1
|
Yong R, Mu R, Han C, Chao T, Liu Y, Dong L, Wang C. Optimizing a 5-factor cocktail to prepare reparative macrophages for wound healing. J Leukoc Biol 2025; 117:qiae096. [PMID: 38630870 DOI: 10.1093/jleuko/qiae096] [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: 01/05/2024] [Revised: 03/14/2024] [Accepted: 04/06/2024] [Indexed: 04/19/2024] Open
Abstract
The treatment of nonhealing wounds, such as diabetic ulcers, remains a critical clinical challenge. Recent breakthroughs in cell therapy have shown great promise, with one primary focus on preparing cells with comprehensive reparative functions and foreseeable safety. In our previous study, we recapitulated the proregenerative and immunosuppressive functions of tumor-associated macrophages in non-tumor-derived macrophages, endowing the latter with characteristics for promoting diabetic wound healing-termed tumor-associated macrophage-educated macrophages. To eliminate the use of tumor-derived sources and devise a more controllable method to prepare tumor-associated macrophage-educated macrophage-like cells, in this study, we identify a cocktail comprising 5 recombinant proteins as an essential condition to induce nonpolarized macrophages into therapeutic cells with prohealing functions. The screened 5 factors are osteopontin, macrophage inflammatory protein 2, chemokine (C-C motif) ligand 8, vascular endothelial growth factor B, and macrophage colony-stimulating factor. We demonstrate the rationale for screening these factors and the phenotype of the 5 factor-induced tumor-associated macrophage-educated macrophage-like macrophages prepared from murine bone marrow-derived macrophages, which exhibit angiogenic and immunomodulatory effects in vitro. Then, we induce primary human monocytes from periphery blood into the 5 factor-induced tumor-associated macrophage-educated macrophage-like macrophages, which show prohealing effects in a human primary cell-based ex vivo model (T-Skin™). Our study demonstrates a simple, effective, and controllable approach to induce primary macrophages to possess repairing activities, which may provide insights for developing cell-based therapeutics for nonhealing wounds clinically.
Collapse
Affiliation(s)
- Rong Yong
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| | - Ruoyu Mu
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| | - Congwei Han
- School of Life Sciences & State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, No. 163 Xianlin Avenue, 210023, Nanjing, China
| | - Tzuwei Chao
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| | - Yu Liu
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| | - Lei Dong
- School of Life Sciences & State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, No. 163 Xianlin Avenue, 210023, Nanjing, China
- Chemistry and Biomedicine Innovative Center, Nanjing University, No. 163 Xianlin Avenue, 210023, Nanjing, Jiangsu, China
| | - Chunming Wang
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| |
Collapse
|
2
|
Mani AM, Lamin V, Peach RC, Friesen EH, Wong T, Singh MV, Dokun AO. miRNA-6236 Regulation of Postischemic Skeletal Muscle Angiogenesis. J Am Heart Assoc 2024:e035923. [PMID: 39604034 DOI: 10.1161/jaha.124.035923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Peripheral arterial disease affects >200 million people worldwide and is characterized by impaired blood flow to the lower extremities. There are no effective medical treatments available. Using the mouse hind-limb ischemia model and miRNA sequencing, we identified a novel miRNA, miR-6236, whose expression significantly elevated in ischemic mouse limbs compared with nonischemic limbs. The role of miR-6236 in general or in postischemic angiogenesis is not known. Here we describe its role using in vivo and in vitro models of peripheral arterial disease. METHODS AND RESULTS In primary mouse and human endothelial cells, we studied the effect of simulated ischemia on miR-6236 expression and assessed its role in cell viability, apoptosis, migration, and tube formation during ischemia. Furthermore, we developed miR-6236 null mice and tested its role in postischemic perfusion recovery using the hind-limb ischemia model. Lastly, using bioinformatics and gene expression analysis, we identified putative angiogenic miR-6236 targets. In vitro simulated ischemia-enhanced miR-6236 expression in mouse and human endothelial cells, whereas its inhibition improved viability, migration, tube formation, and reduced apoptosis. In vivo ischemic mouse skeletal muscle tissue showed higher miR-6236 expression compared with nonischemic muscles. Loss of miR-6236 improved impaired postischemic perfusion recovery and poor angiogenesis associated with streptozotocin-induced diabetes in mice. Six of the 8 miR-6236 predicted angiogenic target mRNAs showed expression consistent with regulation by miR-6236 in ischemic skeletal muscle. CONCLUSIONS Our results show for the first time that miR-6236 plays a key role in regulating postischemic perfusion recovery and angiogenesis.
Collapse
Affiliation(s)
- Arul M Mani
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Victor Lamin
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Ronan C Peach
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Eli H Friesen
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Thomas Wong
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Madhu V Singh
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Ayotunde O Dokun
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
- Fraternal Order of Eagles Diabetes Research Centre, Carver College of Medicine University of Iowa Iowa City IA USA
| |
Collapse
|
3
|
Gao M, Dong C, Chen Z, Jiang R, Shaw P, Gao W, Sun Y. Different impact of short-term and long-term hindlimb disuse on bone homeostasis. Gene 2024; 918:148457. [PMID: 38641071 DOI: 10.1016/j.gene.2024.148457] [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: 11/14/2023] [Revised: 02/19/2024] [Accepted: 04/08/2024] [Indexed: 04/21/2024]
Abstract
Disuse osteoporosis is one of the major problems of bone health which commonly occurs in astronauts during long-term spaceflight and bedridden patients. However, the mechanisms underlying such mechanical unloading induced bone loss have not been fully understood. In this study, we employed hindlimb-unloading mice models with different length of tail suspension to investigate if the bone loss was regulated by distinct factors under different duration of disuse. Our micro-CT results showed more significant decrease of bone mass in 6W (6-week) tail-suspension mice compared to the 1W (1-week) tail-suspension ones, as indicated by greater reduction of BV/TV, Tb.N, B.Ar/T.Ar and Ct.Th. RNA-sequencing results showed significant effects of hindlimb disuse on cell locomotion and immune system process which could cause bone loss.Real-time quantitative PCR results indicated a greater number of bone formation related genes that were downregulated in short-term tail-suspension mice compared to the long-term ones. It is, thus, suggested while sustained hindlimb unloading continuously contributes to bone loss, molecular regulation of bone homeostasis tends to reach a balance during this process.
Collapse
Affiliation(s)
- Minhao Gao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chengji Dong
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhuliu Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Renhao Jiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Peter Shaw
- Oujiang Lab, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang 325000, China
| | - Weiyang Gao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Yuanna Sun
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
4
|
Payne S, Neal A, De Val S. Transcription factors regulating vasculogenesis and angiogenesis. Dev Dyn 2024; 253:28-58. [PMID: 36795082 PMCID: PMC10952167 DOI: 10.1002/dvdy.575] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Transcription factors (TFs) play a crucial role in regulating the dynamic and precise patterns of gene expression required for the initial specification of endothelial cells (ECs), and during endothelial growth and differentiation. While sharing many core features, ECs can be highly heterogeneous. Differential gene expression between ECs is essential to pattern the hierarchical vascular network into arteries, veins and capillaries, to drive angiogenic growth of new vessels, and to direct specialization in response to local signals. Unlike many other cell types, ECs have no single master regulator, instead relying on differing combinations of a necessarily limited repertoire of TFs to achieve tight spatial and temporal activation and repression of gene expression. Here, we will discuss the cohort of TFs known to be involved in directing gene expression during different stages of mammalian vasculogenesis and angiogenesis, with a primary focus on development.
Collapse
Affiliation(s)
- Sophie Payne
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| | - Alice Neal
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| | - Sarah De Val
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| |
Collapse
|
5
|
Maharati A, Moghbeli M. Forkhead box proteins as the critical regulators of cisplatin response in tumor cells. Eur J Pharmacol 2023; 956:175937. [PMID: 37541368 DOI: 10.1016/j.ejphar.2023.175937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/11/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023]
Abstract
Cisplatin (CDDP) is one of the most common chemotherapy drugs used in a wide range of cancer patients; however, there is a high rate of CDDP resistance among cancer patients. Considering the side effects of cisplatin in normal tissues, it is necessary to predict the CDDP response in cancer patients. Therefore, identifying the molecular mechanisms involved in CDDP resistance can help to introduce the prognostic markers. Several molecular mechanisms such as apoptosis inhibition, drug efflux, drug detoxification, and increased DNA repair are involved in CDDP resistance. Regarding the key role of transcription factors in regulation of many cellular processes related to drug resistance, in the present review, we discussed the role of Forkhead box (FOX) protein family in CDDP response. It has been reported that FOX proteins mainly promote CDDP resistance through the regulation of DNA repair, autophagy, epithelial-mesenchymal transition (EMT), and signaling pathways. Therefore, FOX proteins can be introduced as the prognostic markers to predict CDDP response in cancer patients. In addition, considering that oncogenic role of FOX proteins, the CDDP treatment along with FOX inhibition can be used as a therapeutic strategy in cancer patients.
Collapse
Affiliation(s)
- Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Saberi A, Kouhjani M, Mohammadi M, Hosta-Rigau L. Novel scaffold platforms for simultaneous induction osteogenesis and angiogenesis in bone tissue engineering: a cutting-edge approach. J Nanobiotechnology 2023; 21:351. [PMID: 37770928 PMCID: PMC10536787 DOI: 10.1186/s12951-023-02115-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/15/2023] [Indexed: 09/30/2023] Open
Abstract
Despite the recent advances in the development of bone graft substitutes, treatment of critical size bone defects continues to be a significant challenge, especially in the elderly population. A current approach to overcome this challenge involves the creation of bone-mimicking scaffolds that can simultaneously promote osteogenesis and angiogenesis. In this context, incorporating multiple bioactive agents like growth factors, genes, and small molecules into these scaffolds has emerged as a promising strategy. To incorporate such agents, researchers have developed scaffolds incorporating nanoparticles, including nanoparticulate carriers, inorganic nanoparticles, and exosomes. Current paper provides a summary of the latest advancements in using various bioactive agents, drugs, and cells to synergistically promote osteogenesis and angiogenesis in bone-mimetic scaffolds. It also discusses scaffold design properties aimed at maximizing the synergistic effects of osteogenesis and angiogenesis, various innovative fabrication strategies, and ongoing clinical studies.
Collapse
Affiliation(s)
- Arezoo Saberi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Kouhjani
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Mohammadi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Leticia Hosta-Rigau
- DTU Health Tech, Centre for Nanomedicine and Theranostics, Technical University of Denmark, Produktionstorvet, Building 423, 2800, Kgs. Lyngby, Denmark.
| |
Collapse
|
7
|
Kopper TJ, Yu X, Graner MW. Immunopathology of Extracellular Vesicles in Macrophage and Glioma Cross-Talk. J Clin Med 2023; 12:3430. [PMID: 37240536 PMCID: PMC10219523 DOI: 10.3390/jcm12103430] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/25/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Glioblastomas (GBM) are a devastating disease with extremely poor clinical outcomes. Resident (microglia) and infiltrating macrophages are a substantial component of the tumor environment. In GBM and other cancers, tumor-derived extracellular vesicles (EVs) suppress macrophage inflammatory responses, impairing their ability to identify and phagocytose cancerous tissues. Furthermore, these macrophages then begin to produce EVs that support tumor growth and migration. This cross-talk between macrophages/microglia and gliomas is a significant contributor to GBM pathophysiology. Here, we review the mechanisms through which GBM-derived EVs impair macrophage function, how subsequent macrophage-derived EVs support tumor growth, and the current therapeutic approaches to target GBM/macrophage EV crosstalk.
Collapse
Affiliation(s)
| | | | - Michael W. Graner
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, 12700 E 19th Ave., Aurora, CO 80045, USA; (T.J.K.); (X.Y.)
| |
Collapse
|
8
|
Costa D, Andreucci M, Ielapi N, Serraino GF, Mastroroberto P, Bracale UM, Serra R. Molecular Determinants of Chronic Venous Disease: A Comprehensive Review. Int J Mol Sci 2023; 24:ijms24031928. [PMID: 36768250 PMCID: PMC9916309 DOI: 10.3390/ijms24031928] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Chronic Venous Disease (CVD) refers to several pathological and hemodynamic alterations of the veins of lower limbs causing a wide range of symptoms and signs with a high prevalence in the general population and with disabling consequences in the most severe forms. The etiology and pathophysiology of CVD is complex and multifactorial, involving genetic, proteomic, and cellular mechanisms that result in changes to the venous structure and functions. Expressions of several genes associated with angiogenesis, vascular development, and the regulation of veins are responsible for the susceptibility to CVD. Current evidence shows that several extracellular matrix alterations (ECM) could be identified and in some cases pharmacologically targeted. This review shows the most up to date information on molecular determinants of CVD in order to provide a complete overview of the current knowledge on this topic. In particular, the article explores the genetic influence, the hormonal influence, ECM imbalance, and histopathology of CVD and the role of endothelial dysfunction in CVD.
Collapse
Affiliation(s)
- Davide Costa
- Department of Law, Economics and Sociology, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Michele Andreucci
- Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Nicola Ielapi
- Department of Public Health and Infectious Disease, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Giuseppe Filiberto Serraino
- Department of Experimental and Clinical Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Pasquale Mastroroberto
- Department of Experimental and Clinical Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | | | - Raffaele Serra
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
- Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
- Correspondence:
| |
Collapse
|
9
|
Jiang Y, Zhao J, Xu J, Zhang H, Zhou J, Li H, Zhang G, Xu K, Jing Z. Glioblastoma-associated microglia-derived exosomal circKIF18A promotes angiogenesis by targeting FOXC2. Oncogene 2022; 41:3461-3473. [DOI: 10.1038/s41388-022-02360-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 05/14/2022] [Accepted: 05/23/2022] [Indexed: 11/09/2022]
|
10
|
Xia S, Yu W, Menden H, Younger ST, Sampath V. FOXC2 Autoregulates Its Expression in the Pulmonary Endothelium After Endotoxin Stimulation in a Histone Acetylation-Dependent Manner. Front Cell Dev Biol 2021; 9:657662. [PMID: 34017833 PMCID: PMC8129010 DOI: 10.3389/fcell.2021.657662] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 04/12/2021] [Indexed: 12/26/2022] Open
Abstract
The innate immune response of pulmonary endothelial cells (EC) to lipopolysaccharide (LPS) induces Forkhead box protein C2 (FOXC2) activation through Toll Like Receptor 4 (TLR4). The mechanisms by which FOXC2 expression is regulated in lung EC under LPS stimulation remain unclear. We postulated that FOXC2 regulates its own expression in sepsis, and its transcriptional autoregulation directs lymphatic EC cell-fate decision. Bioinformatic analysis identified potential FOXC2 binding sites in the FOXC2 promoter. In human lung EC, we verified using chromatin immunoprecipitation (ChIP) and luciferase assays that FOXC2 bound to its own promoter and stimulated its expression after LPS stimulation. Chemical inhibition of histone acetylation by garcinol repressed LPS-induced histone acetylation in the FOXC2 promoter region, and disrupted LPS-mediated FOXC2 binding and transcriptional activation. CRISPR/dCas9/gRNA directed against FOXC2-binding-element (FBE) suppressed LPS-stimulated FOXC2 binding and autoregulation by blocking FBEs in the FOXC2 promoter, and repressed expression of lymphatic EC markers. In a neonatal mouse model of sterile sepsis, LPS-induced FOXC2 binding to FBE and FOXC2 expression in lung EC was attenuated with garcinol treatment. These data reveal a new mechanism of LPS-induced histone acetylation-dependent FOXC2 autoregulation.
Collapse
Affiliation(s)
- Sheng Xia
- Department of Pediatrics, Children's Mercy Kansas City, MO, United States
| | - Wei Yu
- Department of Pediatrics, Children's Mercy Kansas City, MO, United States
| | - Heather Menden
- Department of Pediatrics, Children's Mercy Kansas City, MO, United States
| | - Scott T Younger
- Center for Pediatric Genomic Medicine, Children's Mercy Kansas City, MO, United States
| | - Venkatesh Sampath
- Department of Pediatrics, Children's Mercy Kansas City, MO, United States
| |
Collapse
|
11
|
Serra R, Ssempijja L, Provenzano M, Andreucci M. Genetic biomarkers in chronic venous disease. Biomark Med 2020; 14:75-80. [PMID: 32053001 DOI: 10.2217/bmm-2019-0408] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Raffaele Serra
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology at the Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Calabria, Italy.,Department of Medical & Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Calabria, Italy
| | - Lwanga Ssempijja
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology at the Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Calabria, Italy.,Department of Medical & Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Calabria, Italy
| | - Michele Provenzano
- Department of Health Sciences, University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Calabria, Italy
| | - Michele Andreucci
- Department of Health Sciences, University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Calabria, Italy
| |
Collapse
|
12
|
Rajabi A, Saber A, Pourmahdi M, Emami A, Ravanbakhsh R, Khodavirdipour A, Khodaei M, Akbarzadeh M, Abdolahi S, Hosseinpourfeizi MA, Safaralizadeh R. Anti-Cancer Effect of Melatonin via Downregulation of Delta-like Ligand 4 in Estrogen-Responsive Breast Cancer Cells. Recent Pat Anticancer Drug Discov 2020; 15:329-340. [PMID: 32990541 DOI: 10.2174/1574892815666200929145236] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND The Notch signaling pathway has a key role in angiogenesis and Delta - Like Ligand 4 (DLL4) is one of the main ligands of Notch involved in cell proliferation in sprouting vessels. OBJECTIVE In this study, we aimed to evaluate the expression of DLL4 in primary breast tumors and to examine the effect of melatonin on DLL4 expression in vitro. METHODS Eighty-five breast tumor and paired adjacent non-tumor tissue samples were collected. Apoptosis assay was performed on breast cancer cells to evaluate melatonin effects. Western blot and quantitative RT-PCR were used to measure DLL4 expression. Then, we investigated the effect of melatonin on the expression of DLL4 in four breast cancer cell lines at RNA and protein levels. We also performed a probabilistic neural network analysis to study genes closely associated with DLL4 expression. RESULTS Our results showed a significantly higher expression of DLL4 in tumor tissues compared to non-tumor tissues (P = 0.027). Melatonin treatment substantially attenuated DLL4 expression in BT474 and MCF-7 cells, but not in SK-BR-3 and MDA-MB-231 cells. Also, melatonin induced apoptosis in all four cell lines. Network analysis revealed a set of 15 genes that had close association and interaction with DLL4. DLL4 was overexpressed in breast cancer tissues as compared to the non-tumor tissues. CONCLUSION It can be concluded that melatonin treatment attenuated DLL4 expression only in estrogen- responsive breast cancer cells and is able to induce apoptosis in breast cancer cells.
Collapse
Affiliation(s)
- Ali Rajabi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Ali Saber
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Mahsa Pourmahdi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Ali Emami
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Reyhaneh Ravanbakhsh
- Department of Aquatic Biotechnology, Artemia and Aquaculture Research Institute, Urmia University, Urmia, Iran
| | - Amir Khodavirdipour
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mehran Khodaei
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Molood Akbarzadeh
- Department of Biology, Faculty of Sciences, Azerbaijan Shahid Madani University, Tabriz, Iran
| | - Sepehr Abdolahi
- Department of Biology, Faculty of Sciences, Azerbaijan Shahid Madani University, Tabriz, Iran
| | | | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
13
|
Dong X, Yin J, Yun B, Lü B, Yin G. [Research on influence mechanism of G protein coupled receptor kinase interacting protein 1 on differentiation of bone marrow mesenchymal stem cells into endothelial cells]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 32:257-263. [PMID: 29806272 DOI: 10.7507/1002-1892.201709090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To investigate the mechanism of G protein coupled receptor kinase interacting protein 1 (GIT1) affecting angiogenesis by comparing the differentiation of bone marrow mesenchymal stem cells (BMSCs) differentiated into endothelial cells between GIT1 wild type mice and GIT1 gene knockout mice. Methods Male and female GIT1 heterozygous mice were paired breeding, and the genotypic identification of newborn mice were detected by PCR. The 2nd generation BMSCs isolated from GIT1 wild type mice or GIT1 gene knockout mice were divided into 4 groups, including wild type control group (group A), wild type experimental group (group A1), GIT1 knockout control group (group B), and GIT1 knockout experimental group (group B1). The cells of groups A1 and B1 were cultured with the endothelial induction medium and the cells of groups A and B with normal cluture medium. The expressions of vascular endothelial growth factor receptor 2 (VEGFR-2), VEGFR-3, and phospho-VEGFR-2 (pVEGFR-2), and pVEGFR-3 proteins were detected by Western blot. The endothelial cell markers [von Willebrand factor (vWF), platelet-endothelial cell adhesion molecule 1 (PECAM-1), and vascular endothelial cadherin (VE-Cadherin)] were detected by flow cytometry. The 2nd generation BMSCs of GIT1 wild type mice were divided into 4 groups according to the different culture media: group Ⅰ, primary cell culture medium; group Ⅱ, cell culture medium containing SAR131675 (VEGFR-3 blocker); group Ⅲ, endothelial induction medium; group Ⅳ, endothelial induction medium containing SAR131675. The endothelial cell markers (vWF, PECAM-1, and VE-Cadherin) in 4 groups were also detected by flow cytometry. Results Western blot results showed that there was no obviously difference in protein expressions of VEGFR-2 and pVEGFR-2 between groups; and the expressions of VEGFR-3 and pVEGFR-3 proteins in group A1 were obviously higher than those in groups A, B, and B1. The flow cytometry results showed that the expressions of vWF, PECAM-1, and VE-Cadherin were significantly higher in group A1 than in groups A, B, and B1 ( P<0.05), and in group B1 than in groups A and B ( P<0.05); but no significant difference was found between groups A and B ( P>0.05). In the VEGFR-3 blocked experiment, the flow cytometry results showed that the expressions of vWF, PECAM-1, and VE-Cadherin were significantly higher in group Ⅲ than in groupsⅠ, Ⅱ, and Ⅳ, and in group Ⅳ than in groups Ⅰ and Ⅱ ( P<0.05); but no significant difference was found between groups Ⅰ and Ⅱ ( P>0.05). Conclusion GIT1 mediates BMSCs of mice differentiation into endothelial cells via VEGFR-3, thereby affecting the angiogenesis.
Collapse
Affiliation(s)
- Xiancheng Dong
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing Jiangsu, 210029, P.R.China
| | - Jian Yin
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing Jiangsu, 210029, P.R.China
| | - Bo Yun
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing Jiangsu, 210029, P.R.China
| | - Bin Lü
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing Jiangsu, 210029, P.R.China
| | - Guoyong Yin
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing Jiangsu, 210029,
| |
Collapse
|
14
|
Wang T, Zheng L, Wang Q, Hu YW. Emerging roles and mechanisms of FOXC2 in cancer. Clin Chim Acta 2018; 479:84-93. [PMID: 29341903 DOI: 10.1016/j.cca.2018.01.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 12/20/2022]
Abstract
Forkhead box protein C2 (FOXC2), a transcription factor of the forkhead/winged-helix family, is required for embryonic and prenatal development. FOXC2 acts as a crucial modulator during both angiogenesis and lymphangiogenesis via multiple angiogenic and lymphangiogenic pathways, respectively. Although recent studies have shed light on the emerging role of FOXC2 in cancer, very little is known about the precise underlying mechanisms. The purpose of this review is to summarize the current understanding of FOXC2 and provide potential mechanistic explanations of the relationship between FOXC2 and cancer, as well as discuss the prospect for future research in the promising prognostic value of FOXC2 in cancer.
Collapse
Affiliation(s)
- Teng Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Lei Zheng
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Qian Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
15
|
Rane CK, Patel M, Cai L, Senapedis W, Baloglu E, Minden A. Decrypting the PAK4 transcriptome profile in mammary tumor forming cells using Next Generation Sequencing. Genomics 2017; 110:S0888-7543(17)30128-3. [PMID: 29055713 DOI: 10.1016/j.ygeno.2017.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 10/03/2017] [Accepted: 10/17/2017] [Indexed: 01/05/2023]
Abstract
The p-21 Activated Kinase 4 (PAK4) protein kinase is implicated in many cancers, including breast cancer. Overexpression of PAK4 is sufficient to cause mouse mammary epithelial cells (iMMECs) to become tumorigenic. To gain insight into the long-term gene expression changes that occur downstream to PAK4, we performed Next Generation Sequencing of RNA collected from PAK4 overexpressing iMMECs and wild-type iMMECs. We identified a list of genes whose expression levels were altered in response to PAK4 overexpression in iMMECs. Some of these genes, including FoxC2 and ParvB, are consistent with a role for PAK4 in cancer. In addition, PAK4 regulates many genes that are frequently associated with the inflammatory response, raising the possibility that there is a connection between PAK4, inflammation, and the tumor microenvironment. This study delineates the PAK4 transcriptome profile in transformed mammary cells and can provide translational utility in other types of cancers as well.
Collapse
Affiliation(s)
- Chetan K Rane
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 164 Frelinghuysen Road, Piscataway, NJ 08854, United States
| | - Misaal Patel
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ 08854, United States
| | - Li Cai
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ 08854, United States
| | - William Senapedis
- Karyopharm Therapeutics, Inc., 85 Wells Avenue, Newton, MA 02459, United States
| | - Erkan Baloglu
- Karyopharm Therapeutics, Inc., 85 Wells Avenue, Newton, MA 02459, United States
| | - Audrey Minden
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 164 Frelinghuysen Road, Piscataway, NJ 08854, United States.
| |
Collapse
|
16
|
Zhang W, Duan N, Song T, Li Z, Zhang C, Chen X. The Emerging Roles of Forkhead Box (FOX) Proteins in Osteosarcoma. J Cancer 2017; 8:1619-1628. [PMID: 28775781 PMCID: PMC5535717 DOI: 10.7150/jca.18778] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/27/2017] [Indexed: 12/22/2022] Open
Abstract
Osteosarcoma is the most common bone cancer primarily occurring in children and young adults. Over the past few years, the deregulation of a superfamily transcription factors, known as forkhead box (FOX) proteins, has been demonstrated to contribute to the pathogenesis of osteosarcoma. Molecular mechanism studies have demonstrated that FOX family proteins participate in a variety of signaling pathways and that their expression can be regulated by multiple factors. The dysfunction of FOX genes can alter osteosarcoma cell differentiation, metastasis and progression. In this review, we summarized the evidence that FOX genes play direct or indirect roles in the development and progression of osteosarcoma, and evaluated the emerging role of FOX proteins as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Wentao Zhang
- Department of Orthopaedics, Xi'an Hong-Hui Hospital affiliated to medical college of Xi'an Jiaotong University, Xi'an, Shaanxi, China, 710054
| | - Ning Duan
- Department of Orthopaedics, Xi'an Hong-Hui Hospital affiliated to medical college of Xi'an Jiaotong University, Xi'an, Shaanxi, China, 710054
| | - Tao Song
- Department of Orthopaedics, Xi'an Hong-Hui Hospital affiliated to medical college of Xi'an Jiaotong University, Xi'an, Shaanxi, China, 710054
| | - Zhong Li
- Department of Orthopaedics, Xi'an Hong-Hui Hospital affiliated to medical college of Xi'an Jiaotong University, Xi'an, Shaanxi, China, 710054
| | - Caiguo Zhang
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Xun Chen
- Department of Orthopaedics, Xi'an Hong-Hui Hospital affiliated to medical college of Xi'an Jiaotong University, Xi'an, Shaanxi, China, 710054
| |
Collapse
|
17
|
Shadrina AS, Smetanina MA, Sokolova EA, Sevost'ianova KS, Shevela AI, Demekhova MY, Shonov OA, Ilyukhin EA, Voronina EN, Zolotukhin IA, Kirienko AI, Filipenko ML. Association of polymorphisms near the FOXC2 gene with the risk of varicose veins in ethnic Russians. Phlebology 2015; 31:640-8. [PMID: 26420053 DOI: 10.1177/0268355515607404] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To investigate the association of polymorphisms located near the FOXC2 gene with the risk of varicose veins in ethnic Russians. METHODS Allele, genotype, and haplotype frequencies were determined in the sample of 474 patients with primary varicose veins and in the control group of 478 individuals without a history of chronic venous disease. RESULTS Polymorphisms rs7189489, rs4633732, and rs1035550 showed the association with the increased risk of varicose veins, but none of the observed associations remained significant after correction for multiple testing. Haplotype analysis revealed the association of haplotype rs7189489 C-rs4633732 T-rs34221221 C-rs1035550 C-rs34152738 T-rs12711457 G with the increased risk of varicose veins (OR = 2.67, P = 0.01). CONCLUSIONS Our results provide evidence that the studied polymorphisms do not play a major role in susceptibility to varicose veins development in the Russian population.
Collapse
Affiliation(s)
- Alexandra S Shadrina
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| | - Mariya A Smetanina
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
| | - Ekaterina A Sokolova
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| | | | - Andrey I Shevela
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
| | | | - Oleg A Shonov
- Private Surgery Center "Medalp", Saint Petersburg, Russia
| | | | - Elena N Voronina
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| | - Igor A Zolotukhin
- Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - Maxim L Filipenko
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia Kazan Federal University, Kazan, Republic of Tatarstan, Russia
| |
Collapse
|
18
|
McManus MM, Weiss KR, Hughes DPM. Understanding the role of Notch in osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 804:67-92. [PMID: 24924169 DOI: 10.1007/978-3-319-04843-7_4] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The Notch pathway has been described as an oncogene in osteosarcoma, but the myriad functions of all the members of this complex signaling pathway, both in malignant cells and nonmalignant components of tumors, make it more difficult to define Notch as simply an oncogene or a tumor suppressor. The cell-autonomous behaviors caused by Notch pathway manipulation may vary between cell lines but can include changes in proliferation, migration, invasiveness, oxidative stress resistance, and expression of markers associated with stemness or tumor-initiating cells. Beyond these roles, Notch signaling also plays a vital role in regulating tumor angiogenesis and vasculogenesis, which are vital aspects of osteosarcoma growth and behavior in vivo. Further, osteosarcoma cells themselves express relatively low levels of Notch ligand, making it likely that nonmalignant cells, especially endothelial cells and pericytes, are the major source of Notch activation in osteosarcoma tumors in vivo and in patients. As a result, Notch pathway expression is not expected to be uniform across a tumor but likely to be highest in those areas immediately adjacent to blood vessels. Therapeutic targeting of the Notch pathway is likewise expected to be complicated. Most pharmacologic approaches thus far have focused on inhibition of gamma secretase, a protease of the presenilin complex. This enzyme, however, has numerous other target proteins that would be expected to affect osteosarcoma behavior, including CD44, the WNT/β-catenin pathway, and Her-4. In addition, Notch plays a vital role in tissue and organ homeostasis in numerous systems, and toxicities, especially GI intolerance, have limited the effectiveness of gamma secretase inhibitors. New approaches are in development, and the downstream targets of Notch pathway signaling also may turn out to be good targets for therapy. In summary, a full understanding of the complex functions of Notch in osteosarcoma is only now unfolding, and this deeper knowledge will help position the field to better utilize novel therapies as they are developed.
Collapse
Affiliation(s)
- Madonna M McManus
- The Children's Cancer Hospital at MD Anderson Cancer Center, Houston, TX, USA
| | | | | |
Collapse
|
19
|
Faralli JA, Gagen D, Filla MS, Crotti TN, Peters DM. Dexamethasone increases αvβ3 integrin expression and affinity through a calcineurin/NFAT pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:3306-3313. [PMID: 24100160 DOI: 10.1016/j.bbamcr.2013.09.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 09/05/2013] [Accepted: 09/26/2013] [Indexed: 12/13/2022]
Abstract
The purpose of this study was to determine how dexamethasone (DEX) regulates the expression and activity of αvβ3 integrin. FACS analysis showed that DEX treatment induced expression of an activated αvβ3 integrin. Its expression remained high as long as DEX was present and continued following DEX removal. FACS analysis showed that the upregulation of αvβ3 integrin was the result of an increase in the expression of the β3 integrin subunit. By real time qPCR, DEX treatment induced a 6.2-fold increase (p<0.04) in β3 integrin mRNA by day 2 compared to control and remained elevated for 6days of treatment and then an additional 10days once the DEX was removed. The increase in β3 integrin mRNA levels required only 1day of DEX treatment to increase levels for 4days in the absence of DEX. In contrast, DEX did not alter β1 integrin mRNA or protein levels. The DEX-induced upregulation of β3 integrin mRNA was partly due to an increase in its half-life to 60.7h from 22.5h in control cultures (p<0.05) and could be inhibited by RU486 and cycloheximide, suggesting that DEX-induced de novo protein synthesis of an activation factor was needed. The calcineurin inhibitors cyclosporin A (CsA) and FK506 inhibited the DEX induced increase in β3 integrin mRNA. In summary, the DEX-induced increase in β3 integrin is a secondary glucocorticoid response that results in prolonged expression of αvβ3 integrin and the upregulation of the β3 integrin subunit through the calcineurin/NFAT pathway.
Collapse
Affiliation(s)
- Jennifer A Faralli
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI 53706, USA
| | - Debjani Gagen
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI 53706, USA
| | - Mark S Filla
- Department of Ophthalmology & Visual Sciences, University of Wisconsin, Madison, WI 53706, USA
| | - Tania N Crotti
- Discipline of Anatomy and Pathology, The University of Adelaide, South Australia, Australia
| | - Donna M Peters
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI 53706, USA; Department of Ophthalmology & Visual Sciences, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
20
|
Zhu JL, Song YX, Wang ZN, Gao P, Wang MX, Dong YL, Xing CZ, Xu HM. The clinical significance of mesenchyme forkhead 1 (FoxC2) in gastric carcinoma. Histopathology 2013; 62:1038-48. [PMID: 23614500 DOI: 10.1111/his.12132] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2013] [Indexed: 01/20/2023]
Affiliation(s)
- Jin-Liang Zhu
- Department of Surgical Oncology and General Surgery; First Hospital of China Medical University; Shenyang; China
| | - Yong-Xi Song
- Department of Surgical Oncology and General Surgery; First Hospital of China Medical University; Shenyang; China
| | - Zhen-Ning Wang
- Department of Surgical Oncology and General Surgery; First Hospital of China Medical University; Shenyang; China
| | - Peng Gao
- Department of Surgical Oncology and General Surgery; First Hospital of China Medical University; Shenyang; China
| | - Mei-Xian Wang
- Department of Tumor Pathology and Surgical Oncology; First Hospital of China Medical University; Shenyang; China
| | - Yu-Lan Dong
- Department of Tumor Pathology and Surgical Oncology; First Hospital of China Medical University; Shenyang; China
| | - Cheng-Zhong Xing
- Department of Surgical Oncology and General Surgery; First Hospital of China Medical University; Shenyang; China
| | - Hui-Mian Xu
- Department of Surgical Oncology and General Surgery; First Hospital of China Medical University; Shenyang; China
| |
Collapse
|
21
|
Interactions between VEGFR and Notch signaling pathways in endothelial and neural cells. Cell Mol Life Sci 2013; 70:1779-92. [PMID: 23479133 DOI: 10.1007/s00018-013-1312-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 02/25/2013] [Accepted: 02/25/2013] [Indexed: 12/25/2022]
Abstract
Notch cell interaction mechanism governs cell fate decisions in many different cell contexts throughout the lifetime of all Metazoan species. It links the fate of one cell to that of its neighbors through cell-to-cell contacts, and binding of Notch receptors expressed on one cell to their membrane bound ligands on an adjacent cell. Environmental cues, such as growth factors and extracellular matrix molecules, superimpose a dynamic regulation on this canonical Notch signaling pathway. In this review, we will focus on Notch signaling in the vertebrate vascular and nervous systems and examine its role in angiogenesis, neurogenesis, and neurovascular interactions. We will also highlight the molecular relationships of the Notch pathway with vascular endothelial growth factors (VEGFs) and their high-affinity tyrosine kinase VEGF receptors, key regulators of both angiogenesis and neurogenesis.
Collapse
|
22
|
Tumor lymphangiogenesis as a potential therapeutic target. JOURNAL OF ONCOLOGY 2012; 2012:204946. [PMID: 22481918 PMCID: PMC3307004 DOI: 10.1155/2012/204946] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 10/10/2011] [Accepted: 10/31/2011] [Indexed: 12/18/2022]
Abstract
Metastasis the spread of cancer cells to distant organs, is the main cause of death for cancer patients. Metastasis is often mediated by lymphatic vessels that invade the primary tumor, and an early sign of metastasis is the presence of cancer cells in the regional lymph node (the first lymph node colonized by metastasizing cancer cells from a primary tumor). Understanding the interplay between tumorigenesis and lymphangiogenesis (the formation of lymphatic vessels associated with tumor growth) will provide us with new insights into mechanisms that modulate metastatic spread. In the long term, these insights will help to define new molecular targets that could be used to block lymphatic vessel-mediated metastasis and increase patient survival. Here, we review the molecular mechanisms of embryonic lymphangiogenesis and those that are recapitulated in tumor lymphangiogenesis, with a view to identifying potential targets for therapies designed to suppress tumor lymphangiogenesis and hence metastasis.
Collapse
|
23
|
Kume T. The Role of FoxC2 Transcription Factor in Tumor Angiogenesis. JOURNAL OF ONCOLOGY 2011; 2012:204593. [PMID: 22174714 PMCID: PMC3228356 DOI: 10.1155/2012/204593] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 08/17/2011] [Accepted: 08/29/2011] [Indexed: 12/27/2022]
Abstract
Much has been learned about the mechanisms underlying tumor angiogenesis, and therapies that target vascular endothelial growth factor (VEGF) to limit tumor angiogenesis and subsequent disease progression have recently been approved. However, the transcriptional mechanisms that regulate pathological angiogenesis remain largely unknown. FoxC2, a member of the Forkhead box (Fox) transcription factor family, is critical for vascular formation during development, and recent studies have shown that FoxC2 is expressed in the endothelium of tumors in both humans and mice. In a B16 mouse melanoma model, Foxc2 deficiency reduced tumor growth and neovascularization and was associated with impairments in mural-cell coverage and increases in endothelial-cell apoptosis in tumor blood vessels. FoxC2 is also expressed by tumor cells in human breast, colonic, and esophageal cancer and participates in the epithelial-mesenchymal transition (EMT), a key process that leads to the invasion and metastasis of aggressive tumors. Collectively, these observations suggest that FoxC2 is essential for tumor angiogenesis and disease progression and that FoxC2 may be a viable target for cancer therapy.
Collapse
Affiliation(s)
- Tsutomu Kume
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, 303E Chicago Avenue, Chicago, IL 60611, USA
| |
Collapse
|