrin-β5, which contributes to migration and survival under physiologic and pathological conditions, but also to endothelial-to-mesenchymal transition (EnMT). We found that elevated integrin-β5 expression in pulmonary artery lesions and lung tissue from PAH patients and rats with PH induced by chronic hypoxia and injection of CD117+ rat lung EC clones. These EC clones exhibited elevated expression of integrin-β5 and its heterodimerization partner integrin-αν and showed accelerated barrier formation. Inhibition of integrin-ανβ5 in vitro partially blocked transforming growth factor (TGF)-β1-induced EnMT gene expression in rat lung control ECs and less in rat lung EC clones and human lung microvascular ECs. Inhibition of integrin-ανβ5 promoted endothelial dysfunction as shown by reduced migration in a scratch assay and increased apoptosis in synergism with TGF-β1. In vivo, blocking of integrin-ανβ5 exaggerated PH induced by chronic hypoxia and CD117+ EC clones in rats. In summary, we found a role for integrin-ανβ5 in lung endothelial survival and migration, but also a partial contribution to TGF-β1-induced EnMT gene expression. Our results suggest that integrin-ανβ5 is required for physiologic function of ECs and lung vascular homeostasis.
Collapse
Affiliation(s)
- Neil Blanchard
- Department of Orthopedic SurgeryUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Patrick A. Link
- Departments of Physiology and Biomedical EngineeringMayo ClinicRochesterMichiganUSA
- Department of Biomedical Engineering, School of EngineeringVirginia Commonwealth UniversityCharlottesvilleVirginiaUSA
| | - Daniela Farkas
- Division of Pulmonary Disease, College of Medicine, Department of Internal Medicine, Critical Care & Sleep Medicine, Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Brennan Harmon
- Department of Pediatrics, Division of Emergency MedicineChildren's National Health SystemWashingtonDistrict of ColumbiaUSA
| | - Jaylen Hudson
- Division of Pulmonary Disease, College of Medicine, Department of Internal Medicine, Critical Care & Sleep Medicine, Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Srimathi Bogamuwa
- Division of Pulmonary Disease, College of Medicine, Department of Internal Medicine, Critical Care & Sleep Medicine, Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Bryce Piper
- Division of Pulmonary Disease, College of Medicine, Department of Internal Medicine, Critical Care & Sleep Medicine, Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Kayla Authelet
- Department of Pediatrics, Division of Emergency MedicineChildren's National Health SystemWashingtonDistrict of ColumbiaUSA
| | - Carlyne D. Cool
- Department of PathologyUniversity of Colorado at DenverDenverColoradoUSA
| | - Rebecca L. Heise
- Department of Biomedical Engineering, School of EngineeringVirginia Commonwealth UniversityCharlottesvilleVirginiaUSA
| | - Robert Freishtat
- Department of Pediatrics, Division of Emergency MedicineChildren's National Health SystemWashingtonDistrict of ColumbiaUSA
| | - Laszlo Farkas
- Division of Pulmonary Disease, College of Medicine, Department of Internal Medicine, Critical Care & Sleep Medicine, Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Department of Physiology and BiophysicsVirginia Commonwealth UniversityRichmondVirginiaUSA
| |
Collapse
|
17
|
Integrin-specific hydrogels for growth factor-free vasculogenesis. NPJ Regen Med 2022; 7:57. [PMID: 36167724 PMCID: PMC9515164 DOI: 10.1038/s41536-022-00253-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
Integrin-binding biomaterials have been extensively evaluated for their capacity to enable de novo formation of capillary-like structures/vessels, ultimately supporting neovascularization in vivo. Yet, the role of integrins as vascular initiators in engineered materials is still not well understood. Here, we show that αvβ3 integrin-specific 3D matrices were able to retain PECAM1+ cells from the stromal vascular fraction (SVF) of adipose tissue, triggering vasculogenesis in vitro in the absence of extrinsic growth factors. Our results suggest that αvβ3-RGD-driven signaling in the formation of capillary-like structures prevents the activation of the caspase 8 pathway and activates the FAK/paxillin pathway, both responsible for endothelial cells (ECs) survival and migration. We also show that prevascularized αvβ3 integrin-specific constructs inosculate with the host vascular system fostering in vivo neovascularization. Overall, this work demonstrates the ability of the biomaterial to trigger vasculogenesis in an integrin-specific manner, by activating essential pathways for EC survival and migration within a self-regulatory growth factor microenvironment. This strategy represents an improvement to current vascularization routes for Tissue Engineering constructs, potentially enhancing their clinical applicability.
Collapse
|
18
|
Zhao P, Zhao S, Zhang J, Lai M, Sun L, Yan F. Molecular Imaging of Steroid-Induced Osteonecrosis of the Femoral Head through iRGD-Targeted Microbubbles. Pharmaceutics 2022; 14:pharmaceutics14091898. [PMID: 36145646 PMCID: PMC9505504 DOI: 10.3390/pharmaceutics14091898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/15/2022] [Accepted: 06/25/2022] [Indexed: 11/16/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a disease that is commonly seen in the clinic, but its detection rate remains limited, especially at the early stage. We developed an ultrasound molecular imaging (UMI) approach for early diagnosis of ONFH by detecting the expression of integrin αvβ3 during the pathological changes in steroid-induced osteonecrosis of the femoral head (SIONFH) in rat models. The integrin αvβ3-targeted PLGA or lipid microbubbles modified with iRGD peptides were fabricated and characterized. Their adhesion efficiency to mouse brain microvascular endothelial cells in vitro was examined, and the better LIPOiRGD was used for further in vivo molecular imaging of SIONFH rats at 1, 3 and 5 weeks; revealing significantly higher UMI signals could be observed in the 3-week and 5-week SIONFH rats but not in the 1-week SIONFH rats in comparison with the non-targeted microbubbles (32.75 ± 0.95 vs. 0.17 ± 0.09 for 5 weeks, p < 0.05; 5.60 ± 1.31 dB vs. 0.94 ± 0.81 dB for 3 weeks, p < 0.01; 1.13 ± 0.13 dB vs. 0.73 ± 0.31 dB for 1 week, p > 0.05). These results were consistent with magnetic resonance imaging data and confirmed by immunofluorescence staining experiments. In conclusion, our study provides an alternative UMI approach to the early evaluation of ONFH.
Collapse
Affiliation(s)
- Ping Zhao
- Department of Ultrasound, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Shuai Zhao
- Department of Ultrasound, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
- Department of Ultrasound, Suzhou Hospital of Anhui Medical University (Suzhou Municipal Hospital of Anhui Province), Suzhou 234000, China
| | - Jiaqi Zhang
- Department of Ultrasound, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Manlin Lai
- Department of Ultrasound, The Second People’s Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen 518061, China
| | - Litao Sun
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou 310014, China
- Correspondence: (L.S.); (F.Y.); Tel.: +86-755-8639-2284 (F.Y.); Fax: +86-755-9638-2299 (F.Y.)
| | - Fei Yan
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Correspondence: (L.S.); (F.Y.); Tel.: +86-755-8639-2284 (F.Y.); Fax: +86-755-9638-2299 (F.Y.)
| |
Collapse
|
19
|
Nakashima Y, Yoshida S, Tsukahara M. Semi-3D cultures using Laminin 221 as a coating material for human induced pluripotent stem cells. Regen Biomater 2022; 9:rbac060. [PMID: 36176714 PMCID: PMC9514851 DOI: 10.1093/rb/rbac060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/09/2022] [Accepted: 08/21/2022] [Indexed: 11/19/2022] Open
Abstract
It was previously believed that human induced pluripotent stem cells (hiPSCs) did not show adhesion to the coating material Laminin 221, which is known to have specific affinity for cardiomyocytes. In this study, we report that human mononuclear cell-derived hiPSCs, established with Sendai virus vector, form peninsular-like colonies rather than embryonic stem cell-like colonies; these peninsular-like colonies can be passaged more than 10 times after establishment. Additionally, initialization-deficient cells with residual Sendai virus vector adhered to the coating material Laminin 511 but not to Laminin 221. Therefore, the expression of undifferentiated markers tended to be higher in hiPSCs established on Laminin 221 than on Laminin 511. On Laminin 221, hiPSCs15M66 showed a semi-floating colony morphology. The expression of various markers of cell polarity was significantly lower in hiPSCs cultured on Laminin 221 than in hiPSCs cultured on Laminin 511. Furthermore, 201B7 and 15M66 hiPSCs showed 3D cardiomyocyte differentiation on Laminin 221. Thus, the coating material Laminin 221 provides semi-floating culture conditions for the establishment, culture and induced differentiation of hiPSCs.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto 606-8397, Japan
| | - Shinsuke Yoshida
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto 606-8397, Japan
| | - Masayoshi Tsukahara
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto 606-8397, Japan
| |
Collapse
|
20
|
Adhesion Molecules and Vulnerable Plaques – Promoters of Acute Coronary Syndromes. JOURNAL OF CARDIOVASCULAR EMERGENCIES 2022. [DOI: 10.2478/jce-2022-0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Abstract
Biological factors that characterize extrinsic plaque vulnerability include various pro- and anti-inflammatory cytokines that contribute to the development and progression of atherosclerosis. Adhesion molecules are among the initiators of the atherosclerotic process, by mediation of endothelial inflammation. The soluble forms of these adhesion molecules have been identified in the circulatory blood, with an increased level in case of subjects with atherosclerotic lesions and higher levels in patients with acute coronary syndromes or vulnerable plaques. In addition, several authors have found a significant predictive capacity of these molecules in case of patients presenting with acute coronary and cerebrovascular events. The aim of this manuscript is to provide a short description of the role of adhesion molecules in the development and progression of atherosclerotic lesions towards acute coronary syndromes, as well as their capacity for predicting major adverse cardiovascular events in vulnerable cardiovascular patients.
Collapse
|
21
|
Qin T, Xi X, Wu Z. Downregulation of glycoprotein non-metastatic melanoma protein B prevents high glucose-induced angiogenesis in diabetic retinopathy. Mol Cell Biochem 2022; 478:697-706. [PMID: 36036335 DOI: 10.1007/s11010-022-04537-7] [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: 03/22/2022] [Accepted: 08/04/2022] [Indexed: 11/30/2022]
Abstract
Diabetic retinopathy (DR), a microvascular complication characterized by abnormal angiogenesis, is the most common reason for irreversible blindness. Glycoprotein non-metastatic melanoma protein B (GPNMB), as a transmembrane protein, was found to be associated with angiogenesis. This study aims to investigate the role of GPNMB in DR. The levels of GPNMB and Integrin β1 were detected by real-time PCR and western blot and were found to be increased in human retinal microvascular endothelial cells (HRMECs) with high glucose (HG, 25 mmol/L) treatment. Knockdown of GPNMB was mediated by lentivirus carrying shRNA targeting GPNMB in vivo and in vitro. Functional experiments, including cell counting kit-8 (CCK-8), scratch, and tube formation assays, showed the anti-proliferative, anti-migrative, and anti-angiogenic roles of GPNMB knockdown in HRMECs using the lentivirus system following HG challenge. Additionally, increased GPNMB levels were detected in the retina of DR rats induced by a single intraperitoneal injection of streptozotocin (60 mg/kg) using real-time PCR, western blot, and immunofluorescence assays. Downregulation of GPNMB inhibited the angiogenesis and vascular endothelial growth factor production in the retina of rats with DR. Furthermore, overexpression of Integrin β1 led to increased angiogenesis in DR. Integrin β1 was indicated as a target protein of GPNMB. Upregulated-Integrin β1 restored GPNMB knockdown-induced inhibition of cell viability, migration, and tube formation in HRMECs. In conclusion, we provide evidence for the angiogenic role of GPNMB and demonstrate that silencing GPNMB may represent a therapeutic potential in the treatment of DR.
Collapse
Affiliation(s)
- Tingyu Qin
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, Henan, People's Republic of China.
| | - Xiangying Xi
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, Henan, People's Republic of China
| | - Zhipeng Wu
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
22
|
Hu M, Zhang X, Hu C, Teng T, Tang QZ. A brief overview about the adipokine: Isthmin-1. Front Cardiovasc Med 2022; 9:939757. [PMID: 35958402 PMCID: PMC9360543 DOI: 10.3389/fcvm.2022.939757] [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: 05/09/2022] [Accepted: 06/30/2022] [Indexed: 11/24/2022] Open
Abstract
Isthmin-1 is a secreted protein with multiple capability; however, it truly attracts our attention since the definition as an adipokine in 2021, which exerts indispensable roles in various pathophysiological processes through the endocrine or autocrine manners. In this review, we summarize recent knowledge of isthmin-1, including its distribution, structure, receptor and potential function.
Collapse
Affiliation(s)
- Min Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Xin Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Can Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Teng Teng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
- *Correspondence: Qi-Zhu Tang
| |
Collapse
|
23
|
Son JA, Lee SK, Park J, Jung MJ, An SE, Yang HJ, Son SH, Kim KR, Park KK, Chung WY. Platycodin D Inhibits Vascular Endothelial Growth Factor-Induced Angiogenesis by Blocking the Activation of Mitogen-Activated Protein Kinases and the Production of Interleukin-8. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:1645-1661. [PMID: 35848124 DOI: 10.1142/s0192415x22500690] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Platycodin D is a major constituent in the root of Platycodon grandiflorum and has diverse pharmacologic activities, including anti-inflammatory, anti-allergic, and antitumor activities. Vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8) are potent angiogenic factors and contribute to tumor angiogenesis by directly and indirectly promoting angiogenic processes, including the proliferation, adhesion, migration, and tube formation of endothelial cells. Here, we found that platycodin D at noncytotoxic concentrations inhibited VEGF-induced proliferation, adhesion to the extracellular matrix proteins fibronectin and vitronectin, chemotactic motility, and tube formation of human umbilical vein endothelial cells (HUVECs). Platycodin D reduced the phosphorylation of extracellular signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK) and the secretion of IL-8 in VEGF-stimulated HUVECs. Moreover, platycodin D inhibited tube formation and the phosphorylation of ERK and p38 in IL-8-stimulated HUVECs. The in vitro anti-angiogenic activity of platycodin D was confirmed by in vivo experimental models. Platycodin D inhibited the formation of new blood vessels into mouse Matrigel plugs with VEGF or IL-8. In mice injected with MDA-MB-231 human breast cancer cells, orally administered platycodin D inhibited tumor growth, the number of CD34 [Formula: see text]vessels, and the expression of VEGF and IL-8. Taken together, platycodin D directly and indirectly prevents VEGF-induced and IL-8-induced angiogenesis by blocking the activation of mitogen-activated protein kinases (MAPKs). Platycodin D may be beneficial for the prevention or treatment of tumor angiogenesis and angiogenesis-related human diseases.
Collapse
Affiliation(s)
- Ju-Ah Son
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- BK21 FOUR Project, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
| | - Sun Kyoung Lee
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- BK21 FOUR Project, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
| | - Junhee Park
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- BK21 FOUR Project, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
| | - Min Ju Jung
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- BK21 FOUR Project, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- Department of Applied Life Science, The Graduate School, Yonsei University, Seoul 03722, Republic of Korea
| | - So-Eun An
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- BK21 FOUR Project, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- Department of Applied Life Science, The Graduate School, Yonsei University, Seoul 03722, Republic of Korea
| | - Hye Ji Yang
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- BK21 FOUR Project, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- Department of Applied Life Science, The Graduate School, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung Hwa Son
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- BK21 FOUR Project, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
| | - Ki Rim Kim
- Department of Dental Hygiene, College of Science and Engineering, Kyungpook National University, Sangju 37224, Republic of Korea
| | - Kwang-Kyun Park
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- BK21 FOUR Project, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
| | - Won-Yoon Chung
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- BK21 FOUR Project, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
- Department of Applied Life Science, The Graduate School, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
24
|
Urbanczyk M, Zbinden A, Schenke-Layland K. Organ-specific endothelial cell heterogenicity and its impact on regenerative medicine and biomedical engineering applications. Adv Drug Deliv Rev 2022; 186:114323. [PMID: 35568103 DOI: 10.1016/j.addr.2022.114323] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/23/2022] [Accepted: 05/05/2022] [Indexed: 02/08/2023]
Abstract
Endothelial cells (ECs) are a key cellular component of the vascular system as they form the inner lining of the blood vessels. Recent findings highlight that ECs express extensive phenotypic heterogenicity when following the vascular tree from the major vasculature down to the organ capillaries. However, in vitro models, used for drug development and testing, or to study the role of ECs in health and disease, rarely acknowledge this EC heterogenicity. In this review, we highlight the main differences between different EC types, briefly summarize their different characteristics and focus on the use of ECs in in vitro models. We introduce different approaches on how ECs can be utilized in co-culture test systems in the field of brain, pancreas, and liver research to study the role of the endothelium in health and disease. Finally, we discuss potential improvements to current state-of-the-art in vitro models and future directions.
Collapse
|
25
|
Halder SK, Sapkota A, Milner R. The impact of genetic manipulation of laminin and integrins at the blood-brain barrier. Fluids Barriers CNS 2022; 19:50. [PMID: 35690759 PMCID: PMC9188059 DOI: 10.1186/s12987-022-00346-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/18/2022] [Indexed: 12/26/2022] Open
Abstract
Blood vessels in the central nervous system (CNS) are unique in having high electrical resistance and low permeability, which creates a selective barrier protecting sensitive neural cells within the CNS from potentially harmful components in the blood. The molecular basis of this blood–brain barrier (BBB) is found at the level of endothelial adherens and tight junction protein complexes, extracellular matrix (ECM) components of the vascular basement membrane (BM), and the influence of adjacent pericytes and astrocyte endfeet. Current evidence supports the concept that instructive cues from the BBB ECM are not only important for the development and maturation of CNS blood vessels, but they are also essential for the maintenance of vascular stability and BBB integrity. In this review, we examine the contributions of one of the most abundant ECM proteins, laminin to BBB integrity, and summarize how genetic deletions of different laminin isoforms or their integrin receptors impact BBB development, maturation, and stability.
Collapse
Affiliation(s)
- Sebok K Halder
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, Suite 200, San Diego, CA, 92121, USA
| | - Arjun Sapkota
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, Suite 200, San Diego, CA, 92121, USA
| | - Richard Milner
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, Suite 200, San Diego, CA, 92121, USA.
| |
Collapse
|
26
|
Sengupta S, Mondal M, Prasasvi KR, Mukherjee A, Magod P, Urbach S, Friedmann-Morvinski D, Marin P, Somasundaram K. Differentiated glioma cell-derived Fibromodulin activates Integrin-dependent Notch signaling in endothelial cells to promote tumor angiogenesis and growth. eLife 2022; 11:78972. [PMID: 35642785 PMCID: PMC9259034 DOI: 10.7554/elife.78972] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/29/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer stem cells (CSCs) alone can initiate and maintain tumors, but the function of non-cancer stem cells (non-CSCs) that form the tumor bulk remains poorly understood. Proteomic analysis showed a higher abundance of the extracellular matrix small leucine-rich proteoglycan fibromodulin (FMOD) in the conditioned medium of differentiated glioma cells (DGCs), the equivalent of glioma non-CSCs, compared to that of glioma stem-like cells (GSCs). DGCs silenced for FMOD fail to cooperate with co-implanted GSCs to promote tumor growth. FMOD downregulation neither affects GSC growth and differentiation nor DGC growth and reprogramming in vitro. DGC-secreted FMOD promotes angiogenesis by activating integrin-dependent Notch signaling in endothelial cells. Furthermore, conditional silencing of FMOD in newly generated DGCs in vivo inhibits the growth of GSC-initiated tumors due to poorly developed vasculature and increases mouse survival. Collectively, these findings demonstrate that DGC-secreted FMOD promotes glioma tumor angiogenesis and growth through paracrine signaling in endothelial cells and identifies a DGC-produced protein as a potential therapeutic target in glioma.
Collapse
Affiliation(s)
- Shreoshi Sengupta
- Department of Microbiology and Cell Biology, Indian Institute of Science Bangalore, Bangalore, India
| | - Mainak Mondal
- Department of Microbiology and Cell Biology, Indian Institute of Science Bangalore, Bangalore, India
| | - Kaval Reddy Prasasvi
- Department of Microbiology and Cell Biology, Indian Institute of Science Bangalore, Bangalore, India
| | - Arani Mukherjee
- Department of Microbiology and Cell Biology, Indian Institute of Science Bangalore, Bangalore, India
| | - Prerna Magod
- School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | - Serge Urbach
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | | | - Philippe Marin
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Kumaravel Somasundaram
- Department of Microbiology and Cell Biology, Indian Institute of Science Bangalore, Bangalore, India
| |
Collapse
|
27
|
Zhang J, Wang H, Yuan C, Wu J, Xu J, Chen S, Zhang C, He Y. ITGAL as a Prognostic Biomarker Correlated With Immune Infiltrates in Gastric Cancer. Front Cell Dev Biol 2022; 10:808212. [PMID: 35399517 PMCID: PMC8987306 DOI: 10.3389/fcell.2022.808212] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 02/14/2022] [Indexed: 12/24/2022] Open
Abstract
Integrin alpha L (ITGAL) is a member of the integrin family in which the abnormal expression is linked with carcinogenesis and immune regulation. However, the relation between ITGAL and the prognosis of gastric cancer (GC) and tumor-infiltrating lymphocytes (TILs) are not well understood. The differential expressions of ITGAL in human tumors and the clinical prognosis in GC were systematically analyzed via multiple databases including Gene Expression Profiling Interaction Analysis (GEPIA), UALCAN, Tumor Immune Estimation Resource (TIMER), and Kaplan–Meier (KM) plotter. TIMER, GEPIA, and TISIDB databases were used to comprehensively investigate the correlation between ITGAL and tumor infiltration immune cells. Also, further results were investigated by immunohistochemistry, qRT-PCR, and Western blot. We found that ITGAL expression in GC samples was considerably increased than in peritumor samples. Sample type, subgroup, cancer stage, lymphatic node stage, and worse survival were strongly related to high ITGAL expression. Moreover, upregulated ITGAL expression was strongly connected with immunomodulators, chemokines, and infiltrating levels of CD8+, CD4+ T cell, B cell, monocyte, neutrophil, macrophage, T-cell regulatory, NK cell, and myeloid dendritic cell in stomach adenocarcinoma (STAD). Specifically, immunohistochemistry and bioinformatic analysis showed that ITGAL expression was shown to have strong relationships with various immunological marker sets including PD1 (T-cell exhaustion marker). In conclusion, ITGAL is a prognostic biomarker for GC patients. It might regulate tumor immune microenvironment leading to poor prognosis. Furthermore, studies are essential to explore therapeutic targeting ITGAL.
Collapse
Affiliation(s)
- Junchang Zhang
- Department of Center for Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Han Wang
- Department of Center for Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Cheng Yuan
- Department of Center for Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jing Wu
- Department of Center for Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jiannan Xu
- Department of Center for Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Songyao Chen
- Department of Center for Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Changhua Zhang
- Department of Center for Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- *Correspondence: Changhua Zhang, ; Yulong He,
| | - Yulong He
- Department of Center for Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Changhua Zhang, ; Yulong He,
| |
Collapse
|
28
|
Van Petten de Vasconcelos Azevedo F, Lopes DS, Zóia MAP, Correia LIV, Saito N, Fonseca BB, Polloni L, Teixeira SC, Goulart LR, de Melo Rodrigues Ávila V. A New Approach to Inhibiting Triple-Negative Breast Cancer: In Vitro, Ex Vivo and In Vivo Antiangiogenic Effect of BthTx-II, a PLA 2-Asp-49 from Bothrops jararacussu Venom. Biomolecules 2022; 12:258. [PMID: 35204758 PMCID: PMC8961627 DOI: 10.3390/biom12020258] [Citation(s) in RCA: 4] [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: 09/30/2021] [Revised: 12/28/2021] [Accepted: 01/05/2022] [Indexed: 12/10/2022] Open
Abstract
Phospholipases A2 (PLA2) represent a superfamily of enzymes widely distributed in living organisms, with a broad spectrum of pharmacological activities and therapeutic potential. Anti-angiogenic strategies have become one of the main tools in fighting cancer. In this sense, the present work reports the inhibition of tumor angiogenesis induced by Asp-49 BthTX-II using in vitro, ex vivo and in vivo approaches. We demonstrate that BthTx-II inhibited cell adhesion, proliferation, and migration of human umbilical vein endothelial cells (HUVEC), as well as caused a reduction in the levels of endothelial growth factor (VEGF) during in vitro angiogenesis assays. BthTx-II was also able to inhibit the sprouting angiogenic process, by the ex vivo germination assay of the aortic ring; in addition, this toxin inhibited the migration and proliferation of HUVEC in co-culture with triple-negative breast cancer cells (e.g., MDA-MB-231 cells). Finally, in vivo tumor suppression and anti-angiogenic activities were analyzed using MDA-MB-231 cells with Matrigel injected into the chorioallantoic membrane of chicken embryo (CAM) for 7 days treatment with BthTx-II, showing a considerable reduction in vessel caliber, on the size and weight of tumors. Together, these results suggest an important antiangiogenic and antitumor role for BthTx-II, as a potential prototype for the development of new tools and antitumor drugs in cancer therapy.
Collapse
Affiliation(s)
- Fernanda Van Petten de Vasconcelos Azevedo
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (L.I.V.C.); (L.P.)
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (M.A.P.Z.); (N.S.); (L.R.G.)
| | - Daiana Silva Lopes
- Multidisciplinary Institute of Health, Federal University of Bahia, Vitoria da Conquista, Salvador 40170-110, BA, Brazil;
| | - Mariana Alves Pereira Zóia
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (M.A.P.Z.); (N.S.); (L.R.G.)
| | - Lucas Ian Veloso Correia
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (L.I.V.C.); (L.P.)
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (M.A.P.Z.); (N.S.); (L.R.G.)
| | - Natieli Saito
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (M.A.P.Z.); (N.S.); (L.R.G.)
| | | | - Lorena Polloni
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (L.I.V.C.); (L.P.)
| | - Samuel Cota Teixeira
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil;
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (M.A.P.Z.); (N.S.); (L.R.G.)
| | - Veridiana de Melo Rodrigues Ávila
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlandia, Uberlândia 38408-100, MG, Brazil; (L.I.V.C.); (L.P.)
| |
Collapse
|
29
|
Pérez LA, Leyton L, Valdivia A. Thy-1 (CD90), Integrins and Syndecan 4 are Key Regulators of Skin Wound Healing. Front Cell Dev Biol 2022; 10:810474. [PMID: 35186924 PMCID: PMC8851320 DOI: 10.3389/fcell.2022.810474] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
Acute skin wound healing is a multistage process consisting of a plethora of tightly regulated signaling events in specialized cells. The Thy-1 (CD90) glycoprotein interacts with integrins and the heparan sulfate proteoglycan syndecan 4, generating a trimolecular complex that triggers bi-directional signaling to regulate diverse aspects of the wound healing process. These proteins can act either as ligands or receptors, and they are critical for the successful progression of wound healing. The expression of Thy-1, integrins, and syndecan 4 is controlled during the healing process, and the lack of expression of any of these proteins results in delayed wound healing. Here, we review and discuss the roles and regulatory events along the stages of wound healing that support the relevance of Thy-1, integrins, and syndecan 4 as crucial regulators of skin wound healing.
Collapse
Affiliation(s)
- Leonardo A. Pérez
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
- *Correspondence: Lisette Leyton, ; Alejandra Valdivia,
| | - Alejandra Valdivia
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
- *Correspondence: Lisette Leyton, ; Alejandra Valdivia,
| |
Collapse
|
30
|
Kang M, Yao Y. Laminin regulates oligodendrocyte development and myelination. Glia 2021; 70:414-429. [PMID: 34773273 DOI: 10.1002/glia.24117] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 11/08/2022]
Abstract
Oligodendrocytes are the cells that myelinate axons and provide trophic support to neurons in the CNS. Their dysfunction has been associated with a group of disorders known as demyelinating diseases, such as multiple sclerosis. Oligodendrocytes are derived from oligodendrocyte precursor cells, which differentiate into premyelinating oligodendrocytes and eventually mature oligodendrocytes. The development and function of oligodendrocytes are tightly regulated by a variety of molecules, including laminin, a major protein of the extracellular matrix. Accumulating evidence suggests that laminin actively regulates every aspect of oligodendrocyte biology, including survival, migration, proliferation, differentiation, and myelination. How can laminin exert such diverse functions in oligodendrocytes? It is speculated that the distinct laminin isoforms, laminin receptors, and/or key signaling molecules expressed in oligodendrocytes at different developmental stages are the reasons. Understanding molecular targets and signaling pathways unique to each aspect of oligodendrocyte biology will enable more accurate manipulation of oligodendrocyte development and function, which may have implications in the therapies of demyelinating diseases. Here in this review, we first introduce oligodendrocyte biology, followed by the expression of laminin and laminin receptors in oligodendrocytes and other CNS cells. Next, the functions of laminin in oligodendrocyte biology, including survival, migration, proliferation, differentiation, and myelination, are discussed in detail. Last, key questions and challenges in the field are discussed. By providing a comprehensive review on laminin's roles in OL lineage cells, we hope to stimulate novel hypotheses and encourage new research in the field.
Collapse
Affiliation(s)
- Minkyung Kang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Yao Yao
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
31
|
Wu SC, Kuo PJ, Rau CS, Huang LH, Lin CW, Wu YC, Wu CJ, Tsai CW, Hsieh TM, Liu HT, Huang CY, Hsieh CH. Increased Angiogenesis by Exosomes Secreted by Adipose-Derived Stem Cells upon Lipopolysaccharide Stimulation. Int J Mol Sci 2021; 22:8877. [PMID: 34445582 PMCID: PMC8396299 DOI: 10.3390/ijms22168877] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/19/2022] Open
Abstract
Exosomes secreted by adipose-derived stem cells (ADSCs) enhance angiogenesis and wound healing. However, in clinical settings, wounds may be infected by various bacteria or pathogens. We investigated whether human ADSCs stimulated with lipopolysaccharide (LPS) secrete exosomes (ADSC-LPS-exo) that augment the angiogenesis of human umbilical vein endothelial cells (HUVECs). ExoQuick-TC exosome precipitation solution was used to purify exosomes from human ADSC culture media in the presence or absence of 1 µg/mL LPS treatment for 24 h. The uptake of ADSC-LPS-exo significantly induced the activation of cAMP response element binding protein (CREB), activating protein 1 (AP-1), and nuclear factor-κB (NF-κB) signaling pathways and increased the migration of and tube formation in HUVECs. RNA interference with CREB, AP-1, or NF-κB1 significantly reduced the migration of and tube formation in HUVECs treated with ADSC-LPS-exo. An experiment with an antibody array for 25 angiogenesis-related proteins revealed that only interleukin-8 expression was significantly upregulated in HUVECs treated with ADSC-LPS-exo. In addition, proteomic analysis revealed that eukaryotic translation initiation factor 4E, amyloid beta A4 protein, integrin beta-1, and ras-related C3 botulinum toxin substrate 1 may be potential candidates involved in ADSC-LPS-exo-mediated enhanced angiogenesis.
Collapse
Affiliation(s)
- Shao-Chun Wu
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan;
| | - Pao-Jen Kuo
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Cheng-Shyuan Rau
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (C.-S.R.); (L.-H.H.)
| | - Lien-Hung Huang
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (C.-S.R.); (L.-H.H.)
| | - Chia-Wei Lin
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Yi-Chan Wu
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Chia-Jung Wu
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Chia-Wen Tsai
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Ting-Min Hsieh
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (T.-M.H.); (H.-T.L.)
| | - Hang-Tsung Liu
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (T.-M.H.); (H.-T.L.)
| | - Chun-Ying Huang
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (T.-M.H.); (H.-T.L.)
| | - Ching-Hua Hsieh
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| |
Collapse
|
32
|
Barbera S, Lugano R, Pedalina A, Mongiat M, Santucci A, Tosi GM, Dimberg A, Galvagni F, Orlandini M. The C-type lectin CD93 controls endothelial cell migration via activation of the Rho family of small GTPases. Matrix Biol 2021; 99:1-17. [PMID: 34062268 DOI: 10.1016/j.matbio.2021.05.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023]
Abstract
Endothelial cell migration is essential to angiogenesis, enabling the outgrowth of new blood vessels both in physiological and pathological contexts. Migration requires the activation of several signaling pathways, the elucidation of which expands the opportunity to develop new drugs to be used in antiangiogenic therapy. In the proliferating endothelium, the interaction between the transmembrane glycoprotein CD93 and the extracellular matrix activates signaling pathways that regulate cell adhesion, migration, and vascular maturation. Here we identify a pathway, comprising CD93, the adaptor proteins Cbl and Crk, and the small GTPases Rac1, Cdc42, and RhoA, which we propose acts as a regulator of cytoskeletal movements responsible for endothelial cell migration. In this framework, phosphorylation of Cbl on tyrosine 774 leads to the interaction with Crk, which acts as a downstream integrator in the CD93-mediated signaling regulating cell polarity and migration. Moreover, confocal microscopy analyses of GTPase biosensors show that CD93 drives coordinated activation of Rho-proteins at the cell edge of migratory endothelial cells. In conclusion, together with the demonstration of the key contribution of CD93 to the migratory process in living cells, these findings suggest that the signaling triggered by CD93 converges to the activation and modulation of the Rho GTPase signaling pathways regulating cell dynamics.
Collapse
Affiliation(s)
- Stefano Barbera
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Italy; Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - Roberta Lugano
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - Alessia Pedalina
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Italy
| | - Maurizio Mongiat
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Annalisa Santucci
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Italy
| | - Gian Marco Tosi
- Department of Medicine, Surgery and Neuroscience, Ophthalmology Unit, University of Siena, Italy
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - Federico Galvagni
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Italy
| | - Maurizio Orlandini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Italy.
| |
Collapse
|
33
|
Okamoto T, Park EJ, Kawamoto E, Usuda H, Wada K, Taguchi A, Shimaoka M. Endothelial connexin-integrin crosstalk in vascular inflammation. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166168. [PMID: 33991620 DOI: 10.1016/j.bbadis.2021.166168] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/18/2021] [Accepted: 05/02/2021] [Indexed: 02/06/2023]
Abstract
Cardiovascular diseases including blood vessel disorders represent a major cause of death globally. The essential roles played by local and systemic vascular inflammation in the pathogenesis of cardiovascular diseases have been increasingly recognized. Vascular inflammation triggers the aberrant activation of endothelial cells, which leads to the functional and structural abnormalities in vascular vessels. In addition to humoral mediators such as pro-inflammatory cytokines and prostaglandins, the alteration of physical and mechanical microenvironment - including vascular stiffness and shear stress - modify the gene expression profiles and metabolic profiles of endothelial cells via mechano-transduction pathways, thereby contributing to the pathogenesis of vessel disorders. Notably, connexins and integrins crosstalk each other in response to the mechanical stress, and, thereby, play an important role in regulating the mechano-transduction of endothelial cells. Here, we provide an overview on how the inter-play between connexins and integrins in endothelial cells unfold during the mechano-transduction in vascular inflammation.
Collapse
Affiliation(s)
- Takayuki Okamoto
- Department of Pharmacology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-city, Shimane 693-8501, Japan.
| | - Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan
| | - Eiji Kawamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan; Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan
| | - Haruki Usuda
- Department of Pharmacology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-city, Shimane 693-8501, Japan
| | - Koichiro Wada
- Department of Pharmacology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-city, Shimane 693-8501, Japan
| | - Akihiko Taguchi
- Department of Regenerative Medicine Research, Foundation for Biomedical Research and Innovation at Kobe, 2-2 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan.
| |
Collapse
|
34
|
Targeting RGD-binding integrins as an integrative therapy for diabetic retinopathy and neovascular age-related macular degeneration. Prog Retin Eye Res 2021; 85:100966. [PMID: 33775825 DOI: 10.1016/j.preteyeres.2021.100966] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/15/2021] [Accepted: 03/19/2021] [Indexed: 12/14/2022]
Abstract
Integrins are a class of transmembrane receptors that are involved in a wide range of biological functions. Dysregulation of integrins has been implicated in many pathological processes and consequently, they are attractive therapeutic targets. In the ophthalmology arena, there is extensive evidence suggesting that integrins play an important role in diabetic retinopathy (DR), age-related macular degeneration (AMD), glaucoma, dry eye disease and retinal vein occlusion. For example, there is extensive evidence that arginyl-glycyl-aspartic acid (Arg-Gly-Asp; RGD)-binding integrins are involved in key disease hallmarks of DR and neovascular AMD (nvAMD), specifically inflammation, vascular leakage, angiogenesis and fibrosis. Based on such evidence, drugs that engage integrin-linked pathways have received attention for their potential to block all these vision-threatening pathways. This review focuses on the pathophysiological role that RGD-binding integrins can have in complex multifactorial retinal disorders like DR, diabetic macular edema (DME) and nvAMD, which are leading causes of blindness in developed countries. Special emphasis will be given on how RGD-binding integrins can modulate the intricate molecular pathways and regulate the underlying pathological mechanisms. For instance, the interplay between integrins and key molecular players such as growth factors, cytokines and enzymes will be summarized. In addition, recent clinical advances linked to targeting RGD-binding integrins in the context of DME and nvAMD will be discussed alongside future potential for limiting progression of these diseases.
Collapse
|
35
|
Dhavalikar P, Robinson A, Lan Z, Jenkins D, Chwatko M, Salhadar K, Jose A, Kar R, Shoga E, Kannapiran A, Cosgriff-Hernandez E. Review of Integrin-Targeting Biomaterials in Tissue Engineering. Adv Healthc Mater 2020; 9:e2000795. [PMID: 32940020 PMCID: PMC7960574 DOI: 10.1002/adhm.202000795] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/27/2020] [Indexed: 12/12/2022]
Abstract
The ability to direct cell behavior has been central to the success of numerous therapeutics to regenerate tissue or facilitate device integration. Biomaterial scientists are challenged to understand and modulate the interactions of biomaterials with biological systems in order to achieve effective tissue repair. One key area of research investigates the use of extracellular matrix-derived ligands to target specific integrin interactions and induce cellular responses, such as increased cell migration, proliferation, and differentiation of mesenchymal stem cells. These integrin-targeting proteins and peptides have been implemented in a variety of different polymeric scaffolds and devices to enhance tissue regeneration and integration. This review first presents an overview of integrin-mediated cellular processes that have been identified in angiogenesis, wound healing, and bone regeneration. Then, research utilizing biomaterials are highlighted with integrin-targeting motifs as a means to direct these cellular processes to enhance tissue regeneration. In addition to providing improved materials for tissue repair and device integration, these innovative biomaterials provide new tools to probe the complex processes of tissue remodeling in order to enhance the rational design of biomaterial scaffolds and guide tissue regeneration strategies.
Collapse
Affiliation(s)
- Prachi Dhavalikar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrew Robinson
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ziyang Lan
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Dana Jenkins
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Malgorzata Chwatko
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Karim Salhadar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Anupriya Jose
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ronit Kar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Erik Shoga
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Aparajith Kannapiran
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | | |
Collapse
|
36
|
Shastri MD, Chong WC, Dua K, Peterson GM, Patel RP, Mahmood MQ, Tambuwala M, Chellappan DK, Hansbro NG, Shukla SD, Hansbro PM. Emerging concepts and directed therapeutics for the management of asthma: regulating the regulators. Inflammopharmacology 2020; 29:15-33. [PMID: 33152094 DOI: 10.1007/s10787-020-00770-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/17/2020] [Indexed: 12/19/2022]
Abstract
Asthma is a common, heterogeneous and serious disease, its prevalence has steadily risen in most parts of the world, and the condition is often inadequately controlled in many patients. Hence, there is a major need for new therapeutic approaches. Mild-to-moderate asthma is considered a T-helper cell type-2-mediated inflammatory disorder that develops due to abnormal immune responses to otherwise innocuous allergens. Prolonged exposure to allergens and persistent inflammation results in myofibroblast infiltration and airway remodelling with mucus hypersecretion, airway smooth muscle hypertrophy, and excess collagen deposition. The airways become hyper-responsive to provocation resulting in the characteristic wheezing and obstructed airflow experienced by patients. Extensive research has progressed the understanding of the underlying mechanisms and the development of new treatments for the management of asthma. Here, we review the basis of the disease, covering new areas such as the role of vascularisation and microRNAs, as well as associated potential therapeutic interventions utilising reports from animal and human studies. We also cover novel drug delivery strategies that are being developed to enhance therapeutic efficacy and patient compliance. Potential avenues to explore to improve the future of asthma management are highlighted.
Collapse
Affiliation(s)
- Madhur D Shastri
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Australia
| | - Wai Chin Chong
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, Australia.,Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia.,Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia.,Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Gregory M Peterson
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Australia
| | - Rahul P Patel
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Australia
| | - Malik Q Mahmood
- Faculty of Health, School of Medicine, Deakin University, Melbourne, Australia
| | - Murtaza Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Belfast, Northern Ireland, UK
| | - Dinesh K Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia.,Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia.,Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia. .,Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia. .,Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
37
|
Changchien C, Chen Y, Chang H, Chang S, Tsai W, Tsai H, Wang C, Lee H, Tsai C. Effect of malignant-associated pleural effusion on endothelial viability, motility and angiogenesis in lung cancer. Cancer Sci 2020; 111:3747-3758. [PMID: 32706142 PMCID: PMC7541005 DOI: 10.1111/cas.14584] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 12/30/2022] Open
Abstract
Malignant pleural effusion (MPE) and paramalignant pleural effusion (PPE) remain debilitating complications in lung cancer patients with poor prognosis and limited treatment options. The role of vascular endothelial cells has not been explored in the pleural environment of lung cancer. By integrating MPE and PPE as malignant-associated pleural fluid (MAPF), the current study aimed to evaluate the effect of MAPF on cell proliferation, migration and angiogenesis of HUVEC. First, increased capillaries were identified in the subpleural layer of lung adenocarcinoma. Compatible with pathological observations, the ubiquitous elevation of HUVEC survival was identified in MAPF culture regardless of the underlying cancer type, the driver gene mutation, prior treatments and evidence of malignant cells in pleural fluid. Moreover, MAPF enhanced HUVEC motility with the formation of lamellipodia and filopodia and focal adhesion complex. Tube formation assay revealed angiogenic behavior with the observation of sheet-like structures. HUVEC cultured with MAPF resulted in a significant increase in MAPK phosphorylation. Accompanied with VEGFR2 upregulation in MAPF culture, there was increased expressions of p-STAT3, HIF-1α and Nf-kB. VEGF/VEGFR2 blockade regressed endothelial migration and angiogenesis but not cell proliferation. Our data indicate the angiogenic activities of MAPF on vascular endothelial cells that revealed increased pleural capillaries in lung cancer. Targeting the VEGF/VEGFR2 pathway might modulate the angiogenic propensity of MAPF in future clinical investigations.
Collapse
MESH Headings
- Aged
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Proliferation/genetics
- Cell Survival/genetics
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Female
- Human Umbilical Vein Endothelial Cells
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Lung Neoplasms/complications
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Male
- NF-kappa B/genetics
- Neovascularization, Pathologic/complications
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/pathology
- Pleural Effusion/genetics
- Pleural Effusion, Malignant/complications
- Pleural Effusion, Malignant/genetics
- Pleural Effusion, Malignant/pathology
- STAT3 Transcription Factor/genetics
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor Receptor-2/genetics
Collapse
Affiliation(s)
- Chih‐Ying Changchien
- Department of Internal MedicineTri‐Service General Hospital, National Defense Medical CenterTaipeiTaiwan
- Department of Biology and AnatomyNational Defense Medical CenterTaipeiTaiwan
| | - Ying Chen
- Department of Biology and AnatomyNational Defense Medical CenterTaipeiTaiwan
| | - Hsin‐Han Chang
- Department of Biology and AnatomyNational Defense Medical CenterTaipeiTaiwan
| | - Shan‐Yueh Chang
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineTri‐Service General Hospital, National Defense Medical CenterTaipeiTaiwan
| | - Wen‐Chiuan Tsai
- Department of PathologyTri‐Service General Hospital, National Defense Medical CenterTaipeiTaiwan
| | - Hao‐Chung Tsai
- Division of Chest MedicineDepartment of Internal MedicineTri‐Service General Hospital Songshan Branch, National Defense Medical CenterTaipeiTaiwan
| | - Chieh‐Yung Wang
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineTri‐Service General Hospital, National Defense Medical CenterTaipeiTaiwan
| | - Herng‐Sheng Lee
- Department of Pathology and Laboratory MedicineKaohsiung Veterans General HospitalKaohsiungTaiwan
| | - Chen‐Liang Tsai
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineTri‐Service General Hospital, National Defense Medical CenterTaipeiTaiwan
| |
Collapse
|
38
|
Wu A, Zhang S, Liu J, Huang Y, Deng W, Shu G, Yin G. Integrated Analysis of Prognostic and Immune Associated Integrin Family in Ovarian Cancer. Front Genet 2020; 11:705. [PMID: 32765584 PMCID: PMC7379341 DOI: 10.3389/fgene.2020.00705] [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: 04/30/2020] [Accepted: 06/10/2020] [Indexed: 12/22/2022] Open
Abstract
Human integrin receptors are important for cell-cell and cell-matrix adhesion in normal epithelial cells. Emerging evidences have indicated integrin members are involved in cancer development and progression as well. However, the expression patterns and clinical significance of the whole integrin family in ovarian cancer (OC) have not yet been well understood. In the present study, we utilized the public datasets including GEPIA, GEO, ONCOMINE, cBioPortal, Kaplan-Meier Plotter, TIMER databases, to analyze the expression and prognostic value of integrin members in OC. We found ITGA3/B4/B6/B7/B8 were abnormally overexpressed in OC; ITGA6 was good prognosis predictor in OC; ITGA3/ B4/B8 were poor prognosis predictor specially in advanced OC patients; elevated ITGA3/B4 might promote metastasis and elevated ITGA3/B8 might promote platinum resistance of OC; ITGA3 and ITGB4 might synergistically or independently regulate cell adhesion and proliferation; ITGA4/AL/AM/AX/B2/B7 showed strong correlations with various tumor immune infiltrates (TILs), especially with pro-tumor immunes cell types like monocyte, M2 macrophage and exhaustion T cells infiltration; ITGAL/AM/B2/B7 and residing memory CD8+ T cells marker ITGAE were specially associated with early OC patients outcome. Our results implied that ITGA3/B4 were important prognostic markers of advanced OC, ITGAL/AM/ B2/B7 were immune associated prognosis markers of early OC, together they might render important therapeutic targets for OC.
Collapse
Affiliation(s)
- Anqi Wu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Sai Zhang
- Deparment of Pathology, School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Jiaqi Liu
- Deparment of Pathology, School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Yifeng Huang
- Department of Anesthesia, School of Medicine, Central South University, Changsha, China
| | - Wenyu Deng
- Departmemt of Nursing, School of Nursing, Central South University, Changsha, China
| | - Guang Shu
- Deparment of Pathology, School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Gang Yin
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China.,Deparment of Pathology, School of Basic Medicine Sciences, Central South University, Changsha, China
| |
Collapse
|
39
|
Sundararaman A, Fukushima Y, Norman JC, Uemura A, Mellor H. RhoJ Regulates α5β1 Integrin Trafficking to Control Fibronectin Remodeling during Angiogenesis. Curr Biol 2020; 30:2146-2155.e5. [PMID: 32302585 DOI: 10.1016/j.cub.2020.03.042] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 02/03/2020] [Accepted: 03/17/2020] [Indexed: 01/24/2023]
Abstract
Rho guanosine triphosphatases (GTPases) are master regulators of cell shape and cell movement [1]. The archetypal family members RhoA, Rac1, and Cdc42 arose early in eukaryotic evolution and coordinate a diverse range of cell morphologies and migrations. Evolution of the vertebrates was paralleled by expansion of this family through gene duplication. Emergence of an adaptive immune system and more complex neural systems presented new roles for Rho GTPases, filled by new family members. Cdc42 underwent gene duplication to produce two related proteins-RhoQ and RhoJ [2]. RhoQ is active in neural dynamics; however, RhoJ is highly expressed in endothelial cells under control of the endothelial-specific promoter ERG [3, 4]. RhoJ is required for angiogenesis [5, 6] and has multiple roles in this process [7, 8]. We recently demonstrated that RhoJ regulates the endosomal trafficking of podocalyxin during angiogenesis to control lumen formation [9]. Here, we use vesicle purification and proteomic analysis to identify the endothelial targets of RhoJ-mediated trafficking. We identify α5β1 integrin as a major RhoJ cargo and show that RhoJ regulates the intracellular trafficking of active α5β1 integrin in endothelial cells to repress fibronectin fibrillogenesis. Accordingly, mice lacking RhoJ show deregulated deposition of fibronectin around vessels during developmental angiogenesis. Intriguingly, we show that RhoJ acts in opposition to Cdc42 in this process through competition for a shared partner, PAK3. These studies identify a critical role for RhoJ in matrix remodeling during blood vessel formation and demonstrate a functional interrelationship between RhoJ and its evolutionary parent.
Collapse
Affiliation(s)
| | - Yoko Fukushima
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Jim C Norman
- CRUK Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - Akiyoshi Uemura
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Harry Mellor
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol BS8 1TD, UK.
| |
Collapse
|
40
|
Apolipoprotein(a), an enigmatic anti-angiogenic glycoprotein in human plasma: A curse or cure? Pharmacol Res 2020; 158:104858. [PMID: 32430285 DOI: 10.1016/j.phrs.2020.104858] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 04/09/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023]
Abstract
Angiogenesis is a finely co-ordinated, multi-step developmental process of the new vascular structure. Even though angiogenesis is regularly occurring in physiological events such as embryogenesis, in adults, it is restricted to specific tissue sites where rapid cell-turnover and membrane synthesis occurs. Both excessive and insufficient angiogenesis lead to vascular disorders such as cancer, ocular diseases, diabetic retinopathy, atherosclerosis, intra-uterine growth restriction, ischemic heart disease, stroke etc. Occurrence of altered lipid profile and vascular lipid deposition along with vascular disorders is a hallmark of impaired angiogenesis. Among lipoproteins, lipoprotein(a) needs special attention due to the presence of a multi-kringle protein subunit, apolipoprotein(a) [apo(a)], which is structurally homologous to many naturally occurring anti-angiogenic proteins such as plasminogen and angiostatin. Researchers have constructed different recombinant forms of apo(a) (rhLK68, rhLK8, RHACK2, KV-11, and AU-6) and successfully exploited its potential to inhibit unwanted angiogenesis during tumor metastasis and retinal neovascularization. Similar to naturally occurring anti-angiogenic proteins, apo(a) can directly interfere with angiogenic signaling pathways. Besides this, apo(a) can also exert its anti-angiogenic effect indirectly by inducing endothelial cell apoptosis, by inhibiting endothelial progenitor cell functions or by upregulating nuclear factors in endothelial cells via apo(a)-bound oxPLs. However, the impact of the anti-angiogenic potential of native apo(a) during physiological angiogenesis in embryos and wounded tissues is not yet explored. In this context, we review the studies so far done to demonstrate the anti-angiogenic activity of apo(a) and the recent developments in using apo(a) as a therapeutic agent to treat impaired angiogenesis during vascular disorders, with emphasis on the gaps in the literature.
Collapse
|
41
|
A Combined Proteomics and Bioinformatics Approach Reveals Novel Signaling Pathways and Molecular Targets After Intracerebral Hemorrhage. J Mol Neurosci 2020; 70:1186-1197. [PMID: 32170712 PMCID: PMC7359136 DOI: 10.1007/s12031-020-01526-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/28/2020] [Indexed: 12/18/2022]
Abstract
Intracerebral hemorrhage (ICH) is a non-traumatic cerebrovascular disorder with very high morbidity and mortality and regarded as one of the deadliest stroke subtypes. Notably, there is no effective treatment for ICH. Despite an overall increase in preclinical studies, the pathophysiology of ICH is complex and remains enigmatic. To this end, ICH was induced in male CD-1 mice and the ipsilateral brain tissue was characterized in an unbiased manner using a combination of proteomics and bioinformatics approaches. A total of 4833 proteins were revealed by quantitative proteomic analysis. Of those, 207 proteins exhibited significantly altered expression after ICH in comparison to sham. It was found that 46 proteins were significantly upregulated and 161 proteins were significantly downregulated after ICH compared to sham. The quantitative proteomics approach combined with bioinformatics revealed several novel molecular targets (cyclin-dependent-like kinase 5, E3 ubiquitin-protein ligase, protein phosphatase 2A-alpha, protein phosphatase 2A-beta, serine/threonine-protein kinase PAK1, alpha-actinin-4, calpain-8, axin-1, NCK1, and septin-4), and related signaling pathways, which could play roles in secondary brain injury and long-term neurobehavioral outcomes after ICH warranting further investigation.
Collapse
|
42
|
Yang Z, Yue Z, Ma X, Xu Z. Calcium Homeostasis: A Potential Vicious Cycle of Bone Metastasis in Breast Cancers. Front Oncol 2020; 10:293. [PMID: 32211326 PMCID: PMC7076168 DOI: 10.3389/fonc.2020.00293] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/19/2020] [Indexed: 12/12/2022] Open
Abstract
Cancers have been considered as one of the most severe health problems in the world. Efforts to elucidate the cancer progression reveal the importance of bone metastasis for tumor malignancy, one of the leading causes for high mortality rate. Multiple cancers develop bone metastasis, from which breast cancers exhibit the highest rate and have been well-recognized. Numerous cells and environmental factors have been believed to synergistically facilitate bone metastasis in breast cancers, from which breast cancer cells, osteoclasts, osteoblasts, and their produced cytokines have been well-recognized to form a vicious cycle that aggravates tumor malignancy. Except the cytokines or chemokines, calcium ions are another element largely released from bones during bone metastasis that leads to hypercalcemia, however, have not been well-characterized yet in modulation of bone metastasis. Calcium ions act as a type of unique second messenger that exhibits omnipotent functions in numerous cells, including tumor cells, osteoclasts, and osteoblasts. Calcium ions cannot be produced in the cells and are dynamically fluxed among extracellular calcium pools, intracellular calcium storages and cytosolic calcium signals, namely calcium homeostasis, raising a possibility that calcium ions released from bone during bone metastasis would further enhance bone metastasis and aggravate tumor progression via the vicious cycle due to abnormal calcium homeostasis in breast cancer cells, osteoclasts and osteoblasts. TRPs, VGCCs, SOCE, and P2Xs are four major calcium channels/routes mediating extracellular calcium entry and affect calcium homeostasis. Here we will summarize the overall functions of these four calcium channels in breast cancer cells, osteoclasts and osteoblasts, providing evidence of calcium homeostasis as a vicious cycle in modulation of bone metastasis in breast cancers.
Collapse
Affiliation(s)
- Zhengfeng Yang
- Shanghai Institute of Immunology Center for Microbiota & Immune Related Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiying Yue
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinrun Ma
- Shanghai Institute of Immunology Center for Microbiota & Immune Related Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenyao Xu
- Shanghai Institute of Immunology Center for Microbiota & Immune Related Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
43
|
Demin AM, Vakhrushev AV, Tumashov AA, Krasnov VP. Synthesis of glutaryl-containing derivatives of GRGD and KRGD peptides. Russ Chem Bull 2020. [DOI: 10.1007/s11172-019-2705-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
44
|
Isolation of an Anti-Tumour Disintegrin: Dabmaurin-1, a Peptide Lebein-1-Like, from Daboia mauritanica Venom. Toxins (Basel) 2020; 12:toxins12020102. [PMID: 32033352 PMCID: PMC7076848 DOI: 10.3390/toxins12020102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/28/2020] [Accepted: 01/31/2020] [Indexed: 11/17/2022] Open
Abstract
In the soft treatment of cancer tumours, consequent downregulation of the malignant tissue angiogenesis constitutes an efficient way to stifle tumour development and metastasis spreading. As angiogenesis requires integrin–promoting endothelial cell adhesion, migration, and vessel tube formation, integrins represent potential targets of new therapeutic anti–angiogenic agents. Our work is a contribution to the research of such therapeutic disintegrins in animal venoms. We report isolation of one peptide, named Dabmaurin–1, from the hemotoxic venom of snake Daboia mauritanica, and we evaluate its potential anti–tumour activity through in vitro inhibition of the human vascular endothelial cell HMECs functions involved in tumour angiogenesis. Dabmaurin–1 altered, in a dose–dependent manner, without any significant cytotoxicity, HMEC proliferation, adhesion, and their mesenchymal migration onto various extracellular matrix proteins, as well as formation of capillary–tube mimics on MatrigelTM. Via experiments involving HMEC or specific cancers cells integrins, we demonstrated that the above Dabmaurin–1 effects are possibly due to some anti–integrin properties. Dabmaurin–1 was demonstrated to recognize a broad panel of prooncogenic integrins (αvβ6, αvβ3 or αvβ5) and/or particularly involved in control of angiogenesis (α5β1, α6β4, αvβ3 or αvβ5). Furthermore, mass spectrometry and partial N–terminal sequencing of this peptide revealed, it is close to Lebein–1, a known anti–β1 disintegrin from Macrovipera lebetina venom. Therefore, our results show that if Dabmaurin–1 exhibits in vitro apparent anti–angiogenic effects at concentrations lower than 30 nM, it is likely because it acts as an anti–tumour disintegrin.
Collapse
|
45
|
Gallo E, Kelil A, Bayliss PE, Jeganathan A, Egorova O, Ploder L, Adams JJ, Giblin P, Sidhu SS. In situ antibody phage display yields optimal inhibitors of integrin α11/β1. MAbs 2020; 12:1717265. [PMID: 31980006 PMCID: PMC6999838 DOI: 10.1080/19420862.2020.1717265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/30/2019] [Accepted: 01/09/2020] [Indexed: 01/13/2023] Open
Abstract
Integrins are transmembrane multi-conformation receptors that mediate interactions with the extracellular matrix. In cancer, integrins influence metastasis, proliferation, and survival. Collagen-binding integrin-α11/β1, a marker of aggressive tumors that is involved in stroma-tumor crosstalk, may be an attractive target for anti-cancer therapeutic antibodies. We performed selections with phage-displayed synthetic antibody libraries for binding to either purified integrin-α11/β1 or in situ on live cells. The in-situ strategy yielded many diverse antibodies, and strikingly, most of these antibodies did not recognize purified integrin-α11/β1. Conversely, none of the antibodies selected for binding to purified integrin-α11/β1 were able to efficiently recognize native cell-surface antigen. Most importantly, only the in-situ selection yielded functional antibodies that were able to compete with collagen-I for binding to cell-surface integrin-α11/β1, and thus inhibited cell adhesion. In-depth characterization of a subset of in situ-derived clones as full-length immunoglobulins revealed high affinity cellular binding and inhibitory activities in the single-digit nanomolar range. Moreover, the antibodies showed high selectivity for integrin-α11/β1 with minimal cross-reactivity for close homologs. Taken together, our findings highlight the advantages of in-situ selections for generation of anti-integrin antibodies optimized for recognition and inhibition of native cell-surface proteins, and our work establishes general methods that could be extended to many other membrane proteins.
Collapse
Affiliation(s)
- Eugenio Gallo
- Department of Molecular Genetics, University of Toronto, Donnelly Centre, Toronto, Ontario, Canada
| | - Abdellali Kelil
- Department of Molecular Genetics, University of Toronto, Donnelly Centre, Toronto, Ontario, Canada
| | - Peter E. Bayliss
- Northern Biologics, Inc., Princess Margaret Cancer Research Tower, Toronto, Ontario, Canada
| | - Ajitha Jeganathan
- Northern Biologics, Inc., Princess Margaret Cancer Research Tower, Toronto, Ontario, Canada
| | - Olga Egorova
- Northern Biologics, Inc., Princess Margaret Cancer Research Tower, Toronto, Ontario, Canada
| | - Lynda Ploder
- Department of Molecular Genetics, University of Toronto, Donnelly Centre, Toronto, Ontario, Canada
| | - Jarret J. Adams
- Department of Molecular Genetics, University of Toronto, Donnelly Centre, Toronto, Ontario, Canada
| | - Patricia Giblin
- Northern Biologics, Inc., Princess Margaret Cancer Research Tower, Toronto, Ontario, Canada
| | - Sachdev S. Sidhu
- Department of Molecular Genetics, University of Toronto, Donnelly Centre, Toronto, Ontario, Canada
| |
Collapse
|
46
|
The Many Roles of Cell Adhesion Molecules in Hepatic Fibrosis. Cells 2019; 8:cells8121503. [PMID: 31771248 PMCID: PMC6952767 DOI: 10.3390/cells8121503] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 01/09/2023] Open
Abstract
Fibrogenesis is a progressive scarring event resulting from disrupted regular wound healing due to repeated tissue injury and can end in organ failure, like in liver cirrhosis. The protagonists in this process, either liver-resident cells or patrolling leukocytes attracted to the site of tissue damage, interact with each other by soluble factors but also by direct cell–cell contact mediated by cell adhesion molecules. Since cell adhesion molecules also support binding to the extracellular matrix, they represent excellent biosensors, which allow cells to modulate their behavior based on changes in the surrounding microenvironment. In this review, we focus on selectins, cadherins, integrins and members of the immunoglobulin superfamily of adhesion molecules as well as some non-classical cell adhesion molecules in the context of hepatic fibrosis. We describe their liver-specific contributions to leukocyte recruitment, cell differentiation and survival, matrix remodeling or angiogenesis and touch on their suitability as targets in antifibrotic therapies.
Collapse
|
47
|
Requirement of β1 integrin for endothelium-dependent vasodilation and collateral formation in hindlimb ischemia. Sci Rep 2019; 9:16931. [PMID: 31729436 PMCID: PMC6858366 DOI: 10.1038/s41598-019-53137-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 10/25/2019] [Indexed: 01/07/2023] Open
Abstract
An acute increase in blood flow triggers flow-mediated dilation (FMD), which is mainly mediated by endothelial nitric oxide synthase (eNOS). A long-term increase in blood flow chronically enlarges the arterial lumen, a process called arteriogenesis. In several common human diseases, these processes are disrupted for as yet unknown reasons. Here, we asked whether β1 integrin, a mechanosensory protein in endothelial cells, is required for FMD and arteriogenesis in the ischemic hindlimb. Permanent ligation of the femoral artery in C57BL/6 J mice enlarged pre-existing collateral arteries and increased numbers of arterioles in the thigh. In the lower leg, the numbers of capillaries increased. Notably, injection of β1 integrin-blocking antibody or tamoxifen-induced endothelial cell-specific deletion of the gene for β1 integrin (Itgb1) inhibited both arteriogenesis and angiogenesis. Using high frequency ultrasound, we demonstrated that β1 integrin-blocking antibody or endothelial cell-specific depletion of β1 integrin attenuated FMD of the femoral artery, and blocking of β1 integrin function did not further decrease FMD in eNOS-deficient mice. Our data suggest that endothelial β1 integrin is required for both acute and chronic widening of the arterial lumen in response to hindlimb ischemia, potentially via functional interaction with eNOS.
Collapse
|
48
|
Rabelo LFG, Ferreira BA, Deconte SR, Tomiosso TC, Dos Santos PK, Andrade SP, Selistre de Araújo HS, Araújo FDA. Alternagin-C, a disintegrin-like protein from Bothrops alternatus venom, attenuates inflammation and angiogenesis and stimulates collagen deposition of sponge-induced fibrovascular tissue in mice. Int J Biol Macromol 2019; 140:653-660. [PMID: 31442506 DOI: 10.1016/j.ijbiomac.2019.08.171] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/18/2019] [Accepted: 08/19/2019] [Indexed: 01/12/2023]
Abstract
Alternagin-C (ALT-C), a disintegrin-like protein obtained from the venom of Bothrops alternatus, is able to modulate cellular behaviors such as adhesion, migration and proliferation, as well as the production of various growth factors via α2β1 integrin, important processes during inflammation, angiogenesis and fibrogenesis, which although appear as distinct events, act concomitantly in several chronic inflammatory diseases. Our objective was to investigate the effects of ALT-C on components of the sponge-induced inflammatory response in balb/c mice. The polyester-polyurethane sponges were implanted in mice's subcutaneous layer of the dorsal region and daily injected with saline (control group) or ALT-C (10, 100 or 1000 ng). Nine days after implantation the implants were removed and processed. ALT-C inhibited the inflammatory response, observed through mast cell reduction, NAG-activity and also by the inhibition of TNF-α, CXCL-1 and CCL2/JE/MCP-1 cytokines. ALT-C was also able to reduce hemoglobin content, number of vessels and the concentrations of VEGF and FGF cytokines. Finally, at its highest dose (1000 ng), ALT-C increased all evaluated markers associated with fibrogenesis (collagen production and TGF-β1 levels). All these factors reveal that ALT-C is a strong candidate to be exploited in the development of anti-inflammatory and anti-angiogenic therapies in chronic inflammatory processes.
Collapse
Affiliation(s)
- Luis Fernando Gonçalves Rabelo
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Bruno Antonio Ferreira
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil.
| | - Simone Ramos Deconte
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Tatiana Carla Tomiosso
- Departamento de Biologia Celular, Histologia e Embriologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | | | - Silvia Passos Andrade
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Fernanda de Assis Araújo
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| |
Collapse
|
49
|
Group A Streptococcus Induces LAPosomes via SLO/β1 Integrin/NOX2/ROS Pathway in Endothelial Cells That Are Ineffective in Bacterial Killing and Suppress Xenophagy. mBio 2019; 10:mBio.02148-19. [PMID: 31575768 PMCID: PMC6775456 DOI: 10.1128/mbio.02148-19] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Our previous reports showed that the LC3-associated GAS-containing single membrane vacuoles are inefficient for bacterial clearance in endothelial cells, which may result in bacteremia. However, the characteristics and the induction mechanisms of these LC3-positive vacuoles are still largely unknown. Here we provide the first evidence that these LC3-positive GAS-containing single membrane compartments appear to be LAPosomes, which are induced by NOX2 and ROS. Through NOX2- and ROS-mediated signaling, GAS preferentially induces LAP and inhibits bacteriostatic xenophagy in endothelial cells. We also provide the first demonstration that β1 integrin acts as the receptor for LAP induction through GAS-produced SLO stimulation in endothelial cells. Our findings reveal the underlying mechanisms of LAP induction and autophagy evasion for GAS multiplication in endothelial cells. Group A streptococcus (GAS) is an important human pathogen which can cause fatal diseases after invasion into the bloodstream. Although antibiotics and immune surveillance are the main defenses against GAS infection, GAS utilizes internalization into cells as a major immune evasion strategy. Our previous findings revealed that light chain 3 (LC3)-associated single membrane GAS-containing vacuoles in endothelial cells are compromised for bacterial clearance due to insufficient acidification after fusion with lysosomes. However, the characteristics and the activation mechanisms of these LC3-positive compartments are still largely unknown. In the present study, we demonstrated that the LC3-positive GAS is surrounded by single membrane and colocalizes with NADPH oxidase 2 (NOX2) complex but without ULK1, which are characteristics of LC3-associated phagocytosis (LAP). Inhibition of NOX2 or reactive oxygen species (ROS) significantly reduces GAS multiplication and enhances autolysosome acidification in endothelial cells through converting LAP to conventional xenophagy, which is revealed by enhancement of ULK1 recruitment, attenuation of p70s6k phosphorylation, and formation of the isolation membrane. We also clarify that the inactivation of mTORC1, which is the initiation signal of autophagy, is inhibited by NOX2- and ROS-activated phosphatidylinositol 3-kinase (PI3K)/AKT and MEK/extracellular signal-regulated kinase (ERK) pathways. In addition, streptolysin O (SLO) of GAS is identified as a crucial inducer of ROS for β1 integrin-mediated LAP induction. After downregulation of β1 integrin, GAS multiplication is reduced, accompanied with LAP inhibition and xenophagy induction. These results demonstrate that GAS infection preferentially induces ineffective LAP to evade xenophagic killing in endothelial cells through the SLO/β1 integrin/NOX2/ROS pathway.
Collapse
|
50
|
Li ZH, Zhou Y, Ding YX, Guo QL, Zhao L. Roles of integrin in tumor development and the target inhibitors. Chin J Nat Med 2019; 17:241-251. [PMID: 31076128 DOI: 10.1016/s1875-5364(19)30028-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Indexed: 01/05/2023]
Abstract
Integrin is a large family of cell adhesion molecules (CAMs) which involves in the interaction of cells/cells and cells/ extracellular matrix (ECM) to mediate cell proliferation, differentiation, adhesion, migration, etc. In recent years, aberrant expression of integrin has been clearly found in many tumor studies, indicating that integrin is closely related to tumor formation and development. Meanwhile, it has effects on tumor cell differentiation, cell migration, proliferation and tumor neovascularization. The study of drugs targeting integrins is of great significance for the clinical treatment of tumors. Because of its important role in tumorigenesis and development, integrin has become a promising target for the treatment of cancer. This review summarizes the role of integrin in tumor development and the current state of integrin inhibitors to provide a valuable reference for subsequent research.
Collapse
Affiliation(s)
- Zhao-He Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
| | - You Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
| | - You-Xiang Ding
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
| | - Qing-Long Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
| | - Li Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|