1
|
Papa V, Li Pomi F, Borgia F, Vaccaro M, Pioggia G, Gangemi S. Alarmins in cutaneous malignant melanoma: An updated overview of emerging evidence on their pathogenetic, diagnostic, prognostic, and therapeutic role. J Dermatol 2024; 51:927-938. [PMID: 38775220 PMCID: PMC11483971 DOI: 10.1111/1346-8138.17278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 07/04/2024]
Abstract
Malignant cutaneous melanoma is the leading cause of death for skin cancer to date, with globally increasing incidence rates. In this epidemiological scenario, international scientific research is exerting efforts to identify new clinical strategies aimed at the prognostic amelioration of the disease. Very promising and groundbreaking in this context is the scientific interest related to alarmins and their pioneering utility in the setting of the pathogenetic understanding, diagnosis, prognosis, and therapy for malignant cutaneous melanoma. However, the scientific investigations on this matter should not overlook their still well-presented dual and contradictory role. The aim of our critical analysis is to provide an up-to-date overview of the emerging evidence concerning the dichotomous role of alarmins in the aforementioned clinical settings. Our literature revision was based on the extensive body of both preclinical and clinical findings published on the PubMed database over the past 5 years. In addition to this, we offer a special focus on potentially revolutionary new therapeutic frontiers, which, on the strength of their earliest successes in other clinical areas, could inaugurate a new era of personalized and precision medicine in the field of dermato-oncology.
Collapse
Affiliation(s)
- Vincenzo Papa
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical ImmunologyUniversity of MessinaMessinaItaly
| | - Federica Li Pomi
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.)University of PalermoPalermoItaly
| | - Francesco Borgia
- Department of Clinical and Experimental Medicine, Section of DermatologyUniversity of MessinaMessinaItaly
| | - Mario Vaccaro
- Department of Clinical and Experimental Medicine, Section of DermatologyUniversity of MessinaMessinaItaly
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR)MessinaItaly
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical ImmunologyUniversity of MessinaMessinaItaly
| |
Collapse
|
2
|
Zhang L, Wu J, Yin WW, Hu J, Liao L, Ma J, Xu Z, Wu S. Vasculogenic mimicry-associated novel gene signature predicted prognosis and response to immunotherapy in lung adenocarcinoma. Pathol Res Pract 2024; 253:155048. [PMID: 38147724 DOI: 10.1016/j.prp.2023.155048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 12/28/2023]
Abstract
BACKGROUNDS It was highlighted by recent studies on the biological significance of vasculogenic mimicry (VM) in tumorigenicity and progression. However, it is unclear whether VM also plays a potential role in immune regulation and tumor microenvironment (TME) formation. METHODS To identify patterns of VM alterations and VM-associated genetic features in non-small cell lung adenocarcinoma, we have screened 309 VM regulators and performed consensus molecular typing by the NMF algorithm. The ssGSEA and CIBORSORT algorithms were employed to measure the relative infiltration of distinct immune cell subpopulations. Individual tumors with immune responses were evaluated for alteration patterns of VM with typing-based differential genes. RESULTS In 490 LUAD samples, two distinctive VM alteration patterns connected to different clinical outcomes and biochemical pathways were established. TME characterization showed that the observed VM patterns were primarily saturated with cell proliferation and metabolic pathways and higher in immune cell infiltration of the C1 type. Vasculogenic mimicry-related genes (VMRG) risk scores were constructed to divide patients with lung adenocarcinoma into subgroups with high and low scores. Patients with lower scores had better immunological scores and longer survival times. Upon further investigation, higher scores were positively correlated with higher tumor mutation burden (TMB), M1-type macrophages and immune checkpoint molecules. Nevertheless, in two other immunotherapy cohorts, individuals with lower scores had enhanced immune responses and long-lasting therapeutic benefits. Finally, we monitored the ANLN gene from the VMRG model, which was highly expressed in lung adenocarcinoma tissues and negatively correlated with prognosis; it was also highly expressed in lung adenocarcinoma cell lines, and knockdown of ANLN elicited low expression of VEGFA, MMP2 and MMP9. CONCLUSION This study highlights that VM modifications are significantly associated with the diversity and complexity of TME, revealing new features of the immune microenvironment in lung adenocarcinoma and providing a new strategy for immunotherapy. Screening ANLN as a critical target for vasculogenic mimicry in lung adenocarcinoma provides a novel perspective for the targeted treatment of lung adenocarcinoma.
Collapse
Affiliation(s)
- Lei Zhang
- Department of General Surgery, the Second Affiliated Hospital of Bengbu Medical University, Anhui Province 233080, China
| | - Jiatao Wu
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease,Molecular Diagnosis Center,First Affiliated Hospital,Bengbu Medical University, 287 Changhuai Road, Anhui, Bengbu 233004, China
| | - Wei Wei Yin
- Department of Thoracic Surgery, the Second Affiliated Hospital of Bengbu Medical University, Anhui Province 233080, China
| | - Junjie Hu
- Department of Radiotherapy, the Second Affiliated Hospital of Bengbu Medical University, Anhui Province 233080, China
| | - Lingli Liao
- Department of Clinical Nutrition, the First People's Hospital of Yibin, Sichuan Province 644000, China
| | - Junjie Ma
- Bengbu Medical University, Anhui Province 233030, China
| | - Ziwei Xu
- Bengbu Medical University, Anhui Province 233030, China
| | - Shiwu Wu
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease,Molecular Diagnosis Center,First Affiliated Hospital,Bengbu Medical University, 287 Changhuai Road, Anhui, Bengbu 233004, China; Anhui No. 2 Provincial People's Hospital, Hefei 230041, China.
| |
Collapse
|
3
|
Du J, Du Y, Chen L, Liu H. IL-17 promotes melanoma through TRAF2 as a scaffold protein recruiting PIAS2 and ELAVL1 to induce EPHA5. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119547. [PMID: 37481078 DOI: 10.1016/j.bbamcr.2023.119547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 07/11/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
An abnormal immune response induces melanoma development. IL-17 and the classical downstream signal STAT1 are associated with melanoma development. TRAF2 also mediates the downstream signaling of IL-17; however, its role in IL-17-stimulated melanoma remains unclear. Bioinformatic analysis revealed that TRAF2 can bind to PIAS2 (a SUMO E3 ligase), ELAVL1 (an RNA-binding protein), and EPHA5 (an ephrin receptor of the tyrosine kinase family). To elucidate the IL-17 downstream signal, the IL-17 receptor (R), STAT1, TRAF2, PIAS2, ELAVL1, and EPHA5 were knocked down before melanoma cells were treated with recombinant IL-17A protein. Co-immunoprecipitation and RNA immunoprecipitation were conducted to determine the interaction of TRAF2 with PIAS2, ELAVL1, and EPHA5 proteins, as well as the interaction of ELAVL1 protein with EPHA5 mRNA. STAT1 knockdown suppressed the proliferation and invasion triggered by IL-17A, but the suppressive effects were much weaker than those caused by IL-17R knockdown. This implies that another nonclassical signal mediates IL-17 effects. IL-17A induces TRAF2 recruitment of ELAVL1, PIAS2, and EPHA5 proteins. We speculated that ELAVL1 bound to the AU-rich elements in the 3' untranslated region of the EPHA5 mRNA, thereby enhancing mRNA stability. Furthermore, PIAS2 induced EPHA5 SUMOylation, which suppressed EPHA5 ubiquitination and degradation. Through pre- and post-translational regulation, IL-17A induced EPHA5 expression in melanoma, and EPHA5 knockdown markedly suppressed IL-17A-induced proliferation and invasion. This study revealed a non-classical signaling mechanism responsible for the effects of IL-17 in melanoma.
Collapse
Affiliation(s)
- Junfeng Du
- Department of Plastic Surgery, the first affiliated hospital of Jinan University, No. 613, Huangpu Avenue West, Tianhe District, Guangzhou 510630, China
| | - Yujia Du
- Medical college of Jianghan University, No. 8, Sanjiaohu Road, Wuhan Economic and Technological Development Zone, Wuhan 430014, China
| | - Lang Chen
- Department of Burns and Plastic, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Hongwei Liu
- Department of Plastic Surgery, the first affiliated hospital of Jinan University, No. 613, Huangpu Avenue West, Tianhe District, Guangzhou 510630, China; Innovative Technology Research Institute of Plastic Surgery, Guangzhou 510630, People's Republic of China; Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou 510632, People's Republic of China.
| |
Collapse
|
4
|
Koroknai V, Szász I, Balázs M. Gene Expression Changes in Cytokine and Chemokine Receptors in Association with Melanoma Liver Metastasis. Int J Mol Sci 2023; 24:ijms24108901. [PMID: 37240247 DOI: 10.3390/ijms24108901] [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: 04/18/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Cytokines and chemokines (chemotactic cytokines) are soluble extracellular proteins that bind to specific receptors and play an integral role in the cell-to-cell signaling network. In addition, they can promote the homing of cancer cells into different organs. We investigated the potential relationship between human hepatic sinusoidal endothelial cells (HHSECs) and several melanoma cell lines for the expression of chemokine and cytokine ligands and receptor expression during the invasion of melanoma cells. In order to identify differences in gene expression related to invasion, we selected invasive and non-invasive subpopulations of cells after co-culturing with HHSECs and identified the gene expression patterns of 88 chemokine/cytokine receptors in all cell lines. Cell lines with stable invasiveness and cell lines with increased invasiveness displayed distinct profiles of receptor genes. Cell lines with increased invasive capacity after culturing with conditioned medium showed a set of receptor genes (CXCR1, IL1RL1, IL1RN, IL3RA, IL8RA, IL11RA, IL15RA, IL17RC, and IL17RD) with significantly different expressions. It is very important to emphasize that we detected significantly higher IL11RA gene expression in primary melanoma tissues with liver metastasis as well, compared to those without metastasis. In addition, we assessed protein expression in endothelial cells before and after co-culturing them with melanoma cell lines by applying chemokine and cytokine proteome arrays. This analysis revealed 15 differentially expressed proteins (including CD31, VCAM-1, ANGPT2, CXCL8, and CCL20) in the hepatic endothelial cells after co-culture with melanoma cells. Our results clearly indicate the interaction between liver endothelial and melanoma cells. Furthermore, we assume that overexpression of the IL11RA gene may play a key role in organ-specific metastasis of primary melanoma cells to the liver.
Collapse
Affiliation(s)
- Viktória Koroknai
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Public Health Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - István Szász
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Public Health Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Margit Balázs
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Public Health Research Group, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
5
|
Huang Y, Zhu C, Liu P, Ouyang F, Luo J, Lu C, Tang B, Yang X. L1CAM promotes vasculogenic mimicry formation by miR-143-3p-induced expression of hexokinase 2 in glioma. Mol Oncol 2023; 17:664-685. [PMID: 36708044 PMCID: PMC10061292 DOI: 10.1002/1878-0261.13384] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 12/17/2022] [Accepted: 01/26/2023] [Indexed: 01/29/2023] Open
Abstract
In recent decades, antiangiogenic therapy, which blocks the supply of oxygen and nutrition to tumor cells, has become a promising clinical strategy for the treatment of patients with tumors. However, recent studies revealed that vasculogenic mimicry (VM), which is the process by which vascular morphological structures are formed by highly invasive tumor cells, has been considered a potential factor for the failure of antiangiogenic therapy in patients with tumors. Thus, inhibition of VM formation might be a potential target for improving the outcome of antiangiogenic strategies. However, the mechanism underlying VM formation is still incompletely elucidated. Herein, we report that L1CAM might be a critical regulator of VM formation in glioma, and might be associated with the resistance of glioma to antiangiogenic therapy. We found that the tumor-invasion and tube-formation capabilities of L1CAM-overexpressing cells were significantly enhanced in vitro and in vivo. In addition, the results indicated that miR-143-3p, which might directly target the 3'UTR of the hexokinase 2 (HK2) gene to regulate its protein expression, was subsequently involved in L1CAM-mediated VM formation by glioma cells. Further study revealed that the regulation of MMP2, MMP9, and VEGFA expression was involved in this process. Moreover, we identified that activation of the downstream PI3K/AKT signaling pathway of the L1CAM/HK2 cascade is critical for VM formation by glioma cells. Furthermore, we found that the combined treatment of anti-L1CAM neutralizing monoclonal antibody and bevacizumab increases efficacy beyond that of bevacizumab alone, and suppresses glioma growth in vivo, indicating that the inhibition of L1CAM-mediated VM formation might efficiently improve the effect of antiangiogenic treatment for glioma patients. Together, our findings demonstrated a critical role of L1CAM in regulating VM formation in glioma, and that L1CAM might be a potential target for ameliorating tumor resistance to antiangiogenic therapy in glioma patients.
Collapse
Affiliation(s)
- Yishan Huang
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular ImmunopathologyShantou University Medical CollegeChina
| | - Chenchen Zhu
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular ImmunopathologyShantou University Medical CollegeChina
| | - Pei Liu
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular ImmunopathologyShantou University Medical CollegeChina
| | - Fan Ouyang
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular ImmunopathologyShantou University Medical CollegeChina
| | - Juanjuan Luo
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular ImmunopathologyShantou University Medical CollegeChina
| | - Chunjiao Lu
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular ImmunopathologyShantou University Medical CollegeChina
| | - Bo Tang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Xiaojun Yang
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular ImmunopathologyShantou University Medical CollegeChina
| |
Collapse
|
6
|
Hu H, Ma T, Liu N, Hong H, Yu L, Lyu D, Meng X, Wang B, Jiang X. Immunotherapy checkpoints in ovarian cancer vasculogenic mimicry: Tumor immune microenvironments, and drugs. Int Immunopharmacol 2022; 111:109116. [PMID: 35969899 DOI: 10.1016/j.intimp.2022.109116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 02/09/2023]
Abstract
Vasculogenic mimicry (VM), a vessel-like structure independent of endothelial cells, commonly exists in solid tumors which requires blood vessels to grow. As a special source of blood supply for tumor progression to a more aggressive state, VM has been observed in a variety of human malignant tumors and is tightly associated with tumor proliferation, invasion, metastasis, and poor patient prognosis. So far, various factors, including immune cells and cytokines, were reported to regulate ovarian cancer progression by influencing VM formation. Herein, we review the mechanisms that regulate VM formation in ovarian cancer and the effect of cells, cytokines, and signaling molecules in the tumor microenvironment on VM formation, Furthermore, we summarize the current clinical application of drugs targeting VM formation.
Collapse
Affiliation(s)
- Haitao Hu
- Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, PR China.
| | - Ting Ma
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Nanqi Liu
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Hong Hong
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, PR China.
| | - Lujiao Yu
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, PR China.
| | - Dantong Lyu
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Xin Meng
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Biao Wang
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning Province, PR China.
| | - Xuefeng Jiang
- Department of Immunology, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| |
Collapse
|
7
|
Bahri R, Kiss O, Prise I, Garcia-Rodriguez KM, Atmoko H, Martínez-Gómez JM, Levesque MP, Dummer R, Smith MP, Wellbrock C, Bulfone-Paus S. Human Melanoma-Associated Mast Cells Display a Distinct Transcriptional Signature Characterized by an Upregulation of the Complement Component 3 That Correlates With Poor Prognosis. Front Immunol 2022; 13:861545. [PMID: 35669782 PMCID: PMC9163391 DOI: 10.3389/fimmu.2022.861545] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
Cutaneous melanoma is one of the most aggressive human malignancies and shows increasing incidence. Mast cells (MCs), long-lived tissue-resident cells that are particularly abundant in human skin where they regulate both innate and adaptive immunity, are associated with melanoma stroma (MAMCs). Thus, MAMCs could impact melanoma development, progression, and metastasis by secreting proteases, pro-angiogenic factors, and both pro-inflammatory and immuno-inhibitory mediators. To interrogate the as-yet poorly characterized role of human MAMCs, we have purified MCs from melanoma skin biopsies and performed RNA-seq analysis. Here, we demonstrate that MAMCs display a unique transcriptome signature defined by the downregulation of the FcεRI signaling pathway, a distinct expression pattern of proteases and pro-angiogenic factors, and a profound upregulation of complement component C3. Furthermore, in melanoma tissue, we observe a significantly increased number of C3+ MCs in stage IV melanoma. Moreover, in patients, C3 expression significantly correlates with the MC-specific marker TPSAB1, and the high expression of both markers is linked with poorer melanoma survival. In vitro, we show that melanoma cell supernatants and tumor microenvironment (TME) mediators such as TGF-β, IL-33, and IL-1β induce some of the changes found in MAMCs and significantly modulate C3 expression and activity in MCs. Taken together, these data suggest that melanoma-secreted cytokines such as TGF-β and IL-1β contribute to the melanoma microenvironment by upregulating C3 expression in MAMCs, thus inducing an MC phenotype switch that negatively impacts melanoma prognosis.
Collapse
Affiliation(s)
- Rajia Bahri
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Orsolya Kiss
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Ian Prise
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Karen M. Garcia-Rodriguez
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Haris Atmoko
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Julia M. Martínez-Gómez
- Department of Dermatology, Skin Cancer Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Mitchell P. Levesque
- Department of Dermatology, Skin Cancer Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, Skin Cancer Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Michael P. Smith
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Claudia Wellbrock
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Silvia Bulfone-Paus
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
8
|
Zhang JY, Du Y, Gong LP, Shao YT, Wen JY, Sun LP, He D, Guo JR, Chen JN, Shao CK. EBV-Induced CXCL8 Upregulation Promotes Vasculogenic Mimicry in Gastric Carcinoma via NF-κB Signaling. Front Cell Infect Microbiol 2022; 12:780416. [PMID: 35321317 PMCID: PMC8936189 DOI: 10.3389/fcimb.2022.780416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 02/09/2022] [Indexed: 12/26/2022] Open
Abstract
Epstein–Barr virus (EBV)-associated gastric carcinoma (EBVaGC) is a distinct entity with a conspicuous tumor microenvironment compared with EBV-negative gastric carcinoma. However, the exact role of EBV in gastric carcinogenesis remains elusive. In the present study, we found that EBV upregulated CXCL8 expression, and CXCL8 significantly promoted vasculogenic mimicry (VM) formation of gastric carcinoma (GC) cells. In accordance with these observations, overexpression of CXCL8 increased cell proliferation and migration of AGS and BGC823 cells, while knockdown of CXCL8 with siRNA inhibited cell proliferation and migration of AGS-EBV cells. In addition, activation of NF-κB signaling was involved in VM formation induced by CXCL8, which was blocked by NF-κB inhibitors BAY 11-7082 and BMS345541. Furthermore, EBV-encoded lncRNA RPMS1 activated the NF-κB signaling cascade, which is responsible for EBV-induced VM formation. Both xenografts and clinical samples of EBVaGC exhibit VM histologically, which are correlated with CXCL8 overexpression. Finally, CXCL8 is positively correlated with overall survival in GC patients. In conclusion, EBV-upregulated CXCL8 expression promotes VM formation in GC via NF-κB signaling, and CXCL8 might serve as a novel anti-tumor target for EBVaGC.
Collapse
Affiliation(s)
- Jing-yue Zhang
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu Du
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-ping Gong
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi-ting Shao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jing-yun Wen
- Department of Medical Oncology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-ping Sun
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dan He
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jin-rui Guo
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jian-ning Chen
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Jian-ning Chen, ; Chun-kui Shao,
| | - Chun-kui Shao
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Jian-ning Chen, ; Chun-kui Shao,
| |
Collapse
|
9
|
Yuan Y, Geng B, Xu X, Zhao H, Bai J, Dou Z, Jia S, Yu X, Luo W. Dual VEGF/PDGF knockdown suppresses vasculogenic mimicry formation in choroidal melanoma cells via the Wnt5a/β-catenin/AKT signaling pathway. Acta Histochem 2022; 124:151842. [PMID: 34995928 DOI: 10.1016/j.acthis.2021.151842] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 12/23/2022]
Abstract
OBJECTIVE This study aimed to explore the effects of knocking down both vascular endothelial growth factor (VEGF) and platelet derived growth factor (PDGF) on vasculogenic mimicry (VM) formation in choroidal melanoma (CM) cells. METHODS Cell counting Kit (CCK)-8, monoclonal formation, wound healing, transwell and flow cytometry assays were used to observe the cell effects in CM cell line, ocular choroidal melanoma-1 cells (OCM-1) with respect to proliferation, migration, invasion and apoptosis. Three-dimensional (3D) cultures were also used to characterize VM tube structural effects in OCM-1 cells and western blotting was used to characterize protein expression changes in VM-related markers. RESULTS Dual VEGF/PDGF knockdown suppressed cell proliferation, migration and invasion, but promoted cell apoptosis. It also reduced VM tube structures in OCM-1 cells. VM associated markers including, VE-cadherin, EphA2 and MT1-MMP were also down-regulated in OCM-1 cells. Similarly, Wnt5a, β-catenin and phosphorylated-AKT levels were also down-regulated. Western blotting and 3D cultures further demonstrated that combined Wnt5a silencing with dual VEGF/PDGF knockdown significantly decreased VE-cadherin and EphA2 levels and reduced VM tube structures in OCM-1 cells. CONCLUSIONS Dual VEGF/PDGF knockdown suppressed cell growth and metastasis in OCM-1 cells, and blocked the Wnt5a/β-catenin/AKT signaling pathway thereby inhibiting VM formation.
Collapse
|
10
|
Massimini M, Romanucci M, De Maria R, Della Salda L. An Update on Molecular Pathways Regulating Vasculogenic Mimicry in Human Osteosarcoma and Their Role in Canine Oncology. Front Vet Sci 2021; 8:722432. [PMID: 34631854 PMCID: PMC8494780 DOI: 10.3389/fvets.2021.722432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/23/2021] [Indexed: 01/16/2023] Open
Abstract
Canine tumors are valuable comparative models for human counterparts, especially to explore novel biomarkers and to understand pathways and processes involved in metastasis. Vasculogenic mimicry (VM) is a unique property of malignant cancer cells which promote metastasis. Thus, it represents an opportunity to investigate both the molecular mechanisms and the therapeutic targets of a crucial phenotypic malignant switch. Although this biological process has been largely investigated in different human cancer types, including osteosarcoma, it is still largely unknown in veterinary pathology, where it has been mainly explored in canine mammary tumors. The presence of VM in human osteosarcoma is associated with poor clinical outcome, reduced patient survival, and increased risk of metastasis and it shares the main pathways involved in other type of human tumors. This review illustrates the main findings concerning the VM process in human osteosarcoma, search for the related current knowledge in canine pathology and oncology, and potential involvement of multiple pathways in VM formation, in order to provide a basis for future investigations on VM in canine tumors.
Collapse
|
11
|
Howard E, Hurrell BP, Helou DG, Quach C, Painter JD, Shafiei-Jahani P, Fung M, Gill PS, Soroosh P, Sharpe AH, Akbari O. PD-1 Blockade on Tumor Microenvironment-Resident ILC2s Promotes TNF-α Production and Restricts Progression of Metastatic Melanoma. Front Immunol 2021; 12:733136. [PMID: 34531874 PMCID: PMC8438316 DOI: 10.3389/fimmu.2021.733136] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/12/2021] [Indexed: 11/13/2022] Open
Abstract
While pulmonary ILC2s represent one of the major tissue-resident innate lymphoid cell populations at steady state and are key drivers of cytokine secretion in their occupational niche, their role in pulmonary cancer progression remains unclear. As the programmed cell death protein-1 (PD-1) plays a major role in cancer immunotherapy and immunoregulatory properties, here we investigate the specific effect of PD-1 inhibition on ILC2s during pulmonary B16 melanoma cancer metastasis. We demonstrate that PD-1 inhibition on ILC2s suppresses B16 tumor growth. Further, PD-1 inhibition upregulates pulmonary ILC2-derived TNF-α production, a cytotoxic cytokine that directly induces cell death in B16 cells, independent of adaptive immunity. Together, these results highlight the importance of ILC2s and their anti-tumor role in pulmonary B16 cancer progression during PD-1 inhibitory immunotherapy.
Collapse
Affiliation(s)
- Emily Howard
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Benjamin P. Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Doumet Georges Helou
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Christine Quach
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jacob D. Painter
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Pedram Shafiei-Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Marshall Fung
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Parkash S. Gill
- Department of Medicine, Norris Cancer center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Pejman Soroosh
- Immunometabolism, Janssen Research and Development, San Diego, CA, United States
| | - Arlene H. Sharpe
- Department of Immunology, Harvard Medical School, Boston, MA, United States
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Medicine, Norris Cancer center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
12
|
Treps L, Faure S, Clere N. Vasculogenic mimicry, a complex and devious process favoring tumorigenesis – Interest in making it a therapeutic target. Pharmacol Ther 2021; 223:107805. [DOI: 10.1016/j.pharmthera.2021.107805] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
13
|
Lin TC, Wang KH, Chuang KH, Kao AP, Kuo TC. Interleukin-33 promotes invasiveness of human ovarian endometriotic stromal cells through the ST2/MAPK/MMP-9 pathway activated by 17β-estradiol. Taiwan J Obstet Gynecol 2021; 60:658-664. [PMID: 34247803 DOI: 10.1016/j.tjog.2021.05.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2021] [Indexed: 01/16/2023] Open
Abstract
OBJECTIVE Endometriosis is an estrogen-dependent, benign, and chronic gynecological disorder occurring in women of reproductive age. Although the pathogenesis of endometriosis is poorly understood, implantation theory indicates that viable endometrial cells shed from the endometrium into the pelvic peritoneum or ovaries, possibly through retrograde menstruation, and then reattach, invade, and damage other tissues. Interleukin (IL)-33, a new member of the IL-1 superfamily, is mainly upregulated by stromal cells following proinflammatory stimulation. Matrix metalloproteinases (MMPs) are involved in the degradation and reconstruction of the extracellular matrix. MMP-9 participates in the pathogenesis of endometriosis by promoting the invasion of endometriotic cells. This study investigated the effect of IL-33 on the cell invasion ability of and MMP-9 expression in human stromal cells derived from ovarian endometrioma (hOVEN-SCs). MATERIALS AND METHODS We isolated hOVEN-SCs from human ovarian endometrioma. Gene expression was analyzed using the Illumina Human WG-6 v2 Expression BeadChips microarray platform and through reverse transcription-polymerase chain reaction. Cell migration and invasion were examined by performing the transwell chamber assay. RESULTS We found that 17β-estradiol could increase the expression of IL-33 and ST2 through the estrogen receptor pathway in hOVEN-SCs. Moreover, IL-33 upregulated MMP-9 expression in and enhanced the invasion ability of hOVEN-SCs through the ST2/MAPK signaling pathway. Our results showed that MMP-9 expression was essential for IL-33-induced cell invasion. CONCLUSION Our main finding is that 17β-estradiol could increase IL-33 expression through the estrogen receptor pathway and activate MMP-9 expression in and invasion ability of hOVEN-SCs through the IL-33/ST2/MAPK signaling pathway. The results of this study and further related studies may provide new strategies for the prevention and treatment of endometriosis.
Collapse
Affiliation(s)
- Ta-Chin Lin
- Department of Obstetrics and Gynecology, Kuo General Hospital, Tainan, Taiwan; Center for Reproductive Medicine, Kuo General Hospital, Tainan, Taiwan
| | - Kai-Hung Wang
- Department of Obstetrics and Gynecology, Kuo General Hospital, Tainan, Taiwan; Center for Reproductive Medicine, Kuo General Hospital, Tainan, Taiwan; Department of Laboratory Medicine, Kuo General Hospital, Tainan, Taiwan.
| | - Kuo-Hsiang Chuang
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - An-Pei Kao
- Stemforce Biotechnology Co., Ltd, Chiayi, Taiwan
| | - Tsung-Cheng Kuo
- Department of Obstetrics and Gynecology, Kuo General Hospital, Tainan, Taiwan; Center for Reproductive Medicine, Kuo General Hospital, Tainan, Taiwan
| |
Collapse
|
14
|
Sun S, Cao C, Li J, Meng Q, Cheng B, Shi B, Shan A. Lycopene Modulates Placental Health and Fetal Development Under High-Fat Diet During Pregnancy of Rats. Mol Nutr Food Res 2021; 65:e2001148. [PMID: 34018317 DOI: 10.1002/mnfr.202001148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 04/09/2021] [Indexed: 01/07/2023]
Abstract
Lycopene plays an important role in improving immunity, promoting antioxidant capacity, and regulating fat metabolism. The placenta, an important organ for nutrients exchange between mother and child during pregnancy, directly affects fetal development. This study aims to characterize effects of lycopene on placental health and fetal development under a high-fat diet, and utilize RNA sequencing (RNA-seq) to investigate and integrate the differences of molecular pathways and biological processes in placenta. For placental health, high-fat diet during pregnancy increases placental oxidative stress, inflammation, and fat deposition. However, lycopene reduces the negative effects of high-fat diet on placenta to some extent, and further promotes fetal development. Under high-fat diet, lycopene reduces the levels of Interleukin 17 (IL-17), Interleukin 6 (IL-6), and tumor necrosis factor α (TNF-α) in placenta (p < 0.05) through the IL-17 pathway. Furthermore, lycopene supplementation in high-fat diet increases Glutaredoxin (Glrx) gene and protein expression in the placenta (p < 0.05), increases Glutathione peroxidase (GSH-Px) and Total antioxidant capacity (T-AOC) levels (p < 0.05), decreases reactive oxygen species (ROS) (p < 0.01) and Hydrogen peroxide (H2 O2 ) levels (p < 0.05) in placenta. In addition, lycopene supplementation in high fat diet increases the expression of Lep gene and protein in placenta and increases the level of leptin (p < 0.05). In terms of fetal development, the average fetal weight and fetal litter weight are increased by lycopene compared to high-diet treatment.
Collapse
Affiliation(s)
- Shishuai Sun
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Chunyu Cao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Jibo Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Qingwei Meng
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Baojing Cheng
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Baoming Shi
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| |
Collapse
|
15
|
Zhang J, Qiao Q, Xu H, Zhou R, Liu X. Human cell polyploidization: The good and the evil. Semin Cancer Biol 2021; 81:54-63. [PMID: 33839294 DOI: 10.1016/j.semcancer.2021.04.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 02/06/2023]
Abstract
Therapeutic resistance represents a major cause of death for most lethal cancers. However, the underlying mechanisms of such resistance have remained unclear. The polyploid cells are due to an increase in DNA content, commonly associated with cell enlargement. In human, they play a variety of roles in physiology and pathologic conditions and perform the specialized functions during development, inflammation, and cancer. Recent work shows that cancer cells can be induced into polyploid giant cancer cells (PGCCs) that leads to reprogramming of surviving cancer cells to acquire resistance. In this article, we will review the polyploidy involved in development and inflammation, and the process of PGCCs formation and propagation that benefits to cell survival. We will discuss the potential opportunities in fighting resistant cancers. The increased knowledge of PGCCs will offer a completely new paradigm to explore the therapeutic intervention for lethal cancers.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Qing Qiao
- Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Hong Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Ru Zhou
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xinzhe Liu
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
16
|
Wang H, Wang L, Zheng Q, Lu Z, Chen Y, Shen D, Xue D, Jiang M, Ding L, Zhang J, Wu H, Xia L, Qian J, Li G, Lu J. Oncometabolite L-2-hydroxyglurate directly induces vasculogenic mimicry through PHLDB2 in renal cell carcinoma. Int J Cancer 2021; 148:1743-1755. [PMID: 33320958 PMCID: PMC7986127 DOI: 10.1002/ijc.33435] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/19/2020] [Accepted: 11/27/2020] [Indexed: 12/16/2022]
Abstract
Metabolism reprograming is a hallmark of cancer and plays an important role in tumor progression. The aberrant metabolism in renal cell carcinoma (RCC) leads to accumulation of the oncometabolite l‐2‐hydroxyglurate (L‐2HG). L‐2HG has been reported to inhibit the activity of some α‐ketoglutarate‐dependent dioxygenases such as TET enzymes, which mediate epigenetic alteration, including DNA and histone demethylation. However, the detailed functions of L‐2HG in renal cell carcinoma have not been investigated thoroughly. In our study, we found that L‐2HG was significantly elevated in tumor tissues compared to adjacent tissues. Furthermore, we demonstrated that L‐2HG promoted vasculogenic mimicry (VM) in renal cancer cell lines through reducing the expression of PHLDB2. A mechanism study revealed that activation of the ERK1/2 pathway was involved in L‐2HG‐induced VM formation. In conclusion, these findings highlighted the pathogenic link between L‐2HG and VM and suggested a novel therapeutic target for RCC. What's new? Metabolic reprograming, a hallmark of cancer, influences tumor progression. In the case of renal cell carcinoma (RCC) specifically, progression appears to be facilitated by the oncometabolite L‐2‐hydroxyglurate (L‐2HG), though underlying mechanisms remain enigmatic. Here, the authors investigated the ability of L‐2HG in RCC to promote vasculogenic mimicry (VM), in which aggressive cancer cells form vessel‐like networks that support tumor growth. Analyses of RCC patient tissues revealed elevated L‐2HG levels, wherein tumor cells with greater L‐2HG levels exhibited more VM structures. TCGA data and high‐throughput sequencing analyses further show that L‐2HG contributes to VM formation via reduction of PHLDB2 levels.
Collapse
Affiliation(s)
- Huan Wang
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liya Wang
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiming Zheng
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zeyi Lu
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuanlei Chen
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Danyang Shen
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dingwei Xue
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minxiao Jiang
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lifeng Ding
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Zhang
- Department of Urology, The Affiliated Hangzhou First People's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Haiyang Wu
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liqun Xia
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Qian
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,State Key Laboratory of Modern Optical Instrumentations, Centre for Optical and Electromagnetic Research, College of Optical Science and Engineering, Zhejiang University, Hangzhou, China
| | - Gonghui Li
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jieyang Lu
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
17
|
Fares J, Cordero A, Kanojia D, Lesniak MS. The Network of Cytokines in Brain Metastases. Cancers (Basel) 2021; 13:E142. [PMID: 33466236 PMCID: PMC7795138 DOI: 10.3390/cancers13010142] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 12/20/2022] Open
Abstract
Brain metastases are the most common of all intracranial tumors and a major cause of death in patients with cancer. Cytokines, including chemokines, interferons, interleukins, lymphokines, and tumor necrosis factors are key regulators in the formation of brain metastases. They regulate the infiltration of different cellular subsets into the tumor microenvironment and affect the therapeutic outcomes in patients. Elucidating the cancer cell-cytokine interactions in the setting of brain metastases is crucial for the development of more accurate diagnostics and efficacious therapies. In this review, we focus on cytokines that are found in the tumor microenvironment of brain metastases and elaborate on their trends of expression, regulation, and roles in cellular recruitment and tumorigenesis. We also explore how cytokines can alter the anti-tumor response in the context of brain metastases and discuss ways through which cytokine networks can be manipulated for diagnosis and treatment.
Collapse
Affiliation(s)
| | | | | | - Maciej S. Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (J.F.); (A.C.); (D.K.)
| |
Collapse
|
18
|
Li S, Wang Z, Liu X, Li Y, Shi C, Wu J, Sun S, Li Y, Li S, Xu Y, Song B. Association of Common Variants in the IL-33/ST2 Axis with Ischemic Stroke. Curr Neurovasc Res 2020; 16:494-501. [PMID: 31660817 DOI: 10.2174/1567202616666191029112334] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/09/2019] [Accepted: 10/16/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND Recent studies have reported that the levels of serum interleukin-33 (IL- 33) and its receptor, suppression of tumorigenicity 2 (ST2), are potential biomarkers for susceptibility of cardiovascular diseases. However, the genetic association of the IL-33/ST2 axis with cardiovascular diseases remains controversial. OBJECTIVE We aimed to investigate the association between common variants in the IL-33/ST2 axis and ischemic stroke in the Han Chinese population. METHODS We consecutively enrolled 1166 patients with ischemic stroke and 1079 age- and gender- matched controls. Eight single nucleotide polymorphisms (SNPs) within IL-33/ST2 axis were genotyped using the improved Multiple Ligase Detection Reaction platform. We analyzed the association between the tested SNPs and ischemic stroke at both the genotype and haplotype levels. RESULTS Binary logistic regression analysis indicated that rs10435816 (additive model: odds ratio [OR]=0.72, 95% confidence interval [CI], 0.54-0.95; recessive model: OR=0.72, 95%CI, 0.56- 0.94) was associated with a decreased risk of ischemic stroke after adjustment of confounding factors. Subgroup analysis indicated that rs10435816 (additive model: OR=0.61, 95%CI, 0.41-0.89; recessive model: OR=0.56, 95%CI, 0.40-0.80), rs7025417 (additive model: OR=0.57, 95%CI, 0.39-0.83), rs11792633 (additive model: OR=0.66, 95%CI, 0.46-0.95; recessive model: OR=0.67, 95%CI, 0.49-0.93), and rs7044343 (additive model: OR=0.69, 95%CI, 0.48-0.97; recessive model: OR=0.67, 95%CI, 0.49-0.91) were associated with a decreased risk of large-artery atherosclerosis stroke after adjustment of confounding factors. CONCLUSION Our findings suggested an association between common variants in the IL-33/ST2 axis and a decreased risk of ischemic stroke in the Han Chinese population.
Collapse
Affiliation(s)
- Shuo Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhijie Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xinjing Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yuanzhe Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jun Wu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Shilei Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yusheng Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Shaohua Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Bo Song
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
19
|
Abstract
All organisms growing beyond the oxygen diffusion limit critically depend on a functional vasculature for survival. Yet blood vessels are far more than passive, uniform conduits for oxygen and nutrient supply. A remarkable organotypic heterogeneity is brought about by tissue-specific differentiated endothelial cells (lining the blood vessels' lumen) and allows blood vessels to deal with organ-specific demands for homeostasis. On the flip side, when blood vessels go awry, they promote life-threatening diseases characterized by endothelial cells inappropriately adopting an angiogenic state (eg, tumor vascularization) or becoming dysfunctional (eg, diabetic microvasculopathies), calling respectively for antiangiogenic therapies and proangiogenic/vascular regenerative strategies. In solid tumors, despite initial enthusiasm, growth factor-based (mostly anti-VEGF [vascular endothelial growth factor]) antiangiogenic therapies do not sufficiently live up to the expectations in terms of efficiency and patient survival, in part, due to intrinsic and acquired therapy resistance. Tumors cunningly deploy alternative growth factors than the ones targeted by the antiangiogenic therapies to reinstigate angiogenesis or revert to other ways of securing blood flow, independently of the targeted growth factors. In trying to alleviate tissue ischemia and to repair dysfunctional or damaged endothelium, local in-tissue administration of (genes encoding) proangiogenic factors or endothelial (stem) cells harnessing regenerative potential have been explored. Notwithstanding evaluation in clinical trials, these approaches are often hampered by dosing issues and limited half-life or local retention of the administered agents. Here, without intending to provide an all-encompassing historical overview, we focus on some recent advances in understanding endothelial cell behavior in health and disease and identify novel molecular players and concepts that could eventually be considered for therapeutic targeting.
Collapse
Affiliation(s)
- Guy Eelen
- From the Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Belgium (G.E., L.T., P.C.)
| | - Lucas Treps
- From the Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Belgium (G.E., L.T., P.C.)
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China (X.L., P.C.)
| | - Peter Carmeliet
- From the Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Belgium (G.E., L.T., P.C.).,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China (X.L., P.C.)
| |
Collapse
|
20
|
Quintero-Fabián S, Arreola R, Becerril-Villanueva E, Torres-Romero JC, Arana-Argáez V, Lara-Riegos J, Ramírez-Camacho MA, Alvarez-Sánchez ME. Role of Matrix Metalloproteinases in Angiogenesis and Cancer. Front Oncol 2019; 9:1370. [PMID: 31921634 PMCID: PMC6915110 DOI: 10.3389/fonc.2019.01370] [Citation(s) in RCA: 567] [Impact Index Per Article: 94.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 11/20/2019] [Indexed: 12/15/2022] Open
Abstract
During angiogenesis, new vessels emerge from existing endothelial lined vessels to promote the degradation of the vascular basement membrane and remodel the extracellular matrix (ECM), followed by endothelial cell migration, and proliferation and the new generation of matrix components. Matrix metalloproteinases (MMPs) participate in the disruption, tumor neovascularization, and subsequent metastasis while tissue inhibitors of metalloproteinases (TIMPs) downregulate the activity of these MMPs. Then, the angiogenic response can be directly or indirectly mediated by MMPs through the modulation of the balance between pro- and anti-angiogenic factors. This review analyzes recent knowledge on MMPs and their participation in angiogenesis.
Collapse
Affiliation(s)
- Saray Quintero-Fabián
- Multidisciplinary Research Laboratory, Military School of Graduate of Health, Mexico City, Mexico
| | - Rodrigo Arreola
- Psychiatric Genetics Department, National Institute of Psychiatry "Ramón de la Fuente", Clinical Research Branch, Mexico City, Mexico
| | | | - Julio César Torres-Romero
- Biochemistry and Molecular Genetics Laboratory, Facultad de Química de la Universidad Autónoma de Yucatán, Merida, Mexico
| | - Victor Arana-Argáez
- Pharmacology Laboratory, Facultad de Química de la Universidad Autónoma de Yucatán, Mérida, Mexico
| | - Julio Lara-Riegos
- Biochemistry and Molecular Genetics Laboratory, Facultad de Química de la Universidad Autónoma de Yucatán, Merida, Mexico
| | | | | |
Collapse
|