1
|
Tien NTN, Choi EJ, Thu NQ, Yu SJ, Nguyen DN, Kim DH, Long NP, Lee HS. An exploratory multi-omics study reveals distinct molecular signatures of ulcerative colitis and Crohn's disease and their correlation with disease activity. J Pharm Biomed Anal 2025; 255:116652. [PMID: 39740478 DOI: 10.1016/j.jpba.2024.116652] [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: 10/07/2024] [Revised: 12/09/2024] [Accepted: 12/22/2024] [Indexed: 01/02/2025]
Abstract
Clinically heterogeneous spectrum and molecular phenotypes of inflammatory bowel disease (IBD) remain to be comprehensively elucidated. This exploratory multi-omics study investigated the serum molecular profiles of Crohn's disease (CD) and ulcerative colitis (UC), in association with elevated fecal calprotectin and disease activity states. The serum proteome, metabolome, and lipidome of 75 treated IBD patients were profiled. Single- and multi-omic data analysis was performed to determine differential analytes and integrative biosignatures for biological interpretations. We found that chronic inflammation, phosphatidylcholines and bile acid homeostasis disturbances underlined the differences between CD and UC. Besides, elevated calprotectin was associated with higher levels of inflammatory proteins and sphingomyelins (SM) and lower levels of bile acids, amino acids, and triacylglycerols (TG). Relative to the remission disease state, the active form was characterized by decreased abundances of SMs and increased abundances of inflammatory proteins and TGs. We also observed that molecular changes upon treatment escalation were putatively related to altered levels of inflammatory response proteins, amino acids, and TGs. ISM1, ANGPTL4, chenodeoxycholate, Cer(18:1;2 O/24:1), and TG were identified as candidates subject to further investigation. Altogether, our study revealed that disturbances in immune response, bile acid homeostasis, amino acids, and lipids potentially underlie the clinically heterogeneous spectrum of IBD.
Collapse
Affiliation(s)
- Nguyen Tran Nam Tien
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Eun Jeong Choi
- Department of Internal Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, 47392, Republic of Korea
| | - Nguyen Quang Thu
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Seung Jung Yu
- Department of Internal Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, 47392, Republic of Korea
| | - Duc Ninh Nguyen
- Comparative Pediatrics, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Dong Hyun Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Nguyen Phuoc Long
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea.
| | - Hong Sub Lee
- Department of Internal Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, 47392, Republic of Korea.
| |
Collapse
|
2
|
Desai SA, Patel VP, Bhosle KP, Nagare SD, Thombare KC. The tumor microenvironment: shaping cancer progression and treatment response. J Chemother 2025; 37:15-44. [PMID: 38179655 DOI: 10.1080/1120009x.2023.2300224] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer progression and treatment response. It comprises a complex network of stromal cells, immune cells, extracellular matrix, and blood vessels, all of which interact with cancer cells and influence tumor behaviour. This review article provides an in-depth examination of the TME, focusing on stromal cells, blood vessels, signaling molecules, and ECM, along with commonly available therapeutic compounds that target these components. Moreover, we explore the TME as a novel strategy for discovering new anti-tumor drugs. The dynamic and adaptive nature of the TME offers opportunities for targeting specific cellular interactions and signaling pathways. We discuss emerging approaches, such as combination therapies that simultaneously target cancer cells and modulate the TME. Finally, we address the challenges and future prospects in targeting the TME. Overcoming drug resistance, improving drug delivery, and identifying new therapeutic targets within the TME are among the challenges discussed. We also highlight the potential of personalized medicine and the integration of emerging technologies, such as immunotherapy and nanotechnology, in TME-targeted therapies. This comprehensive review provides insights into the TME and its therapeutic implications. Understanding the TME's complexity and targeting its components offer promising avenues for the development of novel anti-tumor therapies and improved patient outcomes.
Collapse
Affiliation(s)
- Sharav A Desai
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Vipul P Patel
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Kunal P Bhosle
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Sandip D Nagare
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Kirti C Thombare
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| |
Collapse
|
3
|
Purić E, Nilsson UJ, Anderluh M. Galectin-8 inhibition and functions in immune response and tumor biology. Med Res Rev 2024; 44:2236-2265. [PMID: 38613488 DOI: 10.1002/med.22041] [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: 04/13/2023] [Revised: 03/03/2024] [Accepted: 03/29/2024] [Indexed: 04/15/2024]
Abstract
Galectins are among organisms' most abundantly expressed lectins (carbohydrate-binding proteins) that specifically bind β-galactosides. They act not only outside the cell, where they bind to extracellular matrix glycans, but also inside the cell, where they have a significant impact on signaling pathways. Galectin-8 is a galectin family protein encoded by the LGALS8 gene. Its role is evident in both T- and B-cell immunity and in the innate immune response, where it acts directly on dendritic cells and induces some pro-inflammatory cytokines. Galectin-8 also plays an important role in the defense against bacterial and viral infections. It is known to promote antibacterial autophagy by recognizing and binding glycans present on the vacuolar membrane, thus acting as a danger receptor. The most important role of galectin-8 is the regulation of cancer growth, metastasis, tumor progression, and tumor cell survival. Importantly, the expression of galectins is typically higher in tumor tissues than in noncancerous tissues. In this review article, we focus on galectin-8 and its function in immune response, microbial infections, and cancer. Given all of these functions of galectin-8, we emphasize the importance of developing new and selective galectin-8 inhibitors and report the current status of their development.
Collapse
Affiliation(s)
- Edvin Purić
- Department of Pharmaceutical Chemistry, University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| | - Ulf J Nilsson
- Department of Chemistry, Lund University, Lund, Sweden
| | - Marko Anderluh
- Department of Pharmaceutical Chemistry, University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| |
Collapse
|
4
|
Gupta AO, Furcich JW, Nascene DR, Kemp S, King CJ, Nolan EE, Durose W, Miller BS, Orchard PJ, Lund TC. Targeting VEGF-mediated blood-brain barrier disruption in advanced cerebral leukodystrophy. J Neuroimmunol 2024; 393:578395. [PMID: 38897089 DOI: 10.1016/j.jneuroim.2024.578395] [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: 12/19/2023] [Revised: 03/13/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
The earliest clinical manifestation of cerebral adrenoleukodystrophy (CALD) is adrenal insufficiency (AI) characterized by elevations in ACTH and loss of cortisol. We showed high (though physiologically achievable) levels of ACTH increases endothelial permeability, increases anisotropy, and increases VEGF secretion. An ACBD1 knockout endothelial cell line had increased sensitivity to ACTH and VEGF. Inhibition of VEGF via application of anti-VEGF (bevacizumab) improved permeability. Six boys with advanced CALD were treated with bevacizumab combined with dexamethasone and ruxolitinib as immune suppressants. Most boys had decreases in gadolinium enhancement on MRI indicating improvement in endothelial function, though all boys continued to progress symptomatically.
Collapse
Affiliation(s)
- Ashish O Gupta
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Justin W Furcich
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - David R Nascene
- Department of Diagnostic Radiology, University of Minnesota Medical Center, Minneapolis, MN 55455, United States of America
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | - Carina J King
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Erin E Nolan
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Willa Durose
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Bradley S Miller
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Paul J Orchard
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Troy C Lund
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America.
| |
Collapse
|
5
|
van Noorden CJ, Yetkin-Arik B, Serrano Martinez P, Bakker N, van Breest Smallenburg ME, Schlingemann RO, Klaassen I, Majc B, Habic A, Bogataj U, Galun SK, Vittori M, Erdani Kreft M, Novak M, Breznik B, Hira VV. New Insights in ATP Synthesis as Therapeutic Target in Cancer and Angiogenic Ocular Diseases. J Histochem Cytochem 2024; 72:329-352. [PMID: 38733294 PMCID: PMC11107438 DOI: 10.1369/00221554241249515] [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: 12/05/2023] [Accepted: 04/01/2024] [Indexed: 05/13/2024] Open
Abstract
Lactate and ATP formation by aerobic glycolysis, the Warburg effect, is considered a hallmark of cancer. During angiogenesis in non-cancerous tissue, proliferating stalk endothelial cells (ECs) also produce lactate and ATP by aerobic glycolysis. In fact, all proliferating cells, both non-cancer and cancer cells, need lactate for the biosynthesis of building blocks for cell growth and tissue expansion. Moreover, both non-proliferating cancer stem cells in tumors and leader tip ECs during angiogenesis rely on glycolysis for pyruvate production, which is used for ATP synthesis in mitochondria through oxidative phosphorylation (OXPHOS). Therefore, aerobic glycolysis is not a specific hallmark of cancer but rather a hallmark of proliferating cells and limits its utility in cancer therapy. However, local treatment of angiogenic eye conditions with inhibitors of glycolysis may be a safe therapeutic option that warrants experimental investigation. Most types of cells in the eye such as photoreceptors and pericytes use OXPHOS for ATP production, whereas proliferating angiogenic stalk ECs rely on glycolysis for lactate and ATP production. (J Histochem Cytochem XX.XXX-XXX, XXXX).
Collapse
Affiliation(s)
- Cornelis J.F. van Noorden
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
- Ocular Angiogenesis Group, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
| | - Bahar Yetkin-Arik
- Department of Pediatric Pulmonology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
- Centre for Living Technologies, Alliance TU/e, WUR, UU, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Paola Serrano Martinez
- Ocular Angiogenesis Group, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
| | - Noëlle Bakker
- Ocular Angiogenesis Group, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
| | | | - Reinier O. Schlingemann
- Ocular Angiogenesis Group, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
- Department of Ophthalmology, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
- Department of Ophthalmology, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
| | - Bernarda Majc
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Anamarija Habic
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
- Jozef Stefan Postgraduate School, Ljubljana, Slovenia
| | - Urban Bogataj
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - S. Katrin Galun
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Milos Vittori
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Mateja Erdani Kreft
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Metka Novak
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Barbara Breznik
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Vashendriya V.V. Hira
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| |
Collapse
|
6
|
Cardama GA, Bucci PL, Lemos JS, Llavona C, Benavente MA, Hellmén E, Fara ML, Medrano E, Spitzer E, Demarco IA, Sabella P, Garona J, Alonso DF. In Silico and In Vitro Evaluation of Bevacizumab Biosimilar MB02 as an Antitumor Agent in Canine Mammary Carcinoma. Animals (Basel) 2023; 13:2507. [PMID: 37570315 PMCID: PMC10417262 DOI: 10.3390/ani13152507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Canine mammary carcinomas (CMC) are associated with major aggressive clinical behavior and high mortality. The current standard of care is based on surgical resection, without an established effective treatment scheme, highlighting the urgent need to develop novel effective therapies. Vascular endothelial growth factor (VEGF) is a key regulator of tumor angiogenesis and progression in the majority of solid cancers, including human and canine mammary carcinomas. The first therapy developed to target VEGF was bevacizumab, a recombinant humanized monoclonal antibody, which has already been approved as an anticancer agent in several human cancers. The goal of this work was to establish the therapeutic value of MB02 bevacizumab biosimilar in CMC. First, through different in silico approaches using the MUSCLE multiple-sequence alignment tool and the FoldX protein design algorithm, we were able to predict that canine VEGF is recognized by bevacizumab, after showing an extremely high sequence similarity between canine and human VEGF. Further, by using an ELISA-based in vitro binding assay, we confirmed that MB02 biosimilar was able to recognize canine VEGF. Additionally, canine VEGF-induced microvascular endothelial cell proliferation was inhibited in a concentration-dependent manner by MB02 biosimilar. These encouraging results show a high potential for MB02 as a promising therapeutic agent for the management of CMC.
Collapse
Affiliation(s)
- Georgina A. Cardama
- Centro de Oncología Molecular y Traslacional (COMTra), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal B1876, Argentina; (P.L.B.); (J.S.L.); (C.L.); (J.G.)
- Plataforma de Servicios Biotecnológicos, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal B1876, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1040, Argentina;
| | - Paula L. Bucci
- Centro de Oncología Molecular y Traslacional (COMTra), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal B1876, Argentina; (P.L.B.); (J.S.L.); (C.L.); (J.G.)
- Plataforma de Servicios Biotecnológicos, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal B1876, Argentina
| | - Jesús S. Lemos
- Centro de Oncología Molecular y Traslacional (COMTra), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal B1876, Argentina; (P.L.B.); (J.S.L.); (C.L.); (J.G.)
- Plataforma de Servicios Biotecnológicos, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal B1876, Argentina
| | - Candela Llavona
- Centro de Oncología Molecular y Traslacional (COMTra), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal B1876, Argentina; (P.L.B.); (J.S.L.); (C.L.); (J.G.)
- Centro de Medicina Traslacional (CEMET), Hospital de Alta Complejidad en Red S.A.M.I.C. El Cruce “Nestor Kirchner”, Florencio Varela B5401, Argentina
| | - Micaela A. Benavente
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1040, Argentina;
- Laboratorio de Endocrinología, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, Tandil B7000, Argentina
- Centro de Investigación Veterinaria de Tandil (CIVETAN), CONICET—CICPBA—Universidad Nacional del Centro de la Provincia de Buenos Aires, Tandil B7000, Argentina
| | - Eva Hellmén
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences (SLU), 750 07 Uppsala, Sweden;
| | - María Laura Fara
- Laboratorio Elea Phoenix S.A, Los Polvorines B1613, Argentina; (M.L.F.); (E.M.); (E.S.)
| | - Eduardo Medrano
- Laboratorio Elea Phoenix S.A, Los Polvorines B1613, Argentina; (M.L.F.); (E.M.); (E.S.)
| | - Eduardo Spitzer
- Laboratorio Elea Phoenix S.A, Los Polvorines B1613, Argentina; (M.L.F.); (E.M.); (E.S.)
| | | | | | - Juan Garona
- Centro de Oncología Molecular y Traslacional (COMTra), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal B1876, Argentina; (P.L.B.); (J.S.L.); (C.L.); (J.G.)
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1040, Argentina;
- Centro de Medicina Traslacional (CEMET), Hospital de Alta Complejidad en Red S.A.M.I.C. El Cruce “Nestor Kirchner”, Florencio Varela B5401, Argentina
| | - Daniel F. Alonso
- Centro de Oncología Molecular y Traslacional (COMTra), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal B1876, Argentina; (P.L.B.); (J.S.L.); (C.L.); (J.G.)
- Plataforma de Servicios Biotecnológicos, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal B1876, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1040, Argentina;
| |
Collapse
|
7
|
Liu ZL, Chen HH, Zheng LL, Sun LP, Shi L. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct Target Ther 2023; 8:198. [PMID: 37169756 PMCID: PMC10175505 DOI: 10.1038/s41392-023-01460-1] [Citation(s) in RCA: 243] [Impact Index Per Article: 121.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/20/2023] [Accepted: 04/20/2023] [Indexed: 05/13/2023] Open
Abstract
Angiogenesis, the formation of new blood vessels, is a complex and dynamic process regulated by various pro- and anti-angiogenic molecules, which plays a crucial role in tumor growth, invasion, and metastasis. With the advances in molecular and cellular biology, various biomolecules such as growth factors, chemokines, and adhesion factors involved in tumor angiogenesis has gradually been elucidated. Targeted therapeutic research based on these molecules has driven anti-angiogenic treatment to become a promising strategy in anti-tumor therapy. The most widely used anti-angiogenic agents include monoclonal antibodies and tyrosine kinase inhibitors (TKIs) targeting vascular endothelial growth factor (VEGF) pathway. However, the clinical benefit of this modality has still been limited due to several defects such as adverse events, acquired drug resistance, tumor recurrence, and lack of validated biomarkers, which impel further research on mechanisms of tumor angiogenesis, the development of multiple drugs and the combination therapy to figure out how to improve the therapeutic efficacy. Here, we broadly summarize various signaling pathways in tumor angiogenesis and discuss the development and current challenges of anti-angiogenic therapy. We also propose several new promising approaches to improve anti-angiogenic efficacy and provide a perspective for the development and research of anti-angiogenic therapy.
Collapse
Affiliation(s)
- Zhen-Ling Liu
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Huan-Huan Chen
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Li-Li Zheng
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Li-Ping Sun
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China.
| | - Lei Shi
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China.
| |
Collapse
|
8
|
Hu J, Lin F, Yin Y, Shang Y, Xiao Z, Xu W. Adipocyte-derived exosomal miR-30c-5p promotes ovarian angiogenesis in polycystic ovary syndrome via the SOCS3/STAT3/VEGFA pathway. J Steroid Biochem Mol Biol 2023; 230:106278. [PMID: 36870372 DOI: 10.1016/j.jsbmb.2023.106278] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/07/2023] [Accepted: 03/02/2023] [Indexed: 03/06/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a systemic endocrine disease affecting women's reproductive health. Ovarian angiogenesis in PCOS patients is abnormal, manifested by increased ovarian stromal vascularization and upregulated proangiogenic factors such as vascular endothelial growth factor (VEGF). However, the specific mechanisms underlying these changes in PCOS remain unknown. In this study, we induced the adipogenic differentiation in preadipocyte 3T3-L1 cells and found that adipocyte-derived exosomes promoted proliferation, migration, tube formation, and VEGFA expression in human ovarian microvascular endothelial cells (HOMECs) by delivering miR-30c-5p. Mechanistically, dual luciferase reporter assay demonstrated that miR-30c-5p directly targeted the 3'- untranslated region (UTR) of suppressor of cytokine signaling 3 (SOCS3) mRNA. In addition, adipocyte-derived exosomal miR-30c-5p activated signal transducer and activator of transcription 3 (STAT3)/VEGFA pathway in HOMECs via targeting SOCS3. In vivo experiments indicated that tail vein injection of adipocyte-derived exosomes exacerbated endocrine and metabolic disorders and ovarian angiogenesis in mice with PCOS via miR-30c-5p. Taken together, the study revealed that adipocyte-derived exosomal miR-30c-5p promotes ovarian angiogenesis via the SOCS3/STAT3/VEGFA pathway, thereby participating in the development of PCOS.
Collapse
Affiliation(s)
- Jian Hu
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan 430060, China
| | - Fangyou Lin
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yuchen Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan 430060, China
| | - Yunjie Shang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan 430060, China
| | - Zhuoni Xiao
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan 430060, China.
| | - Wangming Xu
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan 430060, China.
| |
Collapse
|
9
|
Hübers C, Abdul Pari AA, Grieshober D, Petkov M, Schmidt A, Messmer T, Heyer CM, Schölch S, Kapel SS, Gengenbacher N, Singhal M, Schieb B, Fricke C, Will R, Remans K, Utikal JS, Reissfelder C, Schlesner M, Hodivala-Dilke KM, Kersten S, Goerdt S, Augustin HG, Felcht M. Primary tumor-derived systemic nANGPTL4 inhibits metastasis. J Exp Med 2023; 220:e20202595. [PMID: 36269299 PMCID: PMC9595206 DOI: 10.1084/jem.20202595] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 08/07/2022] [Accepted: 09/15/2022] [Indexed: 11/04/2022] Open
Abstract
Primary tumors and distant site metastases form a bidirectionally communicating system. Yet, the molecular mechanisms of this crosstalk are poorly understood. Here, we identified the proteolytically cleaved fragments of angiopoietin-like 4 (ANGPTL4) as contextually active protumorigenic and antitumorigenic contributors in this communication ecosystem. Preclinical studies in multiple tumor models revealed that the C-terminal fragment (cANGPTL4) promoted tumor growth and metastasis. In contrast, the N-terminal fragment of ANGPTL4 (nANGPTL4) inhibited metastasis and enhanced overall survival in a postsurgical metastasis model by inhibiting WNT signaling and reducing vascularity at the metastatic site. Tracing ANGPTL4 and its fragments in tumor patients detected full-length ANGPTL4 primarily in tumor tissues, whereas nANGPTL4 predominated in systemic circulation and correlated inversely with disease progression. The study highlights the spatial context of the proteolytic cleavage-dependent pro- and antitumorigenic functions of ANGPTL4 and identifies and validates nANGPTL4 as a novel biomarker of tumor progression and antimetastatic therapeutic agent.
Collapse
Affiliation(s)
- Corinne Hübers
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergy, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University and Centre of Excellence of Dermatology of Baden-Württemberg, Mannheim, Germany
| | - Ashik Ahmed Abdul Pari
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Denise Grieshober
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Martin Petkov
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | | | - Tatjana Messmer
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergy, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University and Centre of Excellence of Dermatology of Baden-Württemberg, Mannheim, Germany
| | - Christian Moritz Heyer
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Biomedical Informatics, Data Mining and Data Analytics, Augsburg University, Augsburg, Germany
| | - Sebastian Schölch
- JCCU Translational Surgical Oncology (A430), German Cancer Research Center, Heidelberg, Germany
- Department of Surgery, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at University Medical Centre Mannheim, Mannheim, Germany
| | - Stephanie S. Kapel
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Nicolas Gengenbacher
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Mahak Singhal
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
- Laboratory of AngioRhythms, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Benjamin Schieb
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Claudine Fricke
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Rainer Will
- Genomics & Proteomics Core Facilities, German Cancer Research Center, Heidelberg, Germany
| | - Kim Remans
- Protein Expression and Purification Core Facility, European Molecular Biology Center, Heidelberg, Germany
| | - Jochen Sven Utikal
- Department of Dermatology, Venereology and Allergy, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University and Centre of Excellence of Dermatology of Baden-Württemberg, Mannheim, Germany
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
| | - Christoph Reissfelder
- Department of Surgery, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at University Medical Centre Mannheim, Mannheim, Germany
| | - Matthias Schlesner
- Biomedical Informatics, Data Mining and Data Analytics, Augsburg University, Augsburg, Germany
| | - Kairbaan M. Hodivala-Dilke
- Center for Tumor Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Center, London, United Kingdom
| | - Sander Kersten
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Sergij Goerdt
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Dermatology, Venereology and Allergy, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University and Centre of Excellence of Dermatology of Baden-Württemberg, Mannheim, Germany
| | - Hellmut G. Augustin
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Moritz Felcht
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergy, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University and Centre of Excellence of Dermatology of Baden-Württemberg, Mannheim, Germany
| |
Collapse
|
10
|
Mohammadi P, Yarani R, Rahimpour A, Ranjbarnejad F, Mendes Lopes de Melo J, Mansouri K. Targeting endothelial cell metabolism in cancerous microenvironment: a new approach for anti-angiogenic therapy. Drug Metab Rev 2022; 54:386-400. [PMID: 36031813 DOI: 10.1080/03602532.2022.2116033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Anti-angiogenic therapy is a practical approach to managing diseases with increased angiogenesis, such as cancer, maculopathies, and retinopathies. Considering the fundamental gaps in the knowledge of the vital pathways involved in angiogenesis and its inhibition and the insufficient efficiency of existing angiogenesis inhibitors, there is an increasing focus on the emergence of new therapeutic strategies aimed at inhibiting pathological angiogenesis. Angiogenesis is forming a new vascular network from existing vessels; endothelial cells (ECs), vascular lining cells, are the main actors of angiogenesis in physiological or pathological conditions. Switching from a quiescent state to a highly migratory and proliferative state during new vessel formation called "angiogenic switch" is driven by a "metabolic switch" in ECs, angiogenic growth factors, and other signals. As the characteristics of ECs change by altering the surrounding environment, they appear to have a different metabolism in a tumor microenvironment (TME). Therefore, pathological angiogenesis can be inhibited by targeting metabolic pathways. In the current review, we aim to discuss the EC metabolic pathways under normal and TME conditions to verify the suitability of targeting them with novel therapies.
Collapse
Affiliation(s)
- Parisa Mohammadi
- Medical Biology Research Center, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Department of Clinical, Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Azam Rahimpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical, Sciences, Tehran, Iran
| | - Fatemeh Ranjbarnejad
- Medical Biology Research Center, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Joana Mendes Lopes de Melo
- Translational Type 1 Diabetes Research, Department of Clinical, Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Kamran Mansouri
- Medical Biology Research Center, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
11
|
Targeting JWA for Cancer Therapy: Functions, Mechanisms and Drug Discovery. Cancers (Basel) 2022; 14:cancers14194655. [PMID: 36230577 PMCID: PMC9564207 DOI: 10.3390/cancers14194655] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary JWA has been identified as a potential therapeutic target for several cancers. In this review, we summarize the tumor suppressive functions of the JWA gene and its role in anti-cancer drug development. The focus is on elucidating the key regulatory proteins up and downstream of JWA and their signaling networks. We also discuss current strategies for targeting JWA (JWA peptides, small molecule agonists, and JWA-targeted Pt (IV) prodrugs). Abstract Tumor heterogeneity limits the precision treatment of targeted drugs. It is important to find new tumor targets. JWA, also known as ADP ribosylation factor-like GTPase 6 interacting protein 5 (ARL6IP5, GenBank: AF070523, 1998), is a microtubule-associated protein and an environmental response gene. Substantial evidence shows that JWA is low expressed in a variety of malignancies and is correlated with overall survival. As a tumor suppressor, JWA inhibits tumor progression by suppressing multiple oncogenes or activating tumor suppressor genes. Low levels of JWA expression in tumors have been reported to be associated with multiple aspects of cancer progression, including angiogenesis, proliferation, apoptosis, metastasis, and chemotherapy resistance. In this review, we will discuss the structure and biological functions of JWA in tumors, examine the potential therapeutic strategies for targeting JWA and explore the directions for future investigation.
Collapse
|
12
|
Zhu L, Guo Z, Zhang J, Yang Y, Liu C, Zhang L, Gu Z, Li Y, Ding Z, Shi G. Recombinant Human Arresten and Canstatin Inhibit Angiogenic Behaviors of HUVECs via Inhibiting the PI3K/Akt Signaling Pathway. Int J Mol Sci 2022; 23:ijms23168995. [PMID: 36012259 PMCID: PMC9409110 DOI: 10.3390/ijms23168995] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/05/2022] [Accepted: 08/07/2022] [Indexed: 11/30/2022] Open
Abstract
Angiogenetic inhibitors are crucial in tumor therapy, and endogenous angiogenesis inhibitors have attracted considerable attention due to their effectiveness, safety, and multi-targeting ability. Arresten and canstatin, which have anti-angiogenesis effects, are the c-terminal fragments of the α1 and α2 chains of type IV collagen, respectively. In this study, human arresten and canstatin were recombinantly expressed in Escherichia coli (E. coli), and their effects on the proliferation, migration and tube formation of human umbilical vein endothelial cells (HUVECs) were evaluated. Regarding the cell cycle distribution test and 5-ethynyl-2′-deoxyuridine (EdU) assays, arresten and canstatin could repress the proliferation of HUVECs at a range of concentrations. Transwell assay indicated that the migration of HUVECs was significantly decreased in the presence of arresten and canstatin, while tube formation assays suggested that the total tube length and junction number of HUVECs were significantly inhibited by these two proteins; moreover, they could also reduce the expression of vascular endothelial growth factor (VEGF) and the phosphorylation levels of PI3K and Akt, which indicated that the activation of the 3-kinase/serine/threonine-kinase (PI3K/Akt) signaling pathway was inhibited. These findings may have important implications for the soluble recombinant expression of human arresten and canstatin, and for the related therapy of cancer.
Collapse
Affiliation(s)
- Lingyu Zhu
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
| | - Zitao Guo
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
| | - Ji Zhang
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
| | - Yuliang Yang
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
| | - Chunyu Liu
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
| | - Liang Zhang
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
- Correspondence: ; Tel.: +86-510-85918235
| | - Zhenghua Gu
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
| | - Youran Li
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
| | - Zhongyang Ding
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
| | - Guiyang Shi
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
13
|
Baquet-Walscheid K, Wildschütz L, Kasper M, Busch M, Thanos S, Bauer D, Stoll M, König S, Heiligenhaus A. Assessment of angiogenesis-related parameters in juvenile idiopathic arthritis-associated uveitis. Mol Biol Rep 2022; 49:6093-6102. [PMID: 35359237 DOI: 10.1007/s11033-022-07398-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/17/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE Juvenile idiopathic arthritis-associated uveitis (JIAU) may run a chronic and treatment-resistant course, and occasionally, alterations of the iris vasculature may be observed clinically. METHODS Iris tissue (IT), aqueous humor (AH) and serum samples from patients with clinically inactive JIAU (n = 30), acute anterior uveitis (AAU; n = 18), and primary open angle glaucoma (POAG; n = 20) were obtained during trabeculectomy or cataract surgery. Samples were analyzed by RNA-Seq, qRT-PCR, LC-IMS, Western-Blot, and LEGENDplex™ analysis. Pattern of iris vasculature in JIAU patients was assessed qualitatively via fluorescein and indocyanine green angiography (FLA/ICGA). RESULTS RNA-Seq of IT showed significantly differential expression (DE) of 136 genes between JIAU and POAG, of which 15 were associated with angiogenesis. qRT-PCR, performed to validate RNA-Seq results, showed upregulation of the angiogenesis-related genes Kdr, Angpt-1, Tie-1, Tie-2 and Mmrn2 in IT (JIAU vs POAG, p > 0.05). LC-IMS of IT revealed a total number of 56 DE proteins (JIAU vs POAG), of which Angiopoetin, Lumican and Decorin were associated with angiogenesis and showed increased (p > 0.05) expression on Western-Blot analysis. LEGENDplex™ analysis showed upregulation of ANGPT-2 in AH from JIAU compared to AAU and POAG, whereas VEGF was upregulated in AAU. Iris vascular leakage, hypoperfusion and neovascularization were observed by FLA/ICGA in JIA patients with treatment-refractory complicated course of uveitis. CONCLUSION Angiogenesis-related factors could play a role in long-standing complicated JIAU, leading to clinically visible alterations in selected cases.
Collapse
Affiliation(s)
- Karoline Baquet-Walscheid
- Department of Ophthalmology and Ophtha-Lab, St. Franziskus-Hospital, Hohenzollernring 74, 48145, Münster, Germany. .,University of Duisburg-Essen, Essen, Germany.
| | - Lena Wildschütz
- Department of Ophthalmology and Ophtha-Lab, St. Franziskus-Hospital, Hohenzollernring 74, 48145, Münster, Germany
| | - Maren Kasper
- Department of Ophthalmology and Ophtha-Lab, St. Franziskus-Hospital, Hohenzollernring 74, 48145, Münster, Germany
| | - Martin Busch
- Department of Ophthalmology and Ophtha-Lab, St. Franziskus-Hospital, Hohenzollernring 74, 48145, Münster, Germany
| | - Solon Thanos
- Institute of Experimental Ophthalmology, University of Muenster, Münster, Germany
| | - Dirk Bauer
- Department of Ophthalmology and Ophtha-Lab, St. Franziskus-Hospital, Hohenzollernring 74, 48145, Münster, Germany
| | - Monika Stoll
- Core Facility Genomics, University of Muenster, Münster, Germany.,Department "Genetic Epidemiology", Institute of Human Genetics, University of Münster, Münster, Germany
| | - Simone König
- Core Unit Proteomics, Interdisciplinary Center for Clinical Research, University of Muenster, Münster, Germany
| | - Arnd Heiligenhaus
- Department of Ophthalmology and Ophtha-Lab, St. Franziskus-Hospital, Hohenzollernring 74, 48145, Münster, Germany.,University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
14
|
Kirman DC, Renganathan B, Chui WK, Chen MW, Kaya NA, Ge R. Cell surface nucleolin is a novel ADAMTS5 receptor mediating endothelial cell apoptosis. Cell Death Dis 2022; 13:172. [PMID: 35197459 PMCID: PMC8866485 DOI: 10.1038/s41419-022-04618-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 12/17/2021] [Accepted: 01/19/2022] [Indexed: 12/12/2022]
Abstract
A Disintegrin and Metalloproteinase with ThromboSpondin motif (ADAMTS) 5 functions as an anti-angiogenic and anti-cancer protein independent of its metalloproteinase activity. Both full-length ADAMTS5 and TS5-p45, the autocatalytically cleaved C-terminal 45 kDa truncate of ADAMTS5, inhibits angiogenesis, and induces endothelial cell (EC) apoptosis. However, how ADAMTS5 triggers EC apoptosis remains unclear. This work shows that caspase-8 (Cas-8) and caspase-9 (Cas-9) are involved in TS5-p45-induced EC apoptosis. We identify cell surface nucleolin (NCL) as a novel high-affinity receptor for TS5-p45 in ECs, mediating TS5-p45's cell surface binding and pro-apoptotic function. We show that the central RNA-binding domain (RBD) of NCL is essential and sufficient for its binding to TS5-p45. Upon interacting with EC surface NCL, TS5-p45 is internalized through clathrin- and caveolin-dependent endocytosis and trafficked to the nucleus via late endosomes (LEs). We demonstrate that the nuclear trafficking of TS5-p45 is important for its pro-apoptotic activity as disruption of LE membrane integrity with an endosomolytic peptide suppressed both nuclear trafficking and pro-apoptotic activity of TS5-p45. Through cell surface biotinylation, we revealed that cell surface NCL shuttles extracellular TS5-p45 to the nucleus to mediate apoptosis. Furthermore, blocking the importin α1/ß1 receptor hindered the nuclear trafficking of TS5-p45, suggesting the involvement of the nuclear importing machinery for this nuclear translocation. RNA-seq identified many apoptosis-related genes that are differentially expressed at least two-fold in TS5-p45-treated ECs, with 10 of them qRT-PCR-validated and at least 5 of these genes potentially contributing to TS5-p45-NCL-induced apoptosis. Altogether, our work identifies NCL as a novel cell surface receptor for ADAMTS5 and demonstrates the critical role of NCL-mediated internalization and nuclear trafficking for ADAMTS5-induced EC apoptosis. These findings reveal novel mechanistic insights of the secreted metalloproteinase ADAMTS5 in angiogenesis inhibition.
Collapse
Affiliation(s)
- Dogan Can Kirman
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Bhuvanasundar Renganathan
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Wai Kit Chui
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Ming Wei Chen
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Neslihan Arife Kaya
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, 138672, Singapore
| | - Ruowen Ge
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore.
| |
Collapse
|
15
|
Robles JP, Zamora M, Siqueiros-Marquez L, Adan-Castro E, Ramirez-Hernandez G, Nuñez FF, Lopez-Casillas F, Millar RP, Bertsch T, Martínez de la Escalera G, Triebel J, Clapp C. The HGR motif is the antiangiogenic determinant of vasoinhibin: implications for a therapeutic orally active oligopeptide. Angiogenesis 2022; 25:57-70. [PMID: 34097181 PMCID: PMC8813873 DOI: 10.1007/s10456-021-09800-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023]
Abstract
The hormone prolactin acquires antiangiogenic and antivasopermeability properties after undergoing proteolytic cleavage to vasoinhibin, an endogenous prolactin fragment of 123 or more amino acids that inhibits the action of multiple proangiogenic factors. Preclinical and clinical evidence supports the therapeutic potential of vasoinhibin against angiogenesis-related diseases including diabetic retinopathy, peripartum cardiomyopathy, rheumatoid arthritis, and cancer. However, the use of vasoinhibin in the clinic has been limited by difficulties in its production. Here, we removed this barrier to using vasoinhibin as a therapeutic agent by showing that a short linear motif of just three residues (His46-Gly47-Arg48) (HGR) is the functional determinant of vasoinhibin. The HGR motif is conserved throughout evolution, its mutation led to vasoinhibin loss of function, and oligopeptides containing this sequence inhibited angiogenesis and vasopermeability with the same potency as whole vasoinhibin. Furthermore, the oral administration of an optimized cyclic retro-inverse vasoinhibin heptapeptide containing HGR inhibited melanoma tumor growth and vascularization in mice and exhibited equal or higher antiangiogenic potency than other antiangiogenic molecules currently used as anti-cancer drugs in the clinic. Finally, by unveiling the mechanism that obscures the HGR motif in prolactin, we anticipate the development of vasoinhibin-specific antibodies to solve the on-going challenge of measuring endogenous vasoinhibin levels for diagnostic and interventional purposes, the design of vasoinhibin antagonists for managing insufficient angiogenesis, and the identification of putative therapeutic proteins containing HGR.
Collapse
Affiliation(s)
- Juan Pablo Robles
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Magdalena Zamora
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | | | - Elva Adan-Castro
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | | | - Francisco Freinet Nuñez
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México
| | - Fernando Lopez-Casillas
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), México City, México
| | - Robert P Millar
- Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Thomas Bertsch
- Institute for Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Nuremberg General Hospital & Paracelsus Medical University, Nuremberg, Germany
| | | | - Jakob Triebel
- Institute for Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Nuremberg General Hospital & Paracelsus Medical University, Nuremberg, Germany
| | - Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, México.
| |
Collapse
|
16
|
Debir B, Meaney C, Kohandel M, Unlu MB. The role of calcium oscillations in the phenotype selection in endothelial cells. Sci Rep 2021; 11:23781. [PMID: 34893636 PMCID: PMC8664853 DOI: 10.1038/s41598-021-02720-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/07/2021] [Indexed: 11/10/2022] Open
Abstract
Angiogenesis is an important process in the formation and maintenance of tissues which is driven by a complex system of intracellular and intercellular signaling mechanisms. Endothelial cells taking part in early angiogenesis must select their phenotype as either a tip cells (leading, migratory) or a stalk cells (following). Recent experiments have demonstrated that rapid calcium oscillations within active cells characterize this phenotype selection process and that these oscillations play a necessary role in governing phenotype selection and eventual vessel architecture. In this work, we develop a mathematical model capable of describing these oscillations and their role in phenotype selection then use it to improve our understanding of the biological mechanisms at play. We developed a model based on two previously published and experimentally validated mathematical models of calcium and angiogenesis then use our resulting model to simulate various multi-cell scenarios. We are able to capture essential calcium oscillation dynamics and intercellular communication between neighboring cells. The results of our model show that although the late DLL4 (a transmembrane protein that activates Notch pathway) levels of a cell are connected with its initial IP3 (Inositol 1,4,5-trisphosphate) level, cell-to-cell communication determines its eventual phenotype.
Collapse
Affiliation(s)
- Birses Debir
- Department of Physics, Bogazici University, 34342, Istanbul, Turkey.
| | - Cameron Meaney
- Department of Applied Mathematics, University of Waterloo, Waterloo, ON, Canada
| | - Mohammad Kohandel
- Department of Applied Mathematics, University of Waterloo, Waterloo, ON, Canada
| | - M Burcin Unlu
- Department of Physics, Bogazici University, 34342, Istanbul, Turkey
- Hokkaido University, Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education (GI-CoRE), Sapporo, 060-8648, Japan
| |
Collapse
|
17
|
Krajnak S, Battista MJ, Hasenburg A, Schmidt M. Metronomic Chemotherapy for Metastatic Breast Cancer. Oncol Res Treat 2021; 45:12-17. [PMID: 34794154 DOI: 10.1159/000520236] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/12/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND As disease control and quality of life play a leading role in metastatic breast cancer (MBC), metronomic chemotherapy (MCT) is gaining popularity alongside conventional chemotherapy (CCT) and targeted therapies. SUMMARY MCT, defined as continuous administration of low-dose chemotherapeutic agents, is accepted as a therapy that exerts its effects via immunomodulation, anti-angiogenesis and direct cytotoxic effects. Oral administration of MCT is safe, easy to handle, and allows for flexible drug dosing. Dose accumulations associated with non-tolerable side effects are rare, so the medication can be administered for longer periods of time. Patients with hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative metastatic disease resistant to endocrine-based therapy and not requiring rapid tumor response are generally suitable for MCT. However, MCT may also be promising in patients with triple-negative and HER2-positive tumors without aggressive disease who prefer a lower toxicity profile compared to CCT. The most commonly used agents are cyclophosphamide (CTX), methotrexate (MTX), capecitabine (CAPE), and vinorelbine (VRL), whereby a combination of agents is frequently applied. Key Messages: Based on the growing body of evidence, MCT can be considered as a suitable treatment option in selected MBC patients. Nevertheless, there is an urgent need for randomized controlled trials comparing MCT with CCT, but also with best supportive care. Due to the multimodal mechanisms of action, the combination with targeted and immunological therapies may represent a new promising approach for the treatment of MBC.
Collapse
Affiliation(s)
- Slavomir Krajnak
- Department of Gynecology and Obstetrics, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Marco J Battista
- Department of Gynecology and Obstetrics, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Annette Hasenburg
- Department of Gynecology and Obstetrics, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Marcus Schmidt
- Department of Gynecology and Obstetrics, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
18
|
Zheng W, Zhang S, Guo H, Chen X, Huang Z, Jiang S, Li M. Multi-omics analysis of tumor angiogenesis characteristics and potential epigenetic regulation mechanisms in renal clear cell carcinoma. Cell Commun Signal 2021; 19:39. [PMID: 33761933 PMCID: PMC7992844 DOI: 10.1186/s12964-021-00728-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 02/20/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Tumor angiogenesis, an essential process for cancer proliferation and metastasis, has a critical role in prognostic of kidney renal clear cell carcinoma (KIRC), as well as a target in guiding treatment with antiangiogenic agents. However, tumor angiogenesis subtypes and potential epigenetic regulation mechanisms in KIRC patient remains poorly characterized. System evaluation of angiogenesis subtypes in KIRC patient might help to reveal the mechanisms of KIRC and develop more target treatments for patients. METHOD Ten independent tumor angiogenesis signatures were obtained from molecular signatures database (MSigDB) and gene set variation analysis was performed to calculate the angiogenesis score in silico using the Cancer Genome Atlas (TCGA) KIRC dataset. Tumor angiogenesis subtypes in 539 TCGA-KIRC patients were identified using consensus clustering analysis. The potential regulation mechanisms was studied using gene mutation, copy number variation, and differential methylation analysis (DMA). The master transcription factors (MTF) that cause the difference in tumor angiogenesis signals were completed by transcription factor enrichment analysis. RESULTS The angiogenesis score of a prognosis related angiogenesis signature including 189 genes was significantly correlated with immune score, stroma score, hypoxia score, and vascular endothelial growth factor (VEGF) signal score in 539 TCGA KIRC patients. MMRN2, CLEC14A, ACVRL1, EFNB2, and TEK in candidate gene set showed highest correlation coefficient with angiogenesis score in TCGA-KIRC patients. In addition, all of them were associated with overall survival in both TCGA-KIRC and E-MTAB-1980 KIRC data. Clustering analysis based on 183 genes in angiogenesis signature identified two prognosis related angiogenesis subtypes in TCGA KIRC patients. Two clusters also showed different angiogenesis score, immune score, stroma score, hypoxia score, VEGF signal score, and microenvironment score. DMA identified 59,654 differential methylation sites between two clusters and part of these sites were correlated with tumor angiogenesis genes including CDH13, COL4A3, and RHOB. In addition, RFX2, SOX13, and THRA were identified as top three MTF in regulating angiogenesis signature in KIRC patients. CONCLUSION Our study indicate that evaluation the angiogenesis subtypes of KIRC based on angiogenesis signature with 183 genes and potential epigenetic mechanisms may help to develop more target treatments for KIRC patients. Video Abstract.
Collapse
Affiliation(s)
- Wenzhong Zheng
- Department of Urology, Fujian Province, Fujian Medical University Union Hospital, Gulou District, 29 Xinquan Road, Fuzhou, 200001, People's Republic of China
| | - Shiqiang Zhang
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, People's Republic of China
| | - Huan Guo
- Department of Urology, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, Guangdong, People's Republic of China
| | - Xiaobao Chen
- Department of Urology, Fujian Province, Fujian Medical University Union Hospital, Gulou District, 29 Xinquan Road, Fuzhou, 200001, People's Republic of China
| | - Zhangcheng Huang
- Department of Urology, Fujian Province, Fujian Medical University Union Hospital, Gulou District, 29 Xinquan Road, Fuzhou, 200001, People's Republic of China
| | - Shaoqin Jiang
- Department of Urology, Fujian Province, Fujian Medical University Union Hospital, Gulou District, 29 Xinquan Road, Fuzhou, 200001, People's Republic of China
| | - Mengqiang Li
- Department of Urology, Fujian Province, Fujian Medical University Union Hospital, Gulou District, 29 Xinquan Road, Fuzhou, 200001, People's Republic of China.
| |
Collapse
|
19
|
Arima M, Fujii Y, Sonoda KH. Translational Research in Retinopathy of Prematurity: From Bedside to Bench and Back Again. J Clin Med 2021; 10:331. [PMID: 33477419 PMCID: PMC7830975 DOI: 10.3390/jcm10020331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/09/2021] [Accepted: 01/15/2021] [Indexed: 12/11/2022] Open
Abstract
Retinopathy of prematurity (ROP), a vascular proliferative disease affecting preterm infants, is a leading cause of childhood blindness. Various studies have investigated the pathogenesis of ROP. Clinical experience indicates that oxygen levels are strongly correlated with ROP development, which led to the development of oxygen-induced retinopathy (OIR) as an animal model of ROP. OIR has been used extensively to investigate the molecular mechanisms underlying ROP and to evaluate the efficacy of new drug candidates. Large clinical trials have demonstrated the efficacy of anti-vascular endothelial growth factor (VEGF) agents to treat ROP, and anti-VEGF therapy is presently becoming the first-line treatment worldwide. Anti-VEGF therapy has advantages over conventional treatments, including being minimally invasive with a low risk of refractive error. However, long-term safety concerns and the risk of late recurrence limit this treatment. There is an unmet medical need for novel ROP therapies, which need to be addressed by safe and minimally invasive therapies. The recent progress in biotechnology has contributed greatly to translational research. In this review, we outline how basic ROP research has evolved with clinical experience and the subsequent emergence of new drugs. We discuss previous and ongoing trials and present the candidate molecules expected to become novel targets.
Collapse
Affiliation(s)
- Mitsuru Arima
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 8128582, Japan; (Y.F.); (K.-H.S.)
- Center for Clinical and Translational Research, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka 8128582, Japan
| | - Yuya Fujii
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 8128582, Japan; (Y.F.); (K.-H.S.)
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 8128582, Japan; (Y.F.); (K.-H.S.)
| |
Collapse
|
20
|
Probing the Effects of the FGFR-Inhibitor Derazantinib on Vascular Development in Zebrafish Embryos. Pharmaceuticals (Basel) 2020; 14:ph14010025. [PMID: 33396726 PMCID: PMC7824571 DOI: 10.3390/ph14010025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 11/24/2022] Open
Abstract
Angiogenesis is a fundamental developmental process and a hallmark of cancer progression. Receptor tyrosine kinases (RTK) are targets for cancer therapy which may include their action as anti-angiogenic agents. Derazantinib (DZB) is an inhibitor of the fibroblast growth factor receptors (FGFRs) 1–3 as well as other kinase targets including vascular endothelial growth factor receptor 2 (VEGFR2), colony stimulating factor-1 receptor (CSF1R) and platelet-derived growth factor beta receptor (PDGFRbeta). This study aimed to investigate the effect of DZB on blood vessel morphogenesis and to compare its activity to known specific FGFR and VEGFR inhibitors. For this purpose, we used the developing vasculature in the zebrafish embryo as a model system for angiogenesis in vivo. We show that DZB interferes with multiple angiogenic processes that are linked to FGF and VEGF signalling, revealing a potential dual role for DZB as a potent anti-angiogenic treatment.
Collapse
|
21
|
The membrane-bound and soluble form of melanotransferrin function independently in the diagnosis and targeted therapy of lung cancer. Cell Death Dis 2020; 11:933. [PMID: 33127882 PMCID: PMC7599248 DOI: 10.1038/s41419-020-03124-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 01/28/2023]
Abstract
Melanotransferrin (MFI2) is a newly identified tumor-associated protein, which consists of two forms of proteins, membrane-bound (mMFI2) and secretory (sMFI2). However, little is known about the expression pattern and their relevance in lung cancer. Here, we found that both two forms of MFI2 are highly expressed in lung cancer. The expression of MFI2 in lung cancer was detected by using the public database and qRT-PCR. Overexpression and knockdown cell lines and recombinant sMFI2 protein were used to study the function of mMFI2 and sMFI2. RNA-seq, protein chip, ChIP assay, Immunoprecipitation, ELISA, and immunofluorescence were used to study the molecular biological mechanism of mMFI2 and sMFI2. We found that mMFI2 promoted the expression of EMT’s common marker N-cadherin by downregulating the transcription factor KLI4, which in turn promoted tumor metastasis; sMFI2 could promote the metastasis of autologous tumor cells in an autocrine manner but the mechanism is different from that of mMFI2. In addition, sMFI2 was proved could inhibit the migration of vascular endothelial cells and subsequently enhance angiogenic responses in a paracrine manner. We propose that the expressions and functions of the two forms of MFI2 in lung cancer are relatively independent. Specifically, mMFI2 was a potential lung cancer therapeutic target, while sMFI2 was highly enriched in advanced lung cancer, and could be used as a tumor staging index.
Collapse
|
22
|
Roy A, Patra CR. Vanadium pentoxide nanomaterials and their role in anti-angiogenesis for cancer treatment. Nanomedicine (Lond) 2020; 15:2643-2646. [PMID: 33078973 DOI: 10.2217/nnm-2020-0321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Arpita Roy
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, Telangana 500007, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Chitta Ranjan Patra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, Telangana 500007, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
23
|
Fernandes LM, Khan NM, Trochez CM, Duan M, Diaz-Hernandez ME, Presciutti SM, Gibson G, Drissi H. Single-cell RNA-seq identifies unique transcriptional landscapes of human nucleus pulposus and annulus fibrosus cells. Sci Rep 2020; 10:15263. [PMID: 32943704 PMCID: PMC7499307 DOI: 10.1038/s41598-020-72261-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/19/2020] [Indexed: 12/29/2022] Open
Abstract
Intervertebral disc (IVD) disease (IDD) is a complex, multifactorial disease. While various aspects of IDD progression have been reported, the underlying molecular pathways and transcriptional networks that govern the maintenance of healthy nucleus pulposus (NP) and annulus fibrosus (AF) have not been fully elucidated. We defined the transcriptome map of healthy human IVD by performing single-cell RNA-sequencing (scRNA-seq) in primary AF and NP cells isolated from non-degenerated lumbar disc. Our systematic and comprehensive analyses revealed distinct genetic architecture of human NP and AF compartments and identified 2,196 differentially expressed genes. Gene enrichment analysis showed that SFRP1, BIRC5, CYTL1, ESM1 and CCNB2 genes were highly expressed in the AF cells; whereas, COL2A1, DSC3, COL9A3, COL11A1, and ANGPTL7 were mostly expressed in the NP cells. Further, functional annotation clustering analysis revealed the enrichment of receptor signaling pathways genes in AF cells, while NP cells showed high expression of genes related to the protein synthesis machinery. Subsequent interaction network analysis revealed a structured network of extracellular matrix genes in NP compartments. Our regulatory network analysis identified FOXM1 and KDM4E as signature transcription factor of AF and NP respectively, which might be involved in the regulation of core genes of AF and NP transcriptome.
Collapse
Affiliation(s)
- Lorenzo M Fernandes
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, 30033, USA.,Atlanta VA Medical Center, Decatur, GA, USA
| | - Nazir M Khan
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, 30033, USA.,Atlanta VA Medical Center, Decatur, GA, USA
| | - Camila M Trochez
- Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Meixue Duan
- Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Martha E Diaz-Hernandez
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, 30033, USA.,Atlanta VA Medical Center, Decatur, GA, USA
| | - Steven M Presciutti
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, 30033, USA.,Atlanta VA Medical Center, Decatur, GA, USA
| | - Greg Gibson
- Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, 30033, USA. .,Atlanta VA Medical Center, Decatur, GA, USA.
| |
Collapse
|
24
|
Discovery of small molecules targeting GRP78 for antiangiogenic and anticancer therapy. Eur J Med Chem 2020; 193:112228. [DOI: 10.1016/j.ejmech.2020.112228] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 12/30/2022]
|
25
|
Mei Y, Zhu Y, Teo HY, Liu Y, Song Y, Lim HY, Binte Hanafi Z, Angeli V, Liu H. The indirect antiangiogenic effect of IL-37 in the tumor microenvironment. J Leukoc Biol 2020; 107:783-796. [PMID: 32125036 DOI: 10.1002/jlb.3ma0220-207rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 01/28/2023] Open
Abstract
IL-37, a newly identified IL-1 family cytokine, has been shown to play an important role in inflammatory diseases, autoimmune diseases, and carcinogenesis. IL-37 has been suggested to suppress tumoral angiogenesis, whereas some publications showed that IL-37 promoted angiogenesis through TGF-β signaling in both physiologic and pathologic conditions. Therefore, the function of IL-37 in tumoral angiogenesis is not clear and the underlying mechanism is not known. In this current study, we investigated the direct role of IL-37 on endothelial cells, as well as its indirect effect on angiogenesis through functioning on tumor cells both in vitro and in vivo. We found that IL-37 treatment directly promoted HUVEC migration and tubule formation, indicating IL-37 as a proangiogenic factor. Surprisingly, the supernatants from IL-37 overexpressing tumor cell line promoted HUVEC apoptosis and inhibited its migration and tubule formation. Furthermore, we demonstrated that IL-37 suppressed tumor angiogenesis in a murine orthotopic hepatocellular carcinoma model, suggesting its dominant antiangiogenesis role in vivo. Moreover, microarray and qPCR analysis demonstrated that IL-37 reduced the expressions of proangiogenic factors and increased the expressions of antiangiogenic factors by tumor cells. Matrix metalloproteinase (MMP)2 expression was significantly decreased by IL-37 in both cell lines and murine tumor models. MMP9 and vascular endothelial growth factor expressions were also reduced in murine tumors overexpressing IL-37, as well as in cell lines overexpressing IL-37 under hypoxic conditions. In conclusion, although IL-37 could exert direct proangiogenic effects on endothelial cells, it plays an antiangiogenic role via modulating proangiogenic and antiangiogenic factor expressions by tumor cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Yu Mei
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Ying Zhu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Huey Yee Teo
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Yonghao Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Yuan Song
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Hwee Ying Lim
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Zuhairah Binte Hanafi
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Veronique Angeli
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| |
Collapse
|
26
|
Barui AK, Nethi SK, Haque S, Basuthakur P, Patra CR. Recent Development of Metal Nanoparticles for Angiogenesis Study and Their Therapeutic Applications. ACS APPLIED BIO MATERIALS 2019; 2:5492-5511. [DOI: 10.1021/acsabm.9b00587] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Ayan Kumar Barui
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, Telangana 500007, India
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Susheel Kumar Nethi
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, Telangana 500007, India
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Shagufta Haque
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, Telangana 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Papia Basuthakur
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, Telangana 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Chitta Ranjan Patra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, Telangana 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
27
|
Phenotypic characteristics of human bone marrow-derived endothelial progenitor cells in vitro support cell effectiveness for repair of the blood-spinal cord barrier in ALS. Brain Res 2019; 1724:146428. [PMID: 31493389 DOI: 10.1016/j.brainres.2019.146428] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/18/2019] [Accepted: 08/28/2019] [Indexed: 01/01/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) was recently recognized as a neurovascular disease. Accumulating evidence demonstrated blood-spinal-cord barrier (BSCB) impairment mainly via endothelial cell (EC) degeneration in ALS patients and animal models. BSCB repair may be a therapeutic approach for ALS. We showed benefits of human bone marrow endothelial progenitor cell (hBMEPC) transplantation into symptomatic ALS mice on barrier restoration; however, cellular mechanisms remain unclear. The study aimed to characterize hBMEPCs in vitro under normogenic conditions. hBMEPCs were cultured at different time points. Enzyme-linked immunosorbent assay (ELISA) was used to detect concentrations of angiogenic factors (VEGF-A, angiogenin-1, and endoglin) and angiogenic inhibitor endostatin in conditioned media. Double immunocytochemical staining for CD105, ZO-1, and occludin with F-actin was performed. Results showed predominantly gradual significant post-culture increases of VEGF-A and angiogenin-1 levels. Cultured cells displayed distinct rounded or elongated cellular morphologies and positively immunoexpressed for CD105, indicating EC phenotype. Cytoskeletal F-actin filaments were re-arranged according to cell morphologies. Immunopositive expressions for ZO-1 were detected near inner cell membrane and for occludin on cell membrane surface of adjacent hBMEPCs. Together, secretion of angiogenic factors by cultured cells provides evidence for a potential mechanism underlying endogenous EC repair in ALS through hBMEPC transplantation, leading to restored barrier integrity. Also, ZO-1 and occludin immunoexpressions, confirming hBMEPC interactions in vitro, may reflect post-transplant cell actions in vivo.
Collapse
|
28
|
Liu C, Chen Y, Deng Y, Dong Y, Jiang J, Chen S, Kang W, Deng J, Sun H. Survival-based bioinformatics analysis to identify hub genes and key pathways in non-small cell lung cancer. Transl Cancer Res 2019; 8:1188-1198. [PMID: 35116861 PMCID: PMC8797769 DOI: 10.21037/tcr.2019.06.35] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/21/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Lung cancer is one of the leading causes of cancer mortality worldwide. Here, we performed an integrative bioinformatics analysis to screen hub genes and critical pathways in non-small cell lung cancer (NSCLC) based on the overall survival rate of differentially expressed genes (DEGs). METHODS Four datasets from the gene expression omnibus (GEO) were used to identify the DEGs. To obtain robust DEGs in NSCLC, only the DEGs that co-existed in the four datasets were selected for subsequent analysis. To identify the genes correlated with overall survival, the overall survival of these genes was then analyzed using the Kaplan-Meier plotter database. The genes significantly correlated with survival were used to perform gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) analysis; next, these genes were used to construct a protein-protein interaction network. MCODE and CytoHubba were used to identify the clusters and hub genes. Finally, the hub genes were validated in the Cancer Genome Atlas (TCGA) and the Human Protein Atlas (HPA). RESULTS We found 522 up-regulated DEGs, and 989 down-regulated DEGs between the NSCLC and normal lung tissue, and 895 of them were correlated with a higher overall survival. GO analysis showed that the DEGs that were associated with a higher overall survival were enriched in cell division, cell cycle, DNA replication, angiogenesis, and cell migration. KEGG analysis was consistent with GO analysis and showed that p53 signaling pathway, pyrimidine metabolism, cGMP-PKG signaling pathway and renin secretion pathway were associated with overall survival in NSCLC. In the protein-protein analysis, we identified seven clusters and six hub genes which were BUB1B, CCNB1, CENPE, KIF18A, NDC10, and MAD2L1. Of these genes, CENPE and KIF18A had not been reported until now. Finally, the dysregulated expression of the six hub genes was validated by the data from the TCGA and HPA. CONCLUSIONS We identified the hub genes and potential mechanisms of NSCLC based on multiple-microarray analysis and overall survival; then, validated the hub genes in the TCGA and HPA database. These hub genes may serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Chunliang Liu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu Chen
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yuqi Deng
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu Dong
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jixuan Jiang
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Si Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wenfeng Kang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiong Deng
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Haipeng Sun
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
29
|
Clavreul A, Pourbaghi-Masouleh M, Roger E, Menei P. Nanocarriers and nonviral methods for delivering antiangiogenic factors for glioblastoma therapy: the story so far. Int J Nanomedicine 2019; 14:2497-2513. [PMID: 31040671 PMCID: PMC6461002 DOI: 10.2147/ijn.s194858] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels, is an essential component of glioblastoma (GB) progression. The development of angiogenesis inhibitor therapy, including treatments targeting vascular endothelial growth factor (VEGF) in particular, raised new hopes for the treatment of GB, but no Phase III clinical trial to date has reported survival benefits relative to standard treatment. There are several possible reasons for this limited efficacy, including VEGF-independent angiogenesis, induction of tumor invasion, and inefficient antiangiogenic factor delivery to the tumor. Efforts have been made to overcome these limitations by identifying new angiogenesis inhibitors that target angiogenesis through different mechanisms of action without inducing tumor invasion, and through the development of viral and nonviral delivery methods to improve antiangiogenic activity. Herein, we describe the nonviral methods, including convection-enhanced delivery devices, implantable polymer devices, nanocarriers, and cellular vehicles, to deliver antiangiogenic factors. We focus on those evaluated in intracranial (orthotopic) animal models of GB, the most relevant models of this disease, as they reproduce the clinical scenario of tumor progression and therapy response encountered in GB patients.
Collapse
Affiliation(s)
- Anne Clavreul
- Department of Neurosurgery, CHU, Angers, France, .,CRCINA, INSERM, University of Nantes, University of Angers, Angers, France,
| | - Milad Pourbaghi-Masouleh
- CRCINA, INSERM, University of Nantes, University of Angers, Angers, France, .,Division of Drug Delivery and Tissue Engineering, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Emilie Roger
- MINT, INSERM 1066, CNRS 6021, University of Angers, Angers, France
| | - Philippe Menei
- Department of Neurosurgery, CHU, Angers, France, .,CRCINA, INSERM, University of Nantes, University of Angers, Angers, France,
| |
Collapse
|
30
|
Zhou K, Zhang JW, Wang QZ, Liu WY, Liu JL, Yao L, Cai MM, Ni SY, Cai QY, Wang GJ, Zhou F. Apatinib, a selective VEGFR2 inhibitor, improves the delivery of chemotherapeutic agents to tumors by normalizing tumor vessels in LoVo colon cancer xenograft mice. Acta Pharmacol Sin 2019; 40:556-562. [PMID: 29977004 DOI: 10.1038/s41401-018-0058-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/31/2018] [Indexed: 12/12/2022] Open
Abstract
Tumor vascular normalization has been proposed as a therapeutic strategy for malignant neoplasms, which can also interpret the synergistic effect of anti-angiogenesis agents combined with chemotherapy. Apatinib (Apa), a highly selective VEGFR2 inhibitor, attracts much attentions due to its encouraging anticancer activity, especially in the clinical trials of combined treatment. In this study, we investigated whether Apa could promote vascular normalization in tumor in a certain time window. Mice bearing LoVo colon cancer xenograft were orally administrated Apa (150 mg kg-1 per day) for 5, 7, 10, or 12 days. Apa significantly inhibited tumor growth and decreased the microvessel density. Using multi-photon microscopy and electron microscopy, we found that Apa improved tumor vessel morphology by pruning distorted vessel branches and decreased the gap between endothelial cells after a 7-day treatment. Furthermore, Apa decreased vessel leakage and increased pericyte coverage on vascular endothelial cells, suggesting that tumor vessels were more mature and integrated. The intratumoral distribution of adriamycin (ADR) in Apa group was improved from day 7 to 10 without change in plasma drug concentration. Tumor blood perfusion was also increased in this window, and the expression of hypoxia induced factor 1α was downregulated, suggesting the effect of Apa on alleviating tumor hypoxic micro-environment. In conclusion, Apa may improve the effective perfusion of tumor vessels and increase the intratumoral distribution of ADR in a certain time window via normalizing tumor vessels. This normalization window (7 to 10 days of treatment) may contribute to develop a regimen of combined medication in clinic use of Apa.
Collapse
|
31
|
Wei J, Xue Y, Huo X, Han R, Su X, Jin Y, Zhao W, Chen Y, Zhang H, Dai J, Chen J. Establishment and characterization of the GC-030-35 cell line derived from gastric hepatoid adenocarcinoma. Cancer Manag Res 2019; 11:1275-1287. [PMID: 30799954 PMCID: PMC6371931 DOI: 10.2147/cmar.s186416] [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] [Indexed: 11/23/2022] Open
Abstract
Purpose Gastric hepatoid adenocarcinoma is a rare subtype of primary gastric cancer and is a high-grade form of malignancy. However, the pathogenesis and molecular biology of gastric hepatoid adenocarcinoma remain poorly understood. The aim of this study was to establish and characterize a new human gastric hepatoid adenocarcinoma cell line, GC-030-35. Materials and methods The GC-030-35 cell line was established from tumor cells from a 58-year-old Chinese man with gastric hepatoid adenocarcinoma. The cultured cells underwent immunocytochemistry and flow cytometry to confirm the tumor cell phenotype. RNA sequencing was performed to analyze the differences in gene expression between GC-030-35 cells compared with normal gastric epithelial cells. A zebrafish assay was performed. Gene enrichment analysis and interrogation of the bioinformatics databases, the Gene Ontology (GO) database and the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, were used for pathway analysis. Results Flow cytometry analysis of the GC-030-35 cells showed a positive expression rate for CD44+ of 10.7%, high cell clonality, an average plating efficiency of 32%, cell-doubling time of 29.2 hours, and cell proliferation for >15 generations in serial culture. The zebrafish assay showed the ability of the GC-030-35 cells to proliferate, promote angiogenesis, and metastasize. RNA sequencing identified the functional clustering of 6,601 differentially expressed genes of GC-030-35, which were significantly different when compared with nonneoplastic gastric epithelial cells. Pathway enrichment analysis and interrogation of the GO and KEGG bioinformatics databases identified genes for microbial metabolism in diverse environments (63 genes), metabolism of xenobiotics by cytochrome P450 (CYP450; 25 genes), and the drug metabolism cytochrome P450 (28 genes). Conclusion A human gastric hepatoid adenocarcinoma cell line, GC-030-35, was developed and characterized by comparison with normal gastric epithelial cells. Bioinformatics and gene analysis data showed that the CYP450 gene was significantly differentially expressed by GC-030-35 cells.
Collapse
Affiliation(s)
- Jingsun Wei
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China,
| | - Yiqi Xue
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China,
| | - Xinying Huo
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China,
| | - Rongbo Han
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China,
| | - Xinyu Su
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China,
| | - Yan Jin
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China,
| | - Wenjing Zhao
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China,
| | - Yuetong Chen
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China,
| | - Honghong Zhang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China,
| | - Jiali Dai
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China,
| | - Jinfei Chen
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China,
| |
Collapse
|
32
|
Kim JY, Kim YM. Tumor endothelial cells as a potential target of metronomic chemotherapy. Arch Pharm Res 2019; 42:1-13. [PMID: 30604201 DOI: 10.1007/s12272-018-01102-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022]
Abstract
Drug resistance and toxic side effects are major therapeutic hurdles affecting cancer patients receiving conventional chemotherapy based on the maximum tolerated dose. Metronomic chemotherapy (MCT), a new therapeutic approach developed to avoid these problems generally, consists of the continuous administration of low-dose cytotoxic agents without extended intervals. This therapy targets the tumor microenvironment, rather than exerting a direct effect on tumor cells. As a result, the MCT regimen functionally impairs tumor endothelial cells and circulating endothelial progenitor cells, leading to tumor dormancy via anti-angiogenesis. Over the past 10 years, several studies have highlighted the impact of MCT on the tumor microenvironment and angiogenesis and demonstrated its potential as a switch from the pro-angiogenic to the anti-angiogenic state. However, the mechanisms of action are still obscure. Here, we systematically review the evidence regarding the anti-angiogenic potential of MCT as a crucial determinant of tumor dormancy and cancer treatment.
Collapse
Affiliation(s)
- Ji Yoon Kim
- Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul, 04763, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry School of Medicine, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, 24341, South Korea.
| |
Collapse
|
33
|
Association of specific gene mutations derived from machine learning with survival in lung adenocarcinoma. PLoS One 2018; 13:e0207204. [PMID: 30419062 PMCID: PMC6231670 DOI: 10.1371/journal.pone.0207204] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 10/27/2018] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is the second most common cancer in the United States and the leading cause of mortality in cancer patients. Biomarkers predicting survival of patients with lung cancer have a profound effect on patient prognosis and treatment. However, predictive biomarkers for survival and their relevance for lung cancer are not been well known yet. The objective of this study was to perform machine learning with data from The Cancer Genome Atlas of patients with lung adenocarcinoma (LUAD) to find survival-specific gene mutations that could be used as survival-predicting biomarkers. To identify survival-specific mutations according to various clinical factors, four feature selection methods (information gain, chi-squared test, minimum redundancy maximum relevance, and correlation) were used. Extracted survival-specific mutations of LUAD were applied individually or as a group for Kaplan-Meier survival analysis. Mutations in MMRN2 and GMPPA were significantly associated with patient mortality while those in ZNF560 and SETX were associated with patient survival. Mutations in DNAJC2 and MMRN2 showed significant negative association with overall survival while mutations in ZNF560 showed significant positive association with overall survival. Mutations in MMRN2 showed significant negative association with disease-free survival while mutations in DRD3 and ZNF560 showed positive associated with disease-free survival. Mutations in DRD3, SETX, and ZNF560 showed significant positive association with survival in patients with LUAD while the opposite was true for mutations in DNAJC2, GMPPA, and MMRN2. These gene mutations were also found in other cohorts of LUAD, lung squamous cell carcinoma, and small cell lung cancer. In LUAD of Pan-Lung Cancer cohort, mutations in GMPPA, DNAJC2, and MMRN2 showed significant negative associations with survival of patients while mutations in DRD3 and SETX showed significant positive association with survival. In this study, machine learning was conducted to obtain information necessary to discover specific gene mutations associated with the survival of patients with LUAD. Mutations in the above six genes could predict survival rate and disease-free survival rate in patients with LUAD. Thus, they are important biomarker candidates for prognosis.
Collapse
|
34
|
Renganathan B, Durairaj V, Kirman DC, Esubonteng PKA, Ang SK, Ge R. Recombinant TSR1 of ADAMTS5 Suppresses Melanoma Growth in Mice via an Anti-angiogenic Mechanism. Cancers (Basel) 2018; 10:cancers10060192. [PMID: 29891754 PMCID: PMC6025205 DOI: 10.3390/cancers10060192] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/07/2018] [Accepted: 06/08/2018] [Indexed: 11/17/2022] Open
Abstract
Inhibiting tumor angiogenesis is a well-established approach for anticancer therapeutic development. A Disintegrin-like and Metalloproteinase with ThromboSpondin Motifs 5 (ADAMTS5) is a secreted matrix metalloproteinase in the ADAMTS family that also functions as an anti-angiogenic/anti-tumorigenic molecule. Its anti-angiogenic/anti-tumorigenic function is independent from its proteinase activity, but requires its first thrombospondin type 1 repeat (TSR1). However, it is not known if recombinant TSR1 (rTSR1) can function as an anticancer therapeutic. In this report, we expressed and purified a 75-residue recombinant TSR1 polypeptide from E. coli and investigated its ability to function as an anticancer therapeutic in mice. We demonstrate that rTSR1 is present in the blood circulation as well as in the tumor tissue at 15 min post intraperitoneal injection. Intraperitoneal delivery of rTSR1 potently suppressed subcutaneous B16F10 melanoma growth as a single agent, accompanied by diminished tumor angiogenesis, increased apoptosis, and reduced cell proliferation in the tumor tissue. Consistently, rTSR1 dose-dependently induced the apoptosis of cultured human umbilical vein endothelial cells (HUVECs) in a caspase-dependent manner. This work indicates that rTSR1 of ADAMTS5 can function as a potent anticancer therapy in mice. It thus has the potential to be further developed into an anticancer drug.
Collapse
Affiliation(s)
- Bhuvanasundar Renganathan
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117543, Singapore.
| | - Vinoth Durairaj
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117543, Singapore.
| | - Dogan Can Kirman
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117543, Singapore.
| | - Paa Kow A Esubonteng
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117543, Singapore.
| | - Swee Kim Ang
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117543, Singapore.
| | - Ruowen Ge
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117543, Singapore.
| |
Collapse
|
35
|
Shan Y, Wang B, Zhang J. New strategies in achieving antiangiogenic effect: Multiplex inhibitors suppressing compensatory activations of RTKs. Med Res Rev 2018; 38:1674-1705. [DOI: 10.1002/med.21517] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/19/2018] [Accepted: 05/19/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Yuanyuan Shan
- Department of Pharmacy; The First Affiliated Hospital of Xi'an Jiaotong University; Xi'an China
| | - Binghe Wang
- Department of Chemistry; Center for Diagnostics and Therapeutics; Georgia State University; Atlanta GA USA
| | - Jie Zhang
- School of Pharmacy, Health Science Center; Xi'an Jiaotong University; Xi'an China
| |
Collapse
|
36
|
Natale G, Bocci G. Does metronomic chemotherapy induce tumor angiogenic dormancy? A review of available preclinical and clinical data. Cancer Lett 2018; 432:28-37. [PMID: 29885517 DOI: 10.1016/j.canlet.2018.06.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/11/2018] [Accepted: 06/03/2018] [Indexed: 02/08/2023]
Abstract
Tumor dormancy is the ability of cancer cells to survive in a non-proliferating state. This condition can depend on three main mechanisms: cell cycle arrest (quiescence or cell dormancy), immunosurveillance (immunologic dormancy), or lack of functional blood vessels (angiogenic dormancy). In particular, under angiogenic dormancy, cancer cell proliferation is counterbalanced by apoptosis owing to poor vascularization, impeding tumor mass expansion beyond a microscopic size, with an asymptomatic and non-metastatic state. Tumor vasculogenic or non-angiogenic switch is essential to promote escape from tumor dormancy, leading to tumor mass proliferation and metastasis. In avascular lesions angiogenesis process results blocked from the equilibrium between pro- and anti-angiogenic factors, such as vascular endothelial growth factor (VEGF) and thrombospondin-1 (TSP-1), respectively. The angiogenic switch mainly depends on the disruption of this balance, in favor of pro-angiogenic factors, and on the recruitment of circulating endothelial progenitors (CEPs) that promote the formation of new blood vessels. Metronomic chemotherapy, the regular intake of doses able to sustain low but active concentrations of chemotherapeutic drugs during protracted time periods, is an encouraging therapeutic approach that has shown to upregulate anti-angiogenic factors such as TSP-1 and decline pro-angiogenic factors such as VEGF, suppressing the proangiogenic cells such as CEPs. In this perspective, metronomic chemotherapy may be one of the available therapeutic approaches capable to modulate favorably the angiogenic tumor dormancy, but further research is essential to better define this particular characteristic.
Collapse
Affiliation(s)
- Gianfranco Natale
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, and Museo di Anatomia Umana ''Filippo Civinini'', Università di Pisa, Pisa, Italy
| | - Guido Bocci
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy.
| |
Collapse
|
37
|
Yehya AHS, Asif M, Petersen SH, Subramaniam AV, Kono K, Majid AMSA, Oon CE. Angiogenesis: Managing the Culprits behind Tumorigenesis and Metastasis. MEDICINA (KAUNAS, LITHUANIA) 2018; 54:E8. [PMID: 30344239 PMCID: PMC6037250 DOI: 10.3390/medicina54010008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 03/22/2018] [Accepted: 03/22/2018] [Indexed: 12/14/2022]
Abstract
Deregulated angiogenesis has been identified as a key contributor in a number of pathological conditions including cancer. It is a complex process, which involves highly regulated interaction of multiple signalling molecules. The pro-angiogenic signalling molecule, vascular endothelial growth factor (VEGF) and its cognate receptor 2 (VEGFR-2), which is often highly expressed in majority of human cancers, plays a central role in tumour angiogenesis. Owing to the importance of tumour vasculature in carcinogenesis, tumour blood vessels have emerged as an excellent therapeutic target. The anti-angiogenic therapies have been shown to arrest growth of solid tumours through multiple mechanisms, halting the expansion of tumour vasculature and transient normalization of tumour vasculature which help in the improvement of blood flow resulting in more uniform delivery of cytotoxic agents to the core of tumour mass. This also helps in reduction of hypoxia and interstitial pressure leading to reduced chemotherapy resistance and more uniform delivery of cytotoxic agents at the targeted site. Thus, complimentary combination of different agents that target multiple molecules in the angiogenic cascade may optimize inhibition of angiogenesis and improve clinical benefit in the cancer patients. This review provides an update on the current trend in exploitation of angiogenesis pathways as a strategy in the treatment of cancer.
Collapse
Affiliation(s)
- Ashwaq Hamid Salem Yehya
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia.
| | - Muhammad Asif
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan.
| | - Sven Hans Petersen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117543, Singapore.
| | - Ayappa V Subramaniam
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia.
| | - Koji Kono
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117543, Singapore.
- Department of Surgery, National University of Singapore, Singapore 117543, Singapore.
- School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan.
| | - Amin Malik Shah Abdul Majid
- EMAN Testing and Research Laboratories, Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia.
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, Australian National University, Acton 0200, Australia.
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia.
| |
Collapse
|
38
|
Wang SS, Cen X, Liang XH, Tang YL. Macrophage migration inhibitory factor: a potential driver and biomarker for head and neck squamous cell carcinoma. Oncotarget 2018; 8:10650-10661. [PMID: 27788497 PMCID: PMC5354689 DOI: 10.18632/oncotarget.12890] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/19/2016] [Indexed: 02/05/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF), a pleiotropic proinflammatory cytokine, has been showed to be associated with the immunopathogenesis of many diseases. Recent study demonstrated that MIF promoted tumorigenesis and tumor progression and played a critical role in various kinds of human cancer including head and neck squamous cell carcinoma(HNSCC). Hence, in this paper we retrospected the relationship between MIF and angiogenesis, epithelial-mesenchymal transition (EMT), inflammation, immune response, hypoxia microenvironment, and discussed whether it is a promising biomarker for diagnosis and supervisor of HNSCC.
Collapse
Affiliation(s)
- Sha-Sha Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China.,Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China
| | - Xiao Cen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China.,Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China.,Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China.,Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China
| |
Collapse
|
39
|
Looking for the Word "Angiogenesis" in the History of Health Sciences: From Ancient Times to the First Decades of the Twentieth Century. World J Surg 2018; 41:1625-1634. [PMID: 27491322 DOI: 10.1007/s00268-016-3680-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This review deals with the origin of the term "angiogenesis", with an attention to John Hunter who is credited with this neologism. A part of the literature refers to a Hunter's work dating 1787, and the other part claims the first use of the term "angiogenesis" in the Hunter's masterpiece published in 1794. Since we were unable to find the term "angiogenesis" in Hunter's works, this review attempts to bring a new contribution to the historical research of this important concept, moving from ancient times to the first decades of the twentieth century, when "angiogenesis" begun to appear on titles of scientific articles. The development of the knowledge on the cardiocirculatory system and the principal steps of this fascinating subject were examined, with particular regard to microvascular bed and vessel sprouting, and to the intriguing observations on blood vessel neoformation that have been also made in the premicroscopic era. In Hunter's works, the concept of angiogenesis indeed emerges, but not the term "angiogenesis". The scientific language occurring during Hunter's time was still old-fashioned, and the term "angiogenesis" was not one of those he used, rather a much later neologism that sounds too modern to appear in that context. Would the first appearance of the term "angiogenesis" occur in late nineteenth century in studies dealing with embryogenesis and organ vascularization? The present study aims to explore the scientific literature and to open a debate to better define this matter.
Collapse
|
40
|
Aguilar-Rojas A, Maya-Núñez G, Huerta-Reyes M, Pérez-Solis MA, Silva-García R, Guillén N, Olivo-Marin JC. Activation of human gonadotropin-releasing hormone receptor promotes down regulation of ARHGAP18 and regulates the cell invasion of MDA-MB-231 cells. Mol Cell Endocrinol 2018; 460:94-103. [PMID: 28709956 DOI: 10.1016/j.mce.2017.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/29/2017] [Accepted: 07/10/2017] [Indexed: 02/08/2023]
Abstract
The Gonadotropin-Releasing Hormone Receptor (GnRHR) is expressed mainly in the gonadotrope membrane of the adenohypophysis and its natural ligand, the Gonadotropin-Releasing Hormone (GnRH), is produced in anterior hypothalamus. Furthermore, both molecules are also present in the membrane of cells derived from other reproductive tissues such as the breast, endometrium, ovary, and prostate, as well as in tumors derived from these tissues. The functions of GnRH receptor and its hormone in malignant cells have been related with the decrease of proliferation and the invasiveness of those tumors however, little is known about the molecules associated with the signaling pathways regulated by both molecules in malignant cells. To further analyze the potential mechanisms employed by the GnRHR/GnRH system to reduce the tumorigenesis of the highly invasive breast cancer cell line MDA-MB-231, we performed microarrays experiments to evaluated changes in genes expression and validate these modifications by functional assays. We show that activation of human GnRHR is able to diminish the expression and therefore functions of the Rho GTPase-Activating Protein 18 (ARHGAP18). Decrease of this GAP following GnRHR activation, correlates to the higher of cell adhesion and also with reduction of tumor cell invasion, supporting the notion that GnRHR triggers intracellular signaling pathways that acts through ARHGAP18. On the contrary, although a decline of cellular proliferation was observed during GnRHR activation in MDA-MB-231, this was independent of ARHGAP18 showing the complex system in which is involved the signaling pathways regulated by the GnRHR/GnRH system.
Collapse
Affiliation(s)
- Arturo Aguilar-Rojas
- Instituto Mexicano del Seguro Social (IMSS), Unidad de Investigación Médica en Medicina Reproductiva, UMAE No. 4, Ciudad de México, Mexico; Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, F-75015 Paris, France; Centre National de la Recherche Scientifique, CNRS UMR3691, 25 Rue du Dr Roux, F-75015 Paris, France.
| | - Guadalupe Maya-Núñez
- Instituto Mexicano del Seguro Social (IMSS), Unidad de Investigación Médica en Medicina Reproductiva, UMAE No. 4, Ciudad de México, Mexico
| | - Maira Huerta-Reyes
- IMSS, Unidad de Investigación Médica en Farmacología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI (CMN-SXXI), Ciudad de México, Mexico
| | - Marco Allán Pérez-Solis
- Instituto Mexicano del Seguro Social (IMSS), Unidad de Investigación Médica en Medicina Reproductiva, UMAE No. 4, Ciudad de México, Mexico
| | - Raúl Silva-García
- IMSS, Unidad de Investigación Médica en Inmunología, Hospital de Pediatría, CMN-SXXI, Ciudad de México, Mexico
| | - Nancy Guillén
- Centre National de la Recherche Scientifique, CNRS-ERL9195, 25 Rue du Dr Roux, F-75015 Paris, France
| | - Jean-Christophe Olivo-Marin
- Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, F-75015 Paris, France; Centre National de la Recherche Scientifique, CNRS UMR3691, 25 Rue du Dr Roux, F-75015 Paris, France
| |
Collapse
|
41
|
Munro DAD, Hohenstein P, Coate TM, Davies JA. Refuting the hypothesis that semaphorin-3f/neuropilin-2 exclude blood vessels from the cap mesenchyme in the developing kidney. Dev Dyn 2017; 246:1047-1056. [PMID: 28929539 DOI: 10.1002/dvdy.24592] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/16/2017] [Accepted: 09/16/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND During murine kidney development, new cortical blood vessels form and pattern in cycles that coincide with cycles of collecting duct branching and the accompanying splitting of the cap mesenchyme (nephron progenitor cell populations that "cap" collecting duct ends). At no point in the patterning cycle do blood vessels enter the cap mesenchyme. We hypothesized that the exclusion of blood vessels from the cap mesenchyme may be controlled, at least in part, by an anti-angiogenic signal expressed by the cap mesenchyme cells. RESULTS We show that semaphorin-3f (Sema3f), a known anti-angiogenic factor, is expressed in cap mesenchymal cells and its receptor, neuropilin-2 (Nrp2), is expressed by newly forming blood vessels in the cortex of the developing kidney. We hypothesized that Sema3f/Nrp2 signaling excludes vessels from the cap mesenchyme. Genetic ablation of Sema3f and of Nrp2, however, failed to result in vessels invading the cap mesenchyme. CONCLUSIONS Despite complementary expression patterns, our data suggest that Sema3f and Nrp2 are dispensable for the exclusion of vessels from the cap mesenchyme during kidney development. These results should provoke additional experiments to ascertain the biological significance of Sema3f/Nrp2 expression in the developing kidney. Developmental Dynamics 246:1047-1056, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David A D Munro
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter Hohenstein
- The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Thomas M Coate
- Georgetown University, Department of Biology, Washington, DC
| | - Jamie A Davies
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
42
|
Aleskandarany MA, Sonbul S, Surridge R, Mukherjee A, Caldas C, Diez-Rodriguez M, Ashankyty I, Albrahim KI, Elmouna AM, Aneja R, Martin SG, Ellis IO, Green AR, Rakha EA. Rho-GTPase activating-protein 18: a biomarker associated with good prognosis in invasive breast cancer. Br J Cancer 2017; 117:1176-1184. [PMID: 28829761 PMCID: PMC5674094 DOI: 10.1038/bjc.2017.261] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/03/2017] [Accepted: 07/14/2017] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND The prognostic value of lymphovascular invasion (LVI) in breast cancer (BC) has been demonstrated in several independent studies. However, identification of driver molecules for LVI remains a challenging task. Large-scale transcriptomic profiling of histologically validated LVI can potentially identify genes that regulate LVI. METHODS Integrative bio-informatics analyses of the METABRIC study were performed utilising a subset of strictly defined LVI using histological and immunohistochemical (IHC) criteria. ARHGAP18 was among the top differentially expressed genes between LVI+ and LVI- BC with a 1.8-fold change. The prognostic impact of ARHGAP18 gene expression was assessed in the METABRIC data set (n=1980) and externally validated using the online BC gene expression data sets utilising bc-GenExMiner v4.0 (n=2016). Subsequently, ARHGAP18 protein expression was assessed on a large cohort of invasive BC (n=959) with long-term follow-up using IHC. RESULTS Pooled analysis of ARHGAP18 mRNA expression showed that overexpression was associated with better outcome (P<0.001, hazard ratio (HR)=0.82, 95% CI 0.75-0.90). ARHGAP18 protein was expressed in the cytoplasm and nuclei of the tumour cells and its expression was positively associated with good prognostic variables. Lack of cytoplasmic expression showed associations with LVI (P=0.006), epithelial-mesenchymal transition and the HER+ subtype (P=0.01). Loss of nuclear expression was associated with higher grade, HER2+ and high Ki67LI (P=0.001). Cytoplasmic and nuclear expression showed a positive association with improved survival independent of other variables (P=0.01, HR=0.74, 95% CI 0.60-87). CONCLUSIONS ARHGAP18 expression at transcriptomic and protein levels is associated with improved patients' outcomes whose deregulation may play a role in tumour progression and the development of LVI in BC. Further assessment of its potential therapeutic value in BC is warranted.
Collapse
MESH Headings
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Lobular/genetics
- Carcinoma, Lobular/metabolism
- Carcinoma, Lobular/pathology
- Epithelial-Mesenchymal Transition/genetics
- Female
- GTPase-Activating Proteins/genetics
- GTPase-Activating Proteins/metabolism
- Humans
- Immunohistochemistry
- Middle Aged
- Neoplasm Invasiveness
- Neoplasm Staging
- Prognosis
- Proportional Hazards Models
- RNA, Messenger/metabolism
- Tumor Burden
Collapse
Affiliation(s)
- Mohammed A Aleskandarany
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
- Pathology Department, Faculty of Medicine, Menoufyia University, Menoufyia 110532, Egypt
| | - Sultan Sonbul
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
| | - Rachel Surridge
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
| | - Abhik Mukherjee
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
| | - Carlos Caldas
- Cancer Research UK, Cambridge Research Institute, Cambridge CB 0RE, UK
| | - Maria Diez-Rodriguez
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
| | - Ibraheem Ashankyty
- Molecular Diagnostics and Personalised Therapeutics Unit, University of Ha'il, Ha'il 2440, Saudi Arabia
| | - Khalil I Albrahim
- Molecular Diagnostics and Personalised Therapeutics Unit, University of Ha'il, Ha'il 2440, Saudi Arabia
| | - Ahmed M Elmouna
- Molecular Diagnostics and Personalised Therapeutics Unit, University of Ha'il, Ha'il 2440, Saudi Arabia
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Stewart G Martin
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
| | - Ian O Ellis
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
| | - Andrew R Green
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
| | - Emad A Rakha
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
- Pathology Department, Faculty of Medicine, Menoufyia University, Menoufyia 110532, Egypt
| |
Collapse
|
43
|
He Y, Luo J, Chen Y, Zhou X, Yu S, Jin L, Xiao X, Jia S, Liu Q. ARHGAP18 is a novel gene under positive natural selection that influences HbF levels in β-thalassaemia. Mol Genet Genomics 2017; 293:207-216. [DOI: 10.1007/s00438-017-1377-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 09/25/2017] [Indexed: 10/18/2022]
|
44
|
Abstract
Angiogenesis is the process of developing new blood vessels from the original vascular
network; it is necessary for normal physiological processes, such as embryonic development
and wound healing. Angiogenesis is also involved in pathological events, including
myocardial ischemia and tumor growth. To investigate the molecular mechanisms of this
important process, a variety of methods and models are employed. These strategies can also
be used to provide insight into the etiology of angiogenesis-related diseases, thereby
contributing to the development of new diagnostics and treatments. Commonly used animal
models include the chorioallantoic membrane and yolk sac membrane of chick embryos, the
mouse retina and aortic ring, and angiogenesis reactors implanted into mice. These animal
models have been instrumental in the study of the angiogenic process. For example, the
chorioallantoic membrane undergoes robust angiogenesis during the development of chick
embryos, and, because its surface is easily accessible, this membrane provides a
convenient model for experimentation. Here, we discuss the methods that employ animal
models for the imaging and quantification of angiogenesis. In addition, we propose
potential novel directions for future investigations in this area.
Collapse
Affiliation(s)
- Min Liu
- Key Laboratory of Animal Resistance Biology of Shandong Province, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, 88 East Wenhua Road, Jinan, Shandong 250014, P.R. China
| | - Songbo Xie
- Key Laboratory of Animal Resistance Biology of Shandong Province, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, 88 East Wenhua Road, Jinan, Shandong 250014, P.R. China
| | - Jun Zhou
- Key Laboratory of Animal Resistance Biology of Shandong Province, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, 88 East Wenhua Road, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
45
|
In vitro significance of SOCS-3 and SOCS-4 and potential mechanistic links to wound healing. Sci Rep 2017; 7:6715. [PMID: 28751715 PMCID: PMC5532239 DOI: 10.1038/s41598-017-06886-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/22/2017] [Indexed: 01/22/2023] Open
Abstract
Wound healing and the management of chronic wounds represent a significant burden on the NHS. Members of the suppressor of cytokine signalling (SOCS) family have been implicated in the regulation of a range of cellular processes. The current study aims to explore the importance of SOCS-3 and SOCS-4 in regulating cellular traits associated with wound healing. SOCS-3 over-expression and SOCS-4 knockdown mutant lines were generated and verified using q-PCR and western blotting in human keratinocytes (HaCaT) and endothelial cells (HECV). Over-expression of SOCS-3 resulted in a significantly reduced proliferative rate in HaCaT keratinocytes and also enhanced the tubule formation capacity of HECV cells. SOCS-4 knockdown significantly reduced HaCaT migration and HECV cell tubule formation. Suppression of SOCS-4 influenced the responsiveness of HaCaT and HECV cells to EGF and TGFβ and resulted in a dysregulation of phospho-protein expression in HaCaT cells. SOCS-3 and SOCS-4 appear to play regulatory roles in a number of keratinocyte and endothelial cellular traits associated with the wound healing process and may also be able to regulate the responsiveness of these cells to EGF and TGFβ. This implies a potential regulatory role in the wound healing process and, thus highlights their potential as novel therapies.
Collapse
|
46
|
Humphries B, Wang Z, Li Y, Jhan JR, Jiang Y, Yang C. ARHGAP18 Downregulation by miR-200b Suppresses Metastasis of Triple-Negative Breast Cancer by Enhancing Activation of RhoA. Cancer Res 2017; 77:4051-4064. [PMID: 28619708 DOI: 10.1158/0008-5472.can-16-3141] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/13/2017] [Accepted: 06/05/2017] [Indexed: 11/16/2022]
Abstract
Rho GTPases activated in cancer cells drive proliferation, migration, and metastasis. Thus, RhoGAP proteins, which negatively regulate Rho GTPases, are generally thought to function as tumor suppressors. Here this expectation was challenged by characterization of ARHGAP18, a RhoGAP family member that is selectively overexpressed in highly migratory triple-negative breast cancer (TNBC) cells. In human breast tumors, higher ARHGAP18 levels associated with worse overall survival, recurrence-free survival, and metastasis-free survival. In TNBC cells, ARHGAP18 deletion increased RhoA activation but reduced growth, migration, and metastatic capacity. Mechanistic investigations revealed that ARHGAP18 levels were controlled by miR-200b, the enforced expression of which was sufficient to activate RhoA, enhanced formation of focal adhesions and actin stress fibers, and reduced migration and metastasis. Enforced elevation of ARHGAP18 where miR-200b was stably expressed reduced RhoA activity but increased cell migration. Pharmacologic inhibition of the Rho effector kinase ROCK blocked RhoA signaling and reversed the inhibitory effect of miR-200b on cell migration. Finally, ARHGAP18 overexpression or ROCK inhibition was sufficient to overcome metastatic blockade by miR-200b. Taken together, these results define opposing roles for oncogenic ARHGAP18 and tumor suppressive miR-200b in determining TNBC cell migration and metastatic prowess. Cancer Res; 77(15); 4051-64. ©2017 AACR.
Collapse
Affiliation(s)
- Brock Humphries
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Zhishan Wang
- Department of Physiology, Michigan State University, East Lansing, Michigan
- Department of Toxicology and Cancer Biology, Center for Research on Environmental Diseases, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Yunfei Li
- Department of Physiology, Michigan State University, East Lansing, Michigan
- Department of Toxicology and Cancer Biology, Center for Research on Environmental Diseases, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Jing-Ru Jhan
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Yiguo Jiang
- Institute for Chemical Carcinogenesis, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, Guangdong, P.R. China
| | - Chengfeng Yang
- Department of Physiology, Michigan State University, East Lansing, Michigan.
- Department of Toxicology and Cancer Biology, Center for Research on Environmental Diseases, College of Medicine, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
47
|
Mohajeri A, Sanaei S, Kiafar F, Fattahi A, Khalili M, Zarghami N. The Challenges of Recombinant Endostatin in Clinical Application: Focus on the Different Expression Systems and Molecular Bioengineering. Adv Pharm Bull 2017; 7:21-34. [PMID: 28507934 PMCID: PMC5426730 DOI: 10.15171/apb.2017.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 02/01/2017] [Accepted: 02/15/2017] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis plays an essential role in rapid growing and metastasis of the tumors. Inhibition of angiogenesis is a putative strategy for cancer therapy. Endostatin (Es) is an attractive anti-angiogenesis protein with some clinical application challenges including; short half-life, instability in serum and requirement to high dosage. Therefore, production of recombinant endostatin (rEs) is necessary in large scale. The production of rEs is difficult because of its structural properties and is high-cost. Therefore, this review focused on the different expression systems that involved in rEs production including; mammalian, baculovirus, yeast, and Escherichia coli (E. coli) expression systems. The evaluating of the results of different expression systems declared that none of the mentioned systems can be considered to be generally superior to the other. Meanwhile with considering the advantages and disadvantage of E. coli expression system compared with other systems beside the molecular properties of Es, E. coli expression system can be a preferred expression system for expressing of the Es in large scale. Also, the molecular bioengineering and sustained release formulations that lead to improving of its stability and bioactivity will be discussed. Point mutation (P125A) of Es, addition of RGD moiety or an additional zinc biding site to N-terminal of Es , fusing of Es to anti-HER2 IgG or heavy-chain of IgG, and finally loading of the endostar by PLGA and PEG- PLGA nanoparticles and gold nano-shell particles are the effective bioengineering methods to overcome to clinical changes of endostatin.
Collapse
Affiliation(s)
- Abbas Mohajeri
- Department of Biotechnology, Zahravi Pharmaceutical Company, Tabriz, Iran.,Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sarvin Sanaei
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Kiafar
- Department of Biotechnology, Zahravi Pharmaceutical Company, Tabriz, Iran
| | - Amir Fattahi
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Khalili
- Department of Basic Science, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Nosratollah Zarghami
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences,Tabriz, Iran
| |
Collapse
|
48
|
Martínez-Campa C, Menéndez-Menéndez J, Alonso-González C, González A, Álvarez-García V, Cos S. What is known about melatonin, chemotherapy and altered gene expression in breast cancer. Oncol Lett 2017; 13:2003-2014. [PMID: 28454355 PMCID: PMC5403278 DOI: 10.3892/ol.2017.5712] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 11/17/2016] [Indexed: 02/07/2023] Open
Abstract
Melatonin, synthesized in and released from the pineal gland, has been demonstrated by multiple in vivo and in vitro studies to have an oncostatic role in hormone-dependent tumors. Furthermore, several clinical trials point to melatonin as a promising adjuvant molecule to be considered for cancer treatment. In the past few years, evidence of a broader spectrum of action of melatonin as an antitumor agent has arisen; thus, melatonin appears to also have therapeutic effects in several types of hormone-independent cancer, including ovarian, leukemic, pancreatic, gastric and non-small cell lung carcinoma. In the present study, the latest findings regarding melatonin molecular actions when concomitantly administered with either radiotherapy or chemotherapy in cancer were reviewed, with a particular focus on hormone-dependent breast cancer. Finally, the present study discusses which direction should be followed in the next years to definitely clarify whether or not melatonin administration could protect against non-desirable effects (such as altered gene expression and post-translational protein modifications) caused by chemotherapy or radiotherapy treatments. As treatments move towards personalized medicine, comparative gene expression profiling with and without melatonin may be a powerful tool to better understand the antitumor effects of melatonin, the pineal gland hormone.
Collapse
Affiliation(s)
- Carlos Martínez-Campa
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, 39011 Santander, Spain
- Correspondence to: Dr Carlos Martínez-Campa, Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, Av. Cardenal Herrera Oria s/n, 39011 Santander, Spain, E-mail:
| | - Javier Menéndez-Menéndez
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, 39011 Santander, Spain
| | - Carolina Alonso-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, 39011 Santander, Spain
| | - Alicia González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, 39011 Santander, Spain
| | - Virginia Álvarez-García
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot Watt University, EH14 4AS Edinburgh, UK
| | - Samuel Cos
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, 39011 Santander, Spain
| |
Collapse
|
49
|
Huang B, Peng Y, Li J, Li S, Sun Y, Wang D, Yang B, Chan JYW, Yu H, Leung GPH, Hoi MPM, Zhou GC, Lee SMY. An andrographolide derivative AGP-26b exhibiting anti-angiogenic activity in HUVECs and zebrafish via blocking the VEGFA/VEGFR2 signaling pathway. MOLECULAR BIOSYSTEMS 2017; 13:525-536. [DOI: 10.1039/c6mb00641h] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A new andrographolide derivative AGP-26b exhibits anti-angiogenic activity in HUVECs and zebrafish.
Collapse
|
50
|
Takano S, Ishikawa E, Matsuda M, Sakamoto N, Akutsu H, Yamamoto T, Matsumura A. The anti-angiogenic role of soluble-form VEGF receptor in malignant gliomas. Int J Oncol 2016; 50:515-524. [PMID: 28000842 DOI: 10.3892/ijo.2016.3810] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/21/2016] [Indexed: 11/05/2022] Open
|