1
|
Wu H, Li ZX, Fang K, Zhao ZY, Sun MC, Feng AQ, Leng ZY, Zhang ZH, Chu Y, Zhang L, Chen T, Xu MD. IGF-1-mediated FOXC1 overexpression induces stem-like properties through upregulating CBX7 and IGF-1R in esophageal squamous cell carcinoma. Cell Death Discov 2024; 10:102. [PMID: 38413558 PMCID: PMC10899262 DOI: 10.1038/s41420-024-01864-0] [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: 05/07/2023] [Revised: 01/27/2024] [Accepted: 02/12/2024] [Indexed: 02/29/2024] Open
Abstract
Substantial evidence attests to the pivotal role of cancer stem cells (CSC) in both tumorigenesis and drug resistance. A member of the forkhead box (FOX) family, FOXC1, assumes significance in embryonic development and organogenesis. Furthermore, FOXC1 functions as an overexpressed transcription factor in various tumors, fostering proliferation, enhancing migratory capabilities, and promoting drug resistance, while maintaining stem-cell-like properties. Despite these implications, scant attention has been devoted to its role in esophageal squamous cell carcinoma. Our investigation revealed a pronounced upregulation of FOXC1 expression in ESCC, correlating with a poor prognosis. The downregulation of FOXC1 demonstrated inhibitory effects on ESCC tumorigenesis, proliferation, and tolerance to chemotherapeutic agents, concurrently reducing the levels of stemness-related markers CD133 and CD44. Further studies validated that FOXC1 induces ESCC stemness by transactivating CBX7 and IGF-1R. Additionally, IGF-1 activated the PI3K/AKT/NF-κB and MEK/ERK/NF-κB pathways through its binding to IGF-1R, thereby augmenting FOXC1 expression. Conversely, suppressing FOXC1 impeded ESCC stemness induced by IGF-1. The presence of a positive feedback loop, denoted by IGF-1-FOXC1-IGF-1R, suggests the potential of FOXC1 as a prognostic biomarker for ESCC. Taken together, targeting the IGF-1-FOXC1-IGF-1R axis emerges as a promising approach for anti-CSC therapy in ESCC.
Collapse
Affiliation(s)
- Hao Wu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China
| | - Zhao-Xing Li
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China
| | - Kang Fang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China
| | - Zi-Ying Zhao
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China
| | - Ming-Chuang Sun
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China
| | - An-Qi Feng
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China
| | - Zhu-Yun Leng
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China
| | - Ze-Hua Zhang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China
| | - Yuan Chu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China
| | - Li Zhang
- Department of Pathology, Shanghai East Hospital, School of Medicine, Tongji 8 University, 200120, Shanghai, China
| | - Tao Chen
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China.
| | - Mei-Dong Xu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China.
| |
Collapse
|
2
|
Qiu L, Ma L, Chen D, Zhang N, Cai J, Zhang Q, Wang X, Yi H, Yao H, Fan FY. Novel_circ_003686 regulates the osteogenic differentiation of MSCs in patients with myeloma bone disease through miR-142-5p/IGF1 axis. J Bone Oncol 2023; 43:100509. [PMID: 38021072 PMCID: PMC10654027 DOI: 10.1016/j.jbo.2023.100509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/13/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Objectives Circ_003686 is a novel_circRNA with abnormally low expression found in the samples of multiple myeloma bone disease (MBD) patients. The current research intended to investigate the effects of novel_circ_003686 in osteogenesis-induced differentiation of bone marrow mesenchymal stem cells (BMSCs) in MBD. Methods BMSCs were extracted from MBD patients and normal participants, the pcDNA3.1 encoding the circ_003686 (ov-circ_003686), miR-142-5p-mimic/inhibitor and siRNA oligonucleotides targeting insulin like growth factor 1 (IGF1, si-IGF1) were applied to intervene circ_003686, miR-142-5p and IGF1 levels, respectively. Results: Results showed that ov-circ_003686 could mediate the osteogenesis-induced differentiation of MBD-BMSC, and luciferase assay and RIP experiments confirmed that circ_003686 could bind to miR-142-5p. MiR-142-5p-inhibitor helped osteogenesis-induced differentiation, while miR-142-5p-mimic inhibited osteogenesis-induced differentiation and reversed the promoting effect of ov-circ_003686, suggesting that circ_003686/miR-142-5p axis participated in osteogenesis-induced differentiation of MBD-BMSC. In addition, miR-142-5p binds to the target gene IGF1 and negatively adjust its expression. Si-IGF1 significantly inhibited the osteogenesis-induced differentiation and reversed the promotion effects of miR-142-5p-inhibitor and ov-circ_003686. Moreover, circ_003686/miR-142-5p/IGF1 axis meaningfully regulates protein expressions in the PI3K/AKT pathway. Conclusion In conclusion, this research confirmed that circ_003686 regulated the osteogenesis-induced differentiation of MBD-BMSC by sponging miR-142-5p and mediating IGF1, and the PI3K/AKT pathway may also be involved.
Collapse
Affiliation(s)
| | | | | | - Nan Zhang
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People’s Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Jiao Cai
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People’s Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Qian Zhang
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People’s Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Xiao Wang
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People’s Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Hai Yi
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People’s Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Hao Yao
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People’s Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Fang-Yi Fan
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People’s Liberation Army Western Theatre, Chengdu, SiChuan, China
| |
Collapse
|
3
|
van Loon K, van Breest Smallenburg ME, Huijbers EJM, Griffioen AW, van Beijnum JR. Extracellular vimentin as a versatile immune suppressive protein in cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188985. [PMID: 37717859 DOI: 10.1016/j.bbcan.2023.188985] [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: 06/15/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023]
Abstract
The interest in finding new targets in the tumor microenvironment for anti-cancer therapy has increased rapidly over the years. More specifically, the tumor-associated blood vessels are a promising target. We recently found that the intermediate filament protein vimentin is externalized by endothelial cells of the tumor vasculature. Extracellular vimentin was shown to sustain angiogenesis by mimicking VEGF and supporting cell migration, as well as endothelial cell anergy, the unresponsiveness of the endothelium to proinflammatory cytokines. The latter hampers immune cell infiltration and subsequently provides escape from tumor immunity. Other studies showed that extracellular vimentin plays a role in sustained systemic and local inflammation. Here we will review the reported roles of extracellular vimentin with a particular emphasis on its involvement in the interactions between immune cells and the endothelium in the tumor microenvironment. To this end, we discuss the different ways by which extracellular vimentin modulates the immune system. Moreover, we review how this protein can alter immune cell-vessel wall adhesion by altering the expression of adhesion proteins, attenuating immune cell infiltration into the tumor parenchyma. Finally, we discuss how vimentin-targeting therapy can reverse endothelial cell anergy and promote immune infiltration, supporting anti-tumor immunity.
Collapse
Affiliation(s)
- Karlijn van Loon
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Mathilda E van Breest Smallenburg
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Elisabeth J M Huijbers
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; CimCure BV, Amsterdam, the Netherlands
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; CimCure BV, Amsterdam, the Netherlands
| | - Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; CimCure BV, Amsterdam, the Netherlands.
| |
Collapse
|
4
|
Tien TY, Wu YJ, Su CH, Hsieh CL, Wang BJ, Lee YN, Su Y, Yeh HI. Pannexin 1 Modulates Angiogenic Activities of Human Endothelial Colony-Forming Cells Through IGF-1 Mechanism and Is a Marker of Senescence. Arterioscler Thromb Vasc Biol 2023; 43:1935-1951. [PMID: 37589139 DOI: 10.1161/atvbaha.123.319529] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 08/01/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND We examined the role of Panxs (pannexins) in human endothelial progenitor cell (EPC) senescence. METHODS Young and replication-induced senescent endothelial colony-forming cells (ECFCs) derived from human circulating EPCs were used to examine cellular activities and senescence-associated indicators after transfection of short interference RNA specific to Panx1 or lentivirus-mediated Panx1 overexpression. Hind limb ischemia mice were used as in vivo angiogenesis model. Protein and phospho-kinase arrays were used to determine underlying mechanisms. RESULTS Panx1 was the predominant Panx isoform in human ECFCs and upregulated in both replication-induced senescent ECFCs and circulating EPCs from aged mice and humans. Cellular activities of the young ECFCs were enhanced by Panx1 downregulation but attenuated by its upregulation. In addition, reduction of Panx1 in the senescent ECFCs could rejuvenate cellular activities with reduced senescence-associated indicators, including senescence-associated β-galactosidase activity, p16INK4a (cyclin-dependent kinase inhibitor 2A), p21 (cyclin-dependent kinase inhibitor 1), acetyl-p53 (tumor protein P53), and phospho-histone H2A.X (histone family member X). In mouse ischemic hind limbs injected senescent ECFCs, blood perfusion ratio, salvaged limb outcome, and capillary density were all improved by Panx1 knockdown. IGF-1 (insulin-like growth factor 1) was significantly increased in the supernatant from senescent ECFCs after Panx1 knockdown. The enhanced activities and paracrine effects of Panx1 knockdown senescent ECFCs were completely inhibited by anti-IGF-1 antibodies. FAK (focal adhesion kinase), ERK (extracellular signal-regulated kinase), and STAT3 (signal transducer and activator of transcription 3) were activated in senescent ECFCs with Panx1 knockdown, in which the intracellular calcium level was reduced, and the activation was inhibited by supplemented calcium. The increased IGF-1 in Panx1-knockdown ECFCs was abrogated, respectively, by inhibitors of FAK (PF562271), ERK (U0126), and STAT3 (NSC74859) and supplemented calcium. CONCLUSIONS Panx1 expression is upregulated in human ECFCs/EPCs with replication-induced senescence and during aging. Angiogenic potential of senescent ECFCs is improved by Panx1 reduction through increased IGF-1 production via activation of the FAK-ERK axis following calcium influx reduction. Our findings provide new strategies to evaluate EPC activities and rejuvenate senescent EPCs for therapeutic angiogenesis.
Collapse
Affiliation(s)
- Ting-Yi Tien
- Institute of Biopharmaceutical Science, National Yang Ming Chiao Tung University, Taipei, Taiwan (T.-Y.T., Y.S.)
- Departments of Medical Research (T.-Y.T., C.-L.H., B.-J.W., Y.-N.L.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Yih-Jer Wu
- Internal Medicine (Y.-J.W., C.-H.S., H.-I.Y.), MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (Y.-J.W., C.-H.S., H.-I.Y.)
| | - Cheng-Huang Su
- Internal Medicine (Y.-J.W., C.-H.S., H.-I.Y.), MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (Y.-J.W., C.-H.S., H.-I.Y.)
| | - Chin-Ling Hsieh
- Departments of Medical Research (T.-Y.T., C.-L.H., B.-J.W., Y.-N.L.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Bo-Jeng Wang
- Departments of Medical Research (T.-Y.T., C.-L.H., B.-J.W., Y.-N.L.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Yi-Nan Lee
- Departments of Medical Research (T.-Y.T., C.-L.H., B.-J.W., Y.-N.L.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Yeu Su
- Institute of Biopharmaceutical Science, National Yang Ming Chiao Tung University, Taipei, Taiwan (T.-Y.T., Y.S.)
| | - Hung-I Yeh
- Internal Medicine (Y.-J.W., C.-H.S., H.-I.Y.), MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (Y.-J.W., C.-H.S., H.-I.Y.)
| |
Collapse
|
5
|
Dudley AC, Griffioen AW. Pathological angiogenesis: mechanisms and therapeutic strategies. Angiogenesis 2023; 26:313-347. [PMID: 37060495 PMCID: PMC10105163 DOI: 10.1007/s10456-023-09876-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/26/2023] [Indexed: 04/16/2023]
Abstract
In multicellular organisms, angiogenesis, the formation of new blood vessels from pre-existing ones, is an essential process for growth and development. Different mechanisms such as vasculogenesis, sprouting, intussusceptive, and coalescent angiogenesis, as well as vessel co-option, vasculogenic mimicry and lymphangiogenesis, underlie the formation of new vasculature. In many pathological conditions, such as cancer, atherosclerosis, arthritis, psoriasis, endometriosis, obesity and SARS-CoV-2(COVID-19), developmental angiogenic processes are recapitulated, but are often done so without the normal feedback mechanisms that regulate the ordinary spatial and temporal patterns of blood vessel formation. Thus, pathological angiogenesis presents new challenges yet new opportunities for the design of vascular-directed therapies. Here, we provide an overview of recent insights into blood vessel development and highlight novel therapeutic strategies that promote or inhibit the process of angiogenesis to stabilize, reverse, or even halt disease progression. In our review, we will also explore several additional aspects (the angiogenic switch, hypoxia, angiocrine signals, endothelial plasticity, vessel normalization, and endothelial cell anergy) that operate in parallel to canonical angiogenesis mechanisms and speculate how these processes may also be targeted with anti-angiogenic or vascular-directed therapies.
Collapse
Affiliation(s)
- Andrew C Dudley
- Department of Microbiology, Immunology and Cancer Biology, The University of Virginia, Charlottesville, VA, 22908, USA.
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
6
|
Pardridge WM. Receptor-mediated drug delivery of bispecific therapeutic antibodies through the blood-brain barrier. FRONTIERS IN DRUG DELIVERY 2023; 3:1227816. [PMID: 37583474 PMCID: PMC10426772 DOI: 10.3389/fddev.2023.1227816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
Therapeutic antibody drug development is a rapidly growing sector of the pharmaceutical industry. However, antibody drug development for the brain is a technical challenge, and therapeutic antibodies for the central nervous system account for ~3% of all such agents. The principal obstacle to antibody drug development for brain or spinal cord is the lack of transport of large molecule biologics across the blood-brain barrier (BBB). Therapeutic antibodies can be made transportable through the blood-brain barrier by the re-engineering of the therapeutic antibody as a BBB-penetrating bispecific antibody (BSA). One arm of the BSA is the therapeutic antibody and the other arm of the BSA is a transporting antibody. The transporting antibody targets an exofacial epitope on a BBB receptor, and this enables receptor-mediated transcytosis (RMT) of the BSA across the BBB. Following BBB transport, the therapeutic antibody then engages the target receptor in brain. RMT systems at the BBB that are potential conduits to the brain include the insulin receptor (IR), the transferrin receptor (TfR), the insulin-like growth factor receptor (IGFR) and the leptin receptor. Therapeutic antibodies have been re-engineered as BSAs that target the insulin receptor, TfR, or IGFR RMT systems at the BBB for the treatment of Alzheimer's disease and Parkinson's disease.
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: 189] [Impact Index Per Article: 189.0] [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
|
Yang L, Dong Z, Li S, Chen T. ESM1 promotes angiogenesis in colorectal cancer by activating PI3K/Akt/mTOR pathway, thus accelerating tumor progression. Aging (Albany NY) 2023; 15:2920-2936. [PMID: 37100467 DOI: 10.18632/aging.204559] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 01/23/2023] [Indexed: 04/28/2023]
Abstract
BACKGROUND This study aimed to explore the influence of endothelial cell-specific molecule 1 (ESM1) expression on colorectal cancer (CRC) cells and preliminarily analyze its possible mechanism, so as to lay a foundation for research about potential biological targets of CRC. METHODS First, CRC cells were transfected with ESM1-negative control (NC), ESM1-mimic and ESM1-inhibitor and randomly assigned to ESM1-NC group, ESM1-mimic group and ESM1-inhibitor group, respectively. Then the cells were harvested at 48 h after transfection for subsequent experiments. RESULTS The results manifested that after up-regulation of ESM1, the distance of CRC SW480 and SW620 cell lines migrating to the scratch center rose notably, and the number of migrating cells, basement membrane-penetrating cells, colonies formed and angiogenesis was increased overtly, indicating that ESM1 overexpression can promote tumor angiogenesis in CRC and accelerate tumor progression. Combined with results of bioinformatics analysis, the molecular mechanism by which ESM1 promoted tumor angiogenesis in CRC and accelerated tumor progression was explored through suppressing the protein expression of phosphatidylinositol 3-kinase (PI3K). Western blotting revealed that after intervention with PI3K inhibitor, the protein expressions of phosphorylated PI3K (p-PI3K), phosphorylated protein kinase B (p-Akt) and phosphorylated mammalian target of rapamycin (p-mTOR) were decreased evidently, and the protein expressions of matrix metalloproteinase-2 (MMP-2), MMP-3, MMP-9, Cyclin D1, Cyclin A2, VEGF, COX-2 and HIF-1α subsequently declined. CONCLUSION ESM1 may promote angiogenesis in CRC by activating the PI3K/Akt/mTOR pathway, thus accelerating tumor progression.
Collapse
Affiliation(s)
- Liqun Yang
- General Surgery, Tangshan Fengnan District Hospital, Fengnan, Tangshan 063300, China
| | - Zhigang Dong
- General Surgery, Tangshan Fengnan District Hospital, Fengnan, Tangshan 063300, China
| | - Shuyu Li
- Two Divisions of The Cardiovascular Duct, Affiliated Hospital of North China University of Science and Technology, Lubei, Tangshan 063300, China
| | - Tieliang Chen
- General Surgery, Tangshan Union Hospital, Lunan, Tangshan 063300, China
| |
Collapse
|
9
|
Ucaryilmaz Metin C, Ozcan G. The HIF-1α as a Potent Inducer of the Hallmarks in Gastric Cancer. Cancers (Basel) 2022; 14:2711. [PMID: 35681691 PMCID: PMC9179860 DOI: 10.3390/cancers14112711] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 02/01/2023] Open
Abstract
Hypoxia is the principal architect of the topographic heterogeneity in tumors. Hypoxia-inducible factor-1α (HIF-1α) reinforces all hallmarks of cancer and donates cancer cells with more aggressive characteristics at hypoxic niches. HIF-1α potently induces sustained growth factor signaling, angiogenesis, epithelial-mesenchymal transition, and replicative immortality. Hypoxia leads to the selection of cancer cells that evade growth suppressors or apoptotic triggers and deregulates cellular energetics. HIF-1α is also associated with genetic instability, tumor-promoting inflammation, and escape from immunity. Therefore, HIF-1α may be an important therapeutic target in cancer. Despite that, the drug market lacks safe and efficacious anti-HIF-1α molecules, raising the quest for fully unveiling the complex interactome of HIF-1α in cancer to discover more effective strategies. The knowledge gap is even wider in gastric cancer, where the number of studies on hypoxia is relatively low compared to other well-dissected cancers. A comprehensive review of the molecular mechanisms by which HIF-1α induces gastric cancer hallmarks could provide a broad perspective to the investigators and reveal missing links to explore in future studies. Thus, here we review the impact of HIF-1α on the cancer hallmarks with a specific focus on gastric cancer.
Collapse
Affiliation(s)
| | - Gulnihal Ozcan
- Department of Medical Pharmacology, School of Medicine, Koç University, 34450 Istanbul, Turkey
| |
Collapse
|
10
|
Extracellular vimentin mimics VEGF and is a target for anti-angiogenic immunotherapy. Nat Commun 2022; 13:2842. [PMID: 35606362 PMCID: PMC9126915 DOI: 10.1038/s41467-022-30063-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
Anti-angiogenic cancer therapies possess immune-stimulatory properties by counteracting pro-angiogenic molecular mechanisms. We report that tumor endothelial cells ubiquitously overexpress and secrete the intermediate filament protein vimentin through type III unconventional secretion mechanisms. Extracellular vimentin is pro-angiogenic and functionally mimics VEGF action, while concomitantly acting as inhibitor of leukocyte-endothelial interactions. Antibody targeting of extracellular vimentin shows inhibition of angiogenesis in vitro and in vivo. Effective and safe inhibition of angiogenesis and tumor growth in several preclinical and clinical studies is demonstrated using a vaccination strategy against extracellular vimentin. Targeting vimentin induces a pro-inflammatory condition in the tumor, exemplified by induction of the endothelial adhesion molecule ICAM1, suppression of PD-L1, and altered immune cell profiles. Our findings show that extracellular vimentin contributes to immune suppression and functions as a vascular immune checkpoint molecule. Targeting of extracellular vimentin presents therefore an anti-angiogenic immunotherapy strategy against cancer. The pro-tumorigenic effects of vimentin have been attributed to intracellular functions in tumour cells so far. Here, the authors show that tumour endothelial cells can secrete vimentin as a pro-angiogenic factor and that targeting of vimentin can be used as an immunotherapeutic strategy.
Collapse
|
11
|
Du H, Sun Y, Zhang Y, Wang S, Zhu H, Chen S, Pan H. Interaction of PM 2.5 and pre-pregnancy body mass index on birth weight: A nationwide prospective cohort study. Front Endocrinol (Lausanne) 2022; 13:963827. [PMID: 35957820 PMCID: PMC9360486 DOI: 10.3389/fendo.2022.963827] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/05/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Fine particulate matter (PM2.5), one of the most common air pollutants worldwide, has been associated with many adverse birth outcomes in some studies. Pre-pregnancy body mass index (BMI) is an important indicator of maternal obesity that may also contribute to a wide range of birthweight outcomes. Both PM2.5 and maternal obesity have been found associated with issues on neonatal birthweight respectively, and more attentions and interests are focusing on their combined effect on pregnancy outcomes. PURPOSE To explore the modifying effect of pre-pregnancy BMI on the association between gestational PM2.5 and birthweight; to investigate the interactive effect between gestational PM2.5 and pre-pregnancy BMI on birthweight among pregnant women during three trimesters and the whole pregnancy. METHODS This nationwide cohort study used the National Free Preconception Health Examination Project (NFPHEP) data collected from January 1, 2010, to December 31, 2012. A total population of 248,501 Chinese women from 220 counties registered this project. Pre-pregnancy BMI as a common anthropometric examination was collected during preconception investigation, and gestational PM2.5 was derived from a hindcast model for historical PM2.5 estimation from satellite-retrieved aerosol optic depth. Subgroup analysis was conducted to explore a potential modifying effect on the association between PM2.5 and birthweight during pregnancy by four pre-pregnancy BMI subgroups. Interaction analysis by introducing product terms to multivariable linear regression was also used to examine whether there was an interactive relationship between PM2.5 and pre-pregnancy BMI. RESULTS Totally, 193,461 participants were included in our study. The average concentration of PM2.5 was 75.33 μg/m3. Higher exposure of PM2.5 during the entire pregnancy was associated with higher birthweight (17.15 g per 10 μg/m3; 95% CI:16.15, 18.17). Each 10 μg/m3 increase in PM2.5 during the first, second, and third trimesters was associated with increases in birthweight by 14.93 g (95%CI: 13.96, 15.89), 13.75 g (95% CI: 12.81, 14.69), and 8.79 g (95% CI: 8.09, 9.49), respectively. Higher pre-pregnancy BMI per kg/m2 was associated with an increase of birthweight by 7.012 g (95% CI: 6.121, 7.902). Product terms between PM2.5 and pre-pregnancy BMI were significant for the first, second trimesters, and the entire duration of pregnancy. CONCLUSIONS Our results found both gestational PM2.5 exposure and pre-pregnancy BMI respectively correlated with the increase of birthweight. A negative interaction between pre-pregnancy BMI and gestational PM2.5 was discovered in term of birthweight gain. Avoidance of high-dose exposure to PM2.5 during the early and middle stages of pregnancy and pre-pregnancy overweight/obesity may help prevent high birthweight.
Collapse
Affiliation(s)
- Hanze Du
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuxin Sun
- Eight-Year Program of Clinical Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuelun Zhang
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shirui Wang
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Huijuan Zhu
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Shi Chen
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Shi Chen, ; Hui Pan,
| | - Hui Pan
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: Shi Chen, ; Hui Pan,
| |
Collapse
|
12
|
Effects of Intravitreal Aflibercept on the Systemic Insulin-like Growth Factor-I and Vascular Endothelial Growth Factor-A in Patients with Diabetic Retinopathy and Age-Related Macular Degeneration. J Ophthalmol 2021; 2021:7058505. [PMID: 34956670 PMCID: PMC8694974 DOI: 10.1155/2021/7058505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/18/2021] [Indexed: 11/18/2022] Open
Abstract
Purpose To analyze the effect of intravitreal aflibercept injections on systemic levels of insulin-like growth factor-1 and vascular endothelial growth factor-A in patients with diabetic retinopathy and age-related macular degeneration. Methods Vascular endothelial growth factor-A and insulin-like growth factor-1 levels were determined before and one week and four weeks after intravitreal injection of aflibercept (2.0 mg/50 μl) for 19 patients with age-related macular degeneration (mean age, 76 ± 11 years) and 18 patients with diabetic retinopathy (mean age, 64 ± 14 years). Twenty-two healthy individuals were enrolled as controls. Results A significant decline in systemic vascular endothelial growth factor-A level, from 43 (30–57) pg/ml at baseline to 8 (8–8) pg/ml (p < 0.001) at week one and 17 (8–25) pg/ml (p=0.0054) at week four, was observed in the age-related macular degeneration group. In the diabetic retinopathy group, vascular endothelial growth factor-A levels declined from 53 (35–117) pg/ml to 2 (1–5) pg/ml (p < 0.0001) one week after injection and 16 (13–22) pg/ml four weeks after injection (p=0.0327). At baseline, systemic insulin-like growth factor-1 concentration was higher in the diabetic retinopathy group (57 [37–99] pg/ml) than in the age-related macular degeneration group (35 [24–51] pg/ml) (p=0.0056). A subgroup analysis showed that patients in the proliferative diabetic retinopathy subgroup had significantly higher systemic insulin-like growth factor-1 concentrations (71 [44.7–243] pg/ml) than those in the nonproliferative diabetic retinopathy subgroup (43 [29–66] pg/ml) (p=0.0048). Conclusions The difference between the baseline systemic insulin-like growth factor-1 levels of the age-related macular degeneration and diabetic retinopathy groups and the higher insulin-like growth factor-1 levels in the proliferative diabetic retinopathy subgroup one week after aflibercept therapy suggest that insulin-like growth factor-1 may play a role in the pathomechanism of diabetic retinopathy.
Collapse
|
13
|
Komata Y, Tsubota S, Sakamoto K, Ikematsu S, Kadomatsu K. Screening of novel Midkine binding protein by BioID2-based proximity labeling. NAGOYA JOURNAL OF MEDICAL SCIENCE 2021; 83:495-508. [PMID: 34552285 PMCID: PMC8438011 DOI: 10.18999/nagjms.83.3.495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/09/2020] [Indexed: 12/31/2022]
Abstract
Midkine (MK), a heparin-binding growth factor, is associated with the poor prognosis of the pediatric tumor, neuroblastoma. MK would be a druggable target as many studies showed inhibition of its function in various cancers suppressed tumor developments. To establish the therapy targeting MK, identification of its binding partners, and elucidation of its intracellular signaling are needed. It was reported that exogenous MK induced phosphorylation of ribosomal protein S6 (RPS6) downstream of mTOR signaling. Using RPS6 phosphorylation as a marker of MK response, we searched for MK reactive cell lines. We found that MK cell lines expressing less MK tended to respond better to MK. Next, using an MK reactive neuroblastoma cell line, MK-knocked down SH-SY5Y cells, we employed a proximity-dependent biotin identification method, which was invented to evaluate protein-protein interactions by biotinylation. We confirmed that secreted MK fused to the biotin ligase BioID2 (MK-BioID2) was able to biotinylate proteins from the cells. Biotinylated proteins were identified by liquid chromatography-mass spectrometry analyses. Twenty five proteins were found to be overlapped after three independent experiments, among which insulin-like growth binding protein 2 (IGFBP2) was further analyzed. IGFBP2 was indeed detected with immunoblotting after streptavidin pull down of MK-BioID2 labeled cell extract of MK-knocked down SH-SY5Y cells. Our study suggests that the BioID2 method is useful to identify binding partners of growth factors.
Collapse
Affiliation(s)
- Yosuke Komata
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shoma Tsubota
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuma Sakamoto
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
| | - Shinya Ikematsu
- Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Naha, Japan
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
| |
Collapse
|
14
|
IGF1-mediated HOXA13 overexpression promotes colorectal cancer metastasis through upregulating ACLY and IGF1R. Cell Death Dis 2021; 12:564. [PMID: 34075028 PMCID: PMC8169856 DOI: 10.1038/s41419-021-03833-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/16/2022]
Abstract
Metastasis is the major reason for the high mortality of colorectal cancer (CRC) patients and its molecular mechanism remains unclear. Here, we report a novel role of Homeobox A13 (HOXA13), a member of the Homeobox (HOX) family, in promoting CRC metastasis. The elevated expression of HOXA13 was positively correlated with distant metastasis, higher AJCC stage, and poor prognosis in two independent CRC cohorts. Overexpression of HOXA13 promoted CRC metastasis whereas downregulation of HOXA13 suppressed CRC metastasis. Mechanistically, HOXA13 facilitated CRC metastasis by transactivating ATP-citrate lyase (ACLY) and insulin-like growth factor 1 receptor (IGF1R). Knockdown of ACLY and IGFIR inhibited HOXA13-medicated CRC metastasis, whereas ectopic overexpression of ACLY and IGFIR rescued the decreased CRC metastasis induced by HOXA13 knockdown. Furthermore, Insulin-like growth factor 1 (IGF1), the ligand of IGF1R, upregulated HOXA13 expression through the PI3K/AKT/HIF1α pathway. Knockdown of HOXA13 decreased IGF1-mediated CRC metastasis. In addition, the combined treatment of ACLY inhibitor ETC-1002 and IGF1R inhibitor Linsitinib dramatically suppressed HOXA13-mediated CRC metastasis. In conclusion, HOXA13 is a prognostic biomarker in CRC patients. Targeting the IGF1-HOXA13-IGF1R positive feedback loop may provide a potential therapeutic strategy for the treatment of HOXA13-driven CRC metastasis.
Collapse
|
15
|
IGF2 deficiency causes mitochondrial defects in skeletal muscle. Clin Sci (Lond) 2021; 135:979-990. [PMID: 33825857 PMCID: PMC8055961 DOI: 10.1042/cs20210128] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 01/25/2023]
Abstract
Exercise training improves muscle fitness in many aspects, including induction of mitochondrial biogenesis and maintenance of mitochondrial dynamics. The insulin-like growth factors were recently proposed as key regulators of myogenic factors to regulate muscle development. The present study aimed to investigate the physical exercise impact on insulin-like growth factor 2 (IGF2) and analyzed its functions on skeletal muscle cells in vitro. Using online databases, we stated that IGF2 was relatively highly expressed in skeletal muscle cells and increased after exercise training. Then, IGF2 deficiency in myotubes from C2C12 and primary skeletal muscle cells (PMSCs) led to impaired mitochondrial function, reduced mitochondria-related protein content, and decreased mitochondrial biogenesis. Furthermore, we explored the possible regulatory pathway and found that mitochondrial regulation in skeletal muscle cells might occur through IGF2-Sirtuin 1 (SIRT1)-peroxisome proliferator-activated receptor-γ co-activator-1α (PGC1α) signaling pathway. Therefore, the present study first demonstrated the relationship between IGF2 and mitochondria in skeletal muscle.
Collapse
|
16
|
Chen L, Samanta A, Zhao L, Dudley NR, Buehler T, Vincent RJ, Hauptman J, Girgis M, Dawn B. Vitamin D3 induces mesenchymal-to-endothelial transition and promotes a proangiogenic niche through IGF-1 signaling. iScience 2021; 24:102272. [PMID: 33817577 PMCID: PMC8005757 DOI: 10.1016/j.isci.2021.102272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/09/2021] [Accepted: 03/02/2021] [Indexed: 12/31/2022] Open
Abstract
Although vitamin D3 (VitD3) prevents angiogenesis in cancer, VitD3 deficiency is associated with greater incidence of cardiovascular events in patients. We examined the influence of VitD3 on the angiogenic potential of mesenchymal stem cells (MSCs). VitD3 treatment increased the expression of proangiogenic molecules in MSCs, which exhibited an endothelial cell-like phenotype and promoted vascularization in vitro and in vivo. VitD3 activated the IGF-1 promoter and boosted IGF-1 receptor (IGF-1R) signaling, which was essential for the mesenchymal-to-endothelial transition (MEndoT) of MSCs. VitD3-treated MSCs created a proangiogenic microenvironment for co-cultured arterial endothelial cells, as well as aortic rings. The induction of MEndoT and angiogenesis promotion by VitD3-stimulated MSCs was attenuated by IGF-1R inhibitor picropodophyllin. We conclude that VitD3 promotes MEndoT in MSCs, and VitD3-treated MSCs augment vascularization by producing a proangiogenic niche through continued IGF-1 secretion. These results suggest a potential therapeutic role of VitD3 toward enhancing MSC-induced angiogenesis. Vitamin D3 (VitD3) treatment induces IGF-1 in mesenchymal stem cells (MSCs) VitD3 promotes mesenchymal-to-endothelial transition in MSCs via IGF-1 signaling Continued IGF-1 secretion by VitD3-treated MSCs creates a proangiogenic niche VitD3 may enhance MSC-induced angiogenesis through dual mechanisms
Collapse
Affiliation(s)
- Lei Chen
- Department of Cardiovascular Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Anweshan Samanta
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Lin Zhao
- Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine, 1701 W. Charleston Boulevard, Suite 230, Las Vegas, NV 89102, USA
| | - Nathaniel R Dudley
- Department of Cardiovascular Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Tanner Buehler
- Department of Cardiovascular Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Robert J Vincent
- Department of Cardiovascular Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jeryl Hauptman
- Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine, 1701 W. Charleston Boulevard, Suite 230, Las Vegas, NV 89102, USA
| | - Magdy Girgis
- Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine, 1701 W. Charleston Boulevard, Suite 230, Las Vegas, NV 89102, USA
| | - Buddhadeb Dawn
- Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine, 1701 W. Charleston Boulevard, Suite 230, Las Vegas, NV 89102, USA
| |
Collapse
|
17
|
Di Matteo A, Belloni E, Pradella D, Cappelletto A, Volf N, Zacchigna S, Ghigna C. Alternative splicing in endothelial cells: novel therapeutic opportunities in cancer angiogenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:275. [PMID: 33287867 PMCID: PMC7720527 DOI: 10.1186/s13046-020-01753-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
Alternative splicing (AS) is a pervasive molecular process generating multiple protein isoforms, from a single gene. It plays fundamental roles during development, differentiation and maintenance of tissue homeostasis, while aberrant AS is considered a hallmark of multiple diseases, including cancer. Cancer-restricted AS isoforms represent either predictive biomarkers for diagnosis/prognosis or targets for anti-cancer therapies. Here, we discuss the contribution of AS regulation in cancer angiogenesis, a complex process supporting disease development and progression. We consider AS programs acting in a specific and non-redundant manner to influence morphological and functional changes involved in cancer angiogenesis. In particular, we describe relevant AS variants or splicing regulators controlling either secreted or membrane-bound angiogenic factors, which may represent attractive targets for therapeutic interventions in human cancer.
Collapse
Affiliation(s)
- Anna Di Matteo
- Istituto di Genetica Molecolare, "Luigi Luca Cavalli-Sforza", Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100, Pavia, Italy
| | - Elisa Belloni
- Istituto di Genetica Molecolare, "Luigi Luca Cavalli-Sforza", Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100, Pavia, Italy
| | - Davide Pradella
- Istituto di Genetica Molecolare, "Luigi Luca Cavalli-Sforza", Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100, Pavia, Italy
| | - Ambra Cappelletto
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Nina Volf
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy
| | - Serena Zacchigna
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149, Trieste, Italy. .,Department of Medical, Surgical and Health Sciences, University of Trieste, 34149, Trieste, Italy.
| | - Claudia Ghigna
- Istituto di Genetica Molecolare, "Luigi Luca Cavalli-Sforza", Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100, Pavia, Italy.
| |
Collapse
|
18
|
IGF-binding proteins 3 and 4 are regulators of sprouting angiogenesis. Mol Biol Rep 2020; 47:2561-2572. [PMID: 32133604 DOI: 10.1007/s11033-020-05339-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 02/18/2020] [Indexed: 12/29/2022]
Abstract
PURPOSE We have previously identified insulin-like growth factor 2 (IGF2) and insulin-like growth factor 1 receptor (IGF1R) as essential proteins for tip cell maintenance and sprouting angiogenesis. In this study, we aim to identify other IGF family members involved in endothelial sprouting angiogenesis. METHODS Effects on sprouting were analyzed in human umbilical vein endothelial cells (HUVECs) using the spheroid-based sprouting model, and were quantified as mean number of sprouts per spheroid and average sprout length. RNA silencing technology was used to knockdown gene expression. Recombinant forms of the ligands (IGF1 and IGF2, insulin) and the IGF-binding proteins (IGFBP) 3 and 4 were used to induce excess effects. Effects on the tip cell phenotype were analyzed by measuring the fraction of CD34+ tip cells using flow cytometry and immunohistochemistry in a 3D angiogenesis model. Experiments were performed in the presence and absence of serum. RESULTS Knockdown of IGF2 inhibited sprouting in HUVECs, in particular when cultured in the absence of serum, suggesting that components in serum influence the signaling of IGF2 in angiogenesis in vitro. We then determined the effects of IGFBP3 and IGFBP4, which are both present in serum, on IGF2-IGF1R signaling in sprouting angiogenesis in the absence of serum: knockdown of IGFBP3 significantly reduced sprouting angiogenesis, whereas knockdown of IGFBP4 resulted in increased sprouting angiogenesis in both flow cytometry analysis and immunohistochemical analysis of the 3D angiogenesis model. Other IGF family members except INSR did not affect IGF2-IGF1R signaling. CONCLUSIONS Serum components and IGF binding proteins regulate IGF2 effects on sprouting angiogenesis. Whereas IGFBP3 acts as co-factor for IGF2-IGF1R binding, IGFBP4 inhibits IGF2 signaling.
Collapse
|
19
|
Shapiro MR, Atkinson MA, Brusko TM. Pleiotropic roles of the insulin-like growth factor axis in type 1 diabetes. Curr Opin Endocrinol Diabetes Obes 2019; 26:188-194. [PMID: 31145130 PMCID: PMC7135378 DOI: 10.1097/med.0000000000000484] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW We review studies demonstrating lowered levels of insulin-like growth factors (IGFs) in patients with recent-onset type 1 diabetes (T1D) and discuss their potential roles in the disorder's pathogenesis. RECENT FINDINGS IGFs have long been recognized as a class of hormones that promote growth, development, and cellular metabolism throughout the human body. More recently, studies have noted an association between reduced pancreatic weight/volume and T1D. Thus, we believe it is important to understand pancreatic regulation of IGF expression and bioavailability, as well as the impact of IGFs on pancreatic growth and islet health. Additional studies of IGFs have been extended to their influence on the inflammatory/regulatory balance of monocytes, B cells, and T cells; features which have been previously established to show dysregulation in settings of T1D. SUMMARY These data suggest that IGFs may prevent known impairments in the pancreas and immune system in T1D and underscore the need to extend these studies, some of which were performed in health or other autoimmune diseases, toward T1D specifically. Collectively, the work emphasized here support the potential therapeutic use of IGFs in T1D prevention efforts as pancreatic growth factors and/or immunoregulatory agents.
Collapse
Affiliation(s)
- Melanie R. Shapiro
- Department of Pathology, Immunology, and Laboratory Medicine, The University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Mark A. Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, The University of Florida Diabetes Institute, Gainesville, Florida, USA
- Department of Pediatrics, The University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Todd M. Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, The University of Florida Diabetes Institute, Gainesville, Florida, USA
| |
Collapse
|
20
|
A therapeutic approach towards microRNA29 family in vascular diabetic complications: A boon or curse? J Diabetes Metab Disord 2019; 18:243-254. [PMID: 31275895 DOI: 10.1007/s40200-019-00409-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 05/02/2019] [Indexed: 02/06/2023]
Abstract
Diabetes Mellitus (DM) is one of the major metabolic disorders and its severity leads to death. Enhancement in hyperglycaemic conditions of DM gives rise to endothelial impairment in small and large blood vessels contributing towards microvascular and macrovascular complications respectively. The pathogenesis of diabetic complications is associated with interruption of various signal transduction pathways due to epigenetic modifications such as aberrant histone modifications, DNA methylation and expression of miRNAs along with the long non-coding RNAs (lncRNAs). Amongst these epigenetic alterations, modulated expressions of miRNAs confer to apoptosis and endothelial dysfunction of organs that gives rise to vascular complications. In this review, we principally focussed on physiological role of miR29 family in DM and have discussed crosstalk between miR29 family and numerous genes involved in signal transduction pathways of Diabetic vascular complications. Incidences of diabetic retinopathy exploiting the role of miR29 in regulation of EMT process, differential expression patterns of miR29 and promising therapeutic role of miR29 have been discussed. We have summarised the therapeutic role of miR29 in podocyte impairment and how miR29 regulates the expressions of profibrotic, inflammatory and ECM encoding genes in renal fibrosis under diabetic conditions. We have also highlighted impact of miR29 expression patterns in cardiac angiopathy, cardiomyocyte's apoptosis and cardiac fibrosis. Additionally, we have also presented the contradictory actions of miR29 family in amelioration as well as in enhancement of diabetic complications.
Collapse
|
21
|
Oncofoetal insulin receptor isoform A marks the tumour endothelium; an underestimated pathway during tumour angiogenesis and angiostatic treatment. Br J Cancer 2018; 120:218-228. [PMID: 30559346 PMCID: PMC6342959 DOI: 10.1038/s41416-018-0347-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/17/2018] [Accepted: 10/24/2018] [Indexed: 12/29/2022] Open
Abstract
Background In a genomic screen for determinants of the tumour vasculature, we identified insulin receptor (INSR) to mark the tumour endothelium. As a functional role for insulin/INSR in cancer has been suggested and markers of the tumour endothelium may be attractive therapeutic targets, we investigated the role of INSR in angiogenesis. Methods In a genomic screen for determinants of the tumour vasculature we identified insulin receptor to mark the tumour endothelium. Results The current report demonstrates the following: (i) the heavy overexpression of INSR on angiogenic vasculature in human tumours and the correlation to short survival, (ii) that INSR expression in the tumour vasculature is mainly representing the short oncofoetal and non-metabolic isoform INSR-A, (iii) the angiogenic activity of insulin on endothelial cells (EC) in vitro and in vivo, (iv) suppression of proliferation and sprouting of EC in vitro after antibody targeting or siRNA knockdown, and (v) inhibition of in vivo angiogenesis in the chicken chorioallantoic membrane (CAM) by anti-INSR antibodies. We additionally show, using preclinical mouse as well as patient data, that treatment with the inhibitor sunitinib significantly reduces the expression of INSR-A. Conclusions The current study underscores the oncogenic impact of INSR and suggests that targeting the INSR-A isoform should be considered in therapeutic settings.
Collapse
|
22
|
Tonietto L, Vasquez AF, Dos Santos LA, Weber JB. Histological and structural evaluation of growth hormone and PLGA incorporation in macroporous scaffold of α-tricalcium phosphate cement. J Biomater Appl 2018; 33:866-875. [PMID: 30426862 DOI: 10.1177/0885328218812173] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
An in vivo study was conducted to evaluate the effects of the incorporation of fibers of poly(lactic acid-co-glycolic acid, PLGA) and poly(isoprene) blend and recombinant human growth hormone (rhGH) in a macroporous scaffold of α-tricalcium phosphate cement (α-TCP) samples inserted into calvarial defects (8 mm in diameter) of 48 Wistar rats. The samples of α-TCP + PLGA/poly(isoprene) blend fibers were also submitted to a mechanical test of flexural strength. The animals of the different experimental groups [1] α-TCP (n = 6); [2] α-TCP + PLGA/poly(isoprene) blend fibers (n = 6); [3] α-TCP + rhGH, (n = 6) and [4] α-TCP + PLGA/poly(isoprene) blend fibers + rhGH, (n = 6) (the numbers within square brackets identify the experimental groups), after two weeks (subdivision "a") and four weeks (subdivision "b"), were euthanized and the implants removed for histological analysis. There was no statistical difference (p > 0.05) between the samples with and without fibers in the mechanical test. Light microscopy revealed good integration of the material in the host tissue, represented by tissue penetration into the macropores and adequate angiogenesis. In the two-week period, the groups [3a] and [4a] were significantly superior (p < 0.05) to the other groups with regard to angiogenesis and bone neoformation. In the four-week period, the group [3b] was significantly superior (p < 0.05) to the other groups with regard to bone neoformation. We conclude that the macroporous α-TCP scaffold used in this study has low mechanical resistance, is biocompatible and has significantly improved the osteoconductive capacity when rhGH is incorporated into its structure.
Collapse
Affiliation(s)
- Leonardo Tonietto
- 1 School of Health Sciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Andres F Vasquez
- 2 Laboratory of Biomaterials (LABIOMAT), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Luís A Dos Santos
- 2 Laboratory of Biomaterials (LABIOMAT), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - João Bb Weber
- 1 School of Health Sciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| |
Collapse
|
23
|
Xu J, Wang J, Zhang S. Mechanisms of resistance to irreversible epidermal growth factor receptor tyrosine kinase inhibitors and therapeutic strategies in non-small cell lung cancer. Oncotarget 2017; 8:90557-90578. [PMID: 29163853 PMCID: PMC5685774 DOI: 10.18632/oncotarget.21164] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/28/2017] [Indexed: 11/25/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) T790M mutation is the most frequent mechanism which accounts for about 60% of acquired resistance to first-generation EGFR tyrosine kinase inhibitors (TKIs) in non-small cell lung cancer (NSCLC) patients harboring EGFR activating mutations. Irreversible EGFR-TKIs which include the second-generation and third-generation EGFR-TKIs are developed to overcome T790M mediated resistance. The second-generation EGFR-TKIs inhibit the wide type (WT) EGFR combined with dose-limiting toxicity which limits its application in clinics, while the development of third-generation EGFR-TKIs brings inspiring efficacy either in vitro or in vivo. The acquired resistance, however, will also occur and limit their response. Understanding the mechanisms of resistance to irreversible EGFR-TKIs plays an important role in the choice of subsequent treatment. In this review, we show the currently known mechanisms of resistance which can be summarized as EGFR dependent and independent mechanisms and potential therapeutic strategies to irreversible EGFR-TKIs.
Collapse
Affiliation(s)
- Jing Xu
- Department of Medical Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Jinghui Wang
- Department of Medical Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Shucai Zhang
- Department of Medical Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Schiegnitz E, Kämmerer PW, Schön H, Gülle C, Berres M, Sagheb K, Al-Nawas B. The matrix metalloproteinase and insulin-like growth factor system in oral cancer - a prospective clinical study. Onco Targets Ther 2017; 10:5099-5105. [PMID: 29123408 PMCID: PMC5661464 DOI: 10.2147/ott.s149231] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim The absence of reliable single serum biomarkers for oral premalignant lesion (OPL) and oral squamous cell carcinoma (OSCC) limits early diagnosis, monitoring of advanced disease, and prediction of prognosis. Methods In this prospective study, serum levels of matrix metalloproteinase (MMP)-2, MMP-3, MMP-13, insulin-like growth factor (IGF)-1, and IGF-binding protein (IGFBP)-3 were measured in 81 untreated OSCC patients, 49 healthy subjects, and 75 individuals with OPLs, and correlated with clinicopathological parameters. Results Serum levels of MMP-3 were significantly higher in OSCC patients compared to healthy subjects (p=0.004). Mean IGF-1 and IGFBP-3 levels in OSCC patients were significantly lower in healthy subjects (p=0.001 and p<0.001). OSCC patients with an IGF-1 serum value <130 ng/mL (median) showed a significantly lower survival rate compared to ≥130 ng/mL (p=0.049). Combined use of IGF-1 (<130 ng/mL) and IGFBP-3 (<3.1 μg/mL) resulted in a significantly lower 12-month cumulative survival compared to the complementary set (78.5% vs 93.8%; p=0.031). There was a significantly positive correlation between IGF-1 and IGFBP-3 serum values (rs =0.625, p<0.001). Conclusion This study shows that IGF-1 and IGFBP-3 have a vital role in the pathogenesis of OSCC and indicates for the first time that IGF-1 and IGFBP-3 in combination may be applied as potential tools for prognosis of OSCC.
Collapse
Affiliation(s)
- Eik Schiegnitz
- Department of Oral and Maxillofacial Surgery, Plastic Surgery, University Medical Center, Johannes Gutenberg University of Mainz, Mainz
| | - Peer W Kämmerer
- Department of Oral and Maxillofacial Surgery, Plastic Surgery, University Medical Centre Rostock, Rostock
| | - Holger Schön
- Department of Oral and Maxillofacial Surgery, Plastic Surgery, University Medical Center, Johannes Gutenberg University of Mainz, Mainz
| | - Christoph Gülle
- Department of Oral and Maxillofacial Surgery, Plastic Surgery, University Medical Center, Johannes Gutenberg University of Mainz, Mainz
| | - Manfred Berres
- Department of Mathematics and Technology, University of Applied Sciences Koblenz, Remagen.,Institute of Medical Biometry, Epidemiology and Informatics, Johannes Gutenberg University of Mainz, Mainz
| | - Keyvan Sagheb
- Department of Oral and Maxillofacial Surgery, Plastic Surgery, University Medical Center, Johannes Gutenberg University of Mainz, Mainz
| | - Bilal Al-Nawas
- Department of Oral and Maxillofacial Surgery, Plastic Surgery, University Medical Centre, Martin-Luther University Halle, Halle, Germany
| |
Collapse
|
25
|
Strzalka-Mrozik B, Kimsa-Furdzik M, Kabiesz A, Michalska-Malecka K, Nita M, Mazurek U. Gene expression levels of the insulin-like growth factor family in patients with AMD before and after ranibizumab intravitreal injections. Clin Interv Aging 2017; 12:1401-1408. [PMID: 28919726 PMCID: PMC5592959 DOI: 10.2147/cia.s135030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Purpose The present study focused on the assessment of the mRNA levels of the insulin-like growth factor (IGF) family in patients with the exudative form of age-related macular degeneration (AMD) before and after ranibizumab intravitreal injections. Patients and methods An analysis of the expression profile of the IGF family of genes in patients with AMD was carried out using the oligonucleotide microarray and quantitative reverse transcriptase polymerase chain reaction (RT-qPCR) methods. Results In the peripheral blood mononuclear cells (PBMCs) obtained from AMD group receiving ranibizumab compared to the peripheral blood mononuclear cells from AMD group before ranibizumab treatment using oligonucleotide microarray technique, six statistically significant differentially expressed transcripts related to the IGF family were detected (unpaired t-test, p<0.05, fold change >1.5). Moreover, analysis using the real-time RT-qPCR technique revealed statistically significant differences in the IGF2 and IGF2R mRNA levels (Mann–Whitney U test, p<0.05) between the two groups that were studied. Statistical analyses of both oligonucleotide microarray and real-time RT-qPCR results demonstrated a significant decreased expression only for IGF2 mRNA. Conclusion Our results revealed a changed expression of IGF2 mRNA after ranibizumab treatment.
Collapse
Affiliation(s)
- Barbara Strzalka-Mrozik
- Department of Molecular Biology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland
| | - Malgorzata Kimsa-Furdzik
- Department of Biochemistry, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Adam Kabiesz
- University Center for Ophthalmology and Oncology, Independent Public Clinical Hospital, Medical University of Silesia, Katowice, Poland
| | - Katarzyna Michalska-Malecka
- University Center for Ophthalmology and Oncology, Independent Public Clinical Hospital, Medical University of Silesia, Katowice, Poland.,Department of Ophthalmology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Malgorzata Nita
- Domestic and Specialized Medicine Centre "Dilmed", Katowice, Poland
| | - Urszula Mazurek
- Department of Molecular Biology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
26
|
Abstract
The development of milk during evolution is considered a more recent step to provide the neonate with adequate amounts of energy, nutrients, and specific hormonal signals thereby, granting a fast and efficient rate of postnatal growth and development. Since the insulin- or the insulin-like growth factor (IGF) systems were evolved much earlier, it can be assumed that the functionality of the IGF-system has been integrated into the novel matrix milk containing casein and whey proteins from the beginnings. In fact, IGFs and IGF-binding proteins (IGFBPs) are abundantly present in milk, which is particularly true for fore-milk or colostrum and the potential effects of milk-borne IGF-compounds on the consuming organisms have in fact been addressed by several studies. Those studies examined, if orally administered IGFs can be absorbed by the consumer's gastro-intestinal tract and thus contribute e.g. to the somatic growth of infants. A second line of studies assessed local effects of milk-borne IGFs on growth and development of the gastro-intestinal tract itself. Finally, distinct functions of isolated IGF-compounds for growth and involution of the mammary gland have also been provided in the past. While the consumption of milk seems not to represent a major source of endogenous IGFs, accumulating evidence indicates secondary effects of milk on the endogenous IGF-system, which may be mediated by micronutrients such as branched amino acids and metabolic programming. By contrast, direct effects on growth and development of oesophageal and intestinal cells have been observed if IGFs were administered orally.
Collapse
Affiliation(s)
- Andreas Hoeflich
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| | - Zianka Meyer
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| |
Collapse
|