1
|
Bamias G, Menghini P, Pizarro TT, Cominelli F. Targeting TL1A and DR3: the new frontier of anti-cytokine therapy in IBD. Gut 2024:gutjnl-2024-332504. [PMID: 39266053 DOI: 10.1136/gutjnl-2024-332504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/20/2024] [Indexed: 09/14/2024]
Abstract
TNF-like cytokine 1A (TL1A) and its functional receptor, death-domain receptor 3 (DR3), are members of the TNF and TNFR superfamilies, respectively, with recognised roles in regulating innate and adaptive immune responses; additional existence of a decoy receptor, DcR3, indicates a tightly regulated cytokine system. The significance of TL1A:DR3 signalling in the pathogenesis of inflammatory bowel disease (IBD) is supported by several converging lines of evidence. Herein, we aim to provide a comprehensive understanding of what is currently known regarding the TL1A/DR3 system in the context of IBD. TL1A and DR3 are expressed by cellular subsets with important roles for the initiation and maintenance of intestinal inflammation, serving as potent universal costimulators of effector immune responses, indicating their participation in the pathogenesis of IBD. Recent evidence also supports a homoeostatic role for TL1A:DR3 via regulation of Tregs and innate lymphoid cells. TL1A and DR3 are also expressed by stromal cells and may contribute to inflammation-induced or inflammation-independent intestinal fibrogenesis. Finally, discovery of genetic polymorphisms with functional consequences may allow for patient stratification, including differential responses to TL1A-targeted therapeutics. In conclusion, TL1A:DR3 signalling plays a central and multifaceted role in the immunological pathways that underlie intestinal inflammation, such as that observed in IBD. Such evidence provides the foundation for developing pharmaceutical approaches targeting this ligand-receptor pair in IBD.
Collapse
Affiliation(s)
- Giorgos Bamias
- GI Unit, 3rd Academic Department of Internal Medicine, National and Kapodistrian University of Athens, Sotiria Hospital, Athens, Greece
| | - Paola Menghini
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Theresa T Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Fabio Cominelli
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
2
|
Yang H, Xu G, Li Q, Zhu L. Ligustrazine alleviates the progression of coronary artery calcification by inhibiting caspase-3/GSDME mediated pyroptosis. Biosci Trends 2024:2024.01096. [PMID: 38972749 DOI: 10.5582/bst.2024.01096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
Coronary artery calcification (CAC) is an early marker for atherosclerosis and is mainly induced by the osteoblast-like phenotype conversion of vascular smooth muscle cells (VSMCs). Recent reports indicate that NOD-like receptor protein 3 (NLRP3)-mediated pyroptosis plays a significant role in the calcification of vascular smooth muscle cells (VSMCs), making it a promising target for treating calcific aortic valve disease (CAC). Ligustrazine, or tetramethylpyrazine (TMP), has been found effective in various cardiovascular and cerebrovascular diseases and is suggested to inhibit NLRP3-mediated pyroptosis. However, the function of TMP in CAC is unknown. Herein, influences of TMP on β-glycerophosphate (β-GP)-stimulated VSMCs and OPG-/- mice were explored. Mouse Aortic Vascular Smooth Muscle (MOVAS-1) cells were stimulated by β-GP with si- caspase-3, si- Gasdermin E (GSDME) or TMP. Increased calcification, reactive oxygen species (ROS) level, Interleukin-1beta (IL-1β) and Interleukin-18 (IL-18) levels, lactate dehydrogenase (LDH) release, enhanced apoptosis, and activated cysteine-aspartic acid protease-3 (caspase-3)/GSDME signaling were observed in β-GP-stimulated MOVAS-1 cells, which was sharply alleviated by si-caspase-3, si-GSDME or TMP. Furthermore, the impact of TMP on the β-GP-induced calcification and injury in MOVAS-1 cells was abolished by raptinal, an activator of caspase-3. Subsequently, OPG-/- mice were dosed with TMP or TMP combined with raptinal. Calcium deposition, increased nodules, elevated IL-1β and IL-18 levels, upregulated CASP3 and actin alpha 2, smooth muscle (ACTA2), and activated caspase-3/GSDME signaling in OPG-/- mice were markedly alleviated by TMP, which were notably reversed by the co-administration of raptinal. Collectively, TMP mitigated CAC by inhibiting caspase-3/GSDME mediated pyroptosis.
Collapse
Affiliation(s)
- Honghui Yang
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou, China
| | - Guian Xu
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou, China
| | - Qingman Li
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou, China
| | - Lijie Zhu
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou, China
| |
Collapse
|
3
|
Zhan RR, Wang D, Zhang XL. Progress in research of TNF-like cytokine 1A as a therapeutic target for inflammatory bowel disease. WORLD CHINESE JOURNAL OF DIGESTOLOGY 2024; 32:397-404. [DOI: 10.11569/wcjd.v32.i6.397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
|
4
|
Mignini I, Blasi V, Termite F, Esposto G, Borriello R, Laterza L, Scaldaferri F, Ainora ME, Gasbarrini A, Zocco MA. Fibrostenosing Crohn's Disease: Pathogenetic Mechanisms and New Therapeutic Horizons. Int J Mol Sci 2024; 25:6326. [PMID: 38928032 PMCID: PMC11204249 DOI: 10.3390/ijms25126326] [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: 04/30/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Bowel strictures are well recognized as one of the most severe complications in Crohn's disease, with variable impacts on the prognosis and often needing surgical or endoscopic treatment. Distinguishing inflammatory strictures from fibrotic ones is of primary importance due to the different therapeutic approaches required. Indeed, to better understand the pathogenesis of fibrosis, it is crucial to investigate molecular processes involving genetic factors, cytokines, alteration of the intestinal barrier, and epithelial and endothelial damage, leading to an increase in extracellular matrix synthesis, which ultimately ends in fibrosis. In such a complex mechanism, the gut microbiota also seems to play a role. A better comprehension of molecular processes underlying bowel fibrosis, in addition to radiological and histopathological findings, has led to the identification of high-risk patients for personalized follow-up and testing of new therapies, primarily in preclinical models, targeting specific pathways involving Transforming Growth Factor-β, interleukins, extracellular matrix balance, and gut microbiota. Our review aims to summarize current evidence about molecular factors involved in intestinal fibrosis' pathogenesis, paving the way for potential diagnostic biomarkers or anti-fibrotic treatments for stricturing Crohn's disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Maria Assunta Zocco
- CEMAD Digestive Diseases Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (I.M.); (V.B.); (G.E.); (R.B.); (L.L.); (F.S.); (M.E.A.); (A.G.)
| |
Collapse
|
5
|
Zhou M, Yao Y, Ma S, Zou M, Chen Y, Cai S, Zhao F, Wu H, Xiao F, Abudushalamu G, Fan X, Wu G. Dual-targeted and dual-sensitive self-assembled protein nanocarrier delivering hVEGI-192 for triple-negative breast cancer. Int J Biol Macromol 2023:125475. [PMID: 37353129 DOI: 10.1016/j.ijbiomac.2023.125475] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/11/2023] [Accepted: 06/16/2023] [Indexed: 06/25/2023]
Abstract
Breast cancer is a highly prevalent malignancy worldwide among women with an increasing incidence in recent years. Triple-negative breast cancer (TNBC), a specific type of breast cancer, occurs primarily in young women and exhibits large tumor size, high clinical stage, and extremely poor prognosis with a high rate of lymph node, liver, and lung metastases. TNBC is insensitive to endocrine therapy and trastuzumab treatment, and there is an urgent need for effective therapeutics and treatment guidelines. However, investigations into antiangiogenic agents for the treatment of TNBC are ongoing. In this study, we successfully engineered a self-assembled protein nanocarrier TfRBP9-hVEGI-192-ELP fusion protein (TVEFP) to deliver the therapeutic protein, human vascular endothelial growth inhibitor (hVEGI-192). This was found to be effective in inhibiting tumor angiogenesis in vivo. The protein nanocarrier effectively inhibited the progression of TNBC in vivo and showed the behavior of self-assembly, thermoresponsiveness, enzyme stimulation-responsiveness, tumor-targeting, biocompatibility, and biodegradability. Near-infrared imaging studies showed that fluorescent dye-stained TVEFP effectively aggregated at the tumor site. The TVEFP nanocarrier significantly expands the application of the therapeutic protein hVEGI-192 and improves the imaging and biotherapeutic effects in TNBC, chiefly based on anti-angiogenesis effects.
Collapse
Affiliation(s)
- Meiling Zhou
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yuming Yao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Shuo Ma
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Mingyuan Zou
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yaya Chen
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Shijie Cai
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Fengfeng Zhao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Huina Wu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Feng Xiao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - GuliNazhaer Abudushalamu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Xiaobo Fan
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China.
| | - Guoqiu Wu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Laboratory Medcine, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China.
| |
Collapse
|
6
|
Chen K, Xiu Q, Min Q, Cheng X, Xiao H, Jia Z, Feng J, Shi Y, Zhuo Q, Wang J, Zou J. TL1A induces apoptosis via DR3 in grass carp (Ctenopharyngodon idella). FISH AND SHELLFISH IMMUNOLOGY REPORTS 2023; 4:100090. [PMID: 36970231 PMCID: PMC10033717 DOI: 10.1016/j.fsirep.2023.100090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/09/2023] [Accepted: 02/09/2023] [Indexed: 03/17/2023] Open
Abstract
Tumor necrosis factor like ligand 1A (TL1A), a member of TNF superfamily, regulates inflammatory response and immune defense. TL1A homologues have recently been discovered in fish, but their functions have not been studied. In this study, a TL1A homologue was identified in grass carp (Ctenopharyngodon idella) and its bioactivities were investigated. The grass carp tl1a (Citl1a) gene was constitutively expressed in tissues, with the highest expression detected in the liver. It was upregulated in response to infection with Aeromonas hydrophila. The recombinant CiTL1A was produced in bacteria and was shown to stimulate the expression of il1β, tnfα, caspase 8 and ifnγ in the primary head kidney leucocytes. In addition, co-immunoprecipitation assay revealed that CiTL1A interacted with DR3 and induced apoptosis via activation of DR3. The results demonstrate that TL1A regulates inflammation and apoptosis and is involved in the immune defense against bacterial infection in fish.
Collapse
|
7
|
Wen X, Hu G, Xiao X, Zhang X, Zhang Q, Guo H, Li X, Liu Q, Li H. FGF2 positively regulates osteoclastogenesis via activating the ERK-CREB pathway. Arch Biochem Biophys 2022; 727:109348. [PMID: 35835230 DOI: 10.1016/j.abb.2022.109348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/17/2022] [Accepted: 07/03/2022] [Indexed: 11/02/2022]
Abstract
Fibroblast growth factor 2 (FGF2) plays crucial roles in the growth and development of several tissues. However, its function in bone homeostasis remains controversial. Here, we found that exogenous FGF2 supplementation inhibited the mineralization of bone marrow stromal cells (BMSCs), at least partially, via up-regulating the gene expression of osteoclastogenesis. The FGF receptor (FGFR) allosteric antagonist SSR128129E modestly, whereas the FGFR tyrosine kinase inhibitor AZD4547 significantly antagonized the effects of FGF2. Mechanistically, FGF2 stimulated ERK phosphorylation, and the ERK signaling inhibitor PD325901 strongly blocked FGF2 enhancement of osteoclastogenesis. Moreover, the phosphorylation of CREB was also activated in response to FGF2, thereby potentiating the interaction of p-CREB with the promoter region of Rankl gene. Notably, FGF2-deficient BMSCs exhibited higher mineralization capability and lower osteoclastogenic gene expression. Correspondingly, FGF2-knockout mice showed increased bone mass and attenuated expression of osteoclast-related markers, which were associated with moderate inhibition of the ERK signaling. In conclusion, FGF2 positively regulates osteoclastogenesis via stimulating the ERK-CREB pathway. These findings establish the importance of FGF2 in bone homeostasis, hinting the potential use of FGF2/ERK/CREB specific inhibitors to fight against bone-related disorders, such as osteoporosis.
Collapse
Affiliation(s)
- Xin Wen
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Geng Hu
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, China
| | - Xue Xiao
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Xinzhi Zhang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Qiang Zhang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Hengjun Guo
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Xianyao Li
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, China
| | - Qingxin Liu
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China.
| | - Haifang Li
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China.
| |
Collapse
|
8
|
Abstract
Since the receptor activator of nuclear factor-kappa B ligand (RANKL), its cognate receptor activator of nuclear factor-kappa B (RANK), and the decoy receptor osteoprotegerin (OPG) were discovered, a number of studies have uncovered the crucial role of the RANKL-RANK-OPG pathway in controlling the key aspect of bone homeostasis, the immune system, inflammation, cancer, and other systems under pathophysiological condition. These findings have expanded the understanding of the multifunctional biology of the RANKL-RANK-OPG pathway and led to the development of therapeutic potential targeting this pathway. The successful development and application of anti-RANKL antibody in treating diseases causing bone loss validates the utility of therapeutic approaches based on the modulation of this pathway. Moreover, recent studies have demonstrated the involvement of the RANKL-RANK pathway in osteoblast differentiation and bone formation, shedding light on the RANKL-RANK dual signaling in coupling bone resorption and bone formation. In this review, we will summarize the current understanding of the RANKL-RANK-OPG system in the context of the bone and the immune system as well as the impact of this pathway in disease conditions, including cancer development and metastasis.
Collapse
Affiliation(s)
- Noriko Takegahara
- Departments of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Hyunsoo Kim
- Departments of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Yongwon Choi
- Departments of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Tao R, Liu Q, Huang R, Wang K, Sun Z, Yang P, Wang J. A Novel TNFSF-Based Signature Predicts the Prognosis and Immunosuppressive Status of Lower-Grade Glioma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3194996. [PMID: 35592520 PMCID: PMC9112166 DOI: 10.1155/2022/3194996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 02/22/2022] [Accepted: 03/03/2022] [Indexed: 02/08/2023]
Abstract
Purpose Tumour necrosis factor (TNF) superfamilies play important roles in cell proliferation, migration, differentiation, and apoptosis. We believe that TNF has a huge potential and might cast new insight into antitumour therapies. Therefore, we established this signature based on TNF superfamilies. Results A six-gene signature derived from the TNF superfamilies was established. The Riskscore correlated significantly with the expression of immune checkpoint genes and infiltrating M2 macrophages in the tumour specimen. This signature was also associated with mutations in genes that regulate tumour cell proliferation. Univariate and multivariate regression analyses further confirmed the Riskscore, TNFRSF11b, and TNFRSF12a as independent risk factors in The Cancer Genome Atlas and Chinese Glioma Genome Atlas datasets. Conclusion Our signature could accurately predict the prognosis of lower-grade gliomas (LGG). In addition, this six-gene signature could predict the immunosuppressive status of LGG and provide evidence that TNF superfamilies had correlations with some critical mutations that could be effectively targeted now.
Collapse
Affiliation(s)
- Rui Tao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qi Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ruoyu Huang
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Kuanyu Wang
- Gamma Knife Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhiyan Sun
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Pei Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiangfei Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
The essential anti-angiogenic strategies in cartilage engineering and osteoarthritic cartilage repair. Cell Mol Life Sci 2022; 79:71. [PMID: 35029764 PMCID: PMC9805356 DOI: 10.1007/s00018-021-04105-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/01/2021] [Accepted: 12/18/2021] [Indexed: 01/16/2023]
Abstract
In the cartilage matrix, complex interactions occur between angiogenic and anti-angiogenic components, growth factors, and environmental stressors to maintain a proper cartilage phenotype that allows for effective load bearing and force distribution. However, as seen in both degenerative disease and tissue engineering, cartilage can lose its vascular resistance. This vascularization then leads to matrix breakdown, chondrocyte apoptosis, and ossification. Research has shown that articular cartilage inflammation leads to compromised joint function and decreased clinical potential for regeneration. Unfortunately, few articles comprehensively summarize what we have learned from previous investigations. In this review, we summarize our current understanding of the factors that stabilize chondrocytes to prevent terminal differentiation and applications of these factors to rescue the cartilage phenotype during cartilage engineering and osteoarthritis treatment. Inhibiting vascularization will allow for enhanced phenotypic stability so that we are able to develop more stable implants for cartilage repair and regeneration.
Collapse
|
11
|
Furfaro F, Alfarone L, Gilardi D, Correale C, Allocca M, Fiorino G, Argollo M, Zilli A, Zacharopoulou E, Loy L, Roda G, Danese S. TL1A: A New Potential Target in the Treatment of Inflammatory Bowel Disease. Curr Drug Targets 2021; 22:760-769. [PMID: 33475057 DOI: 10.2174/1389450122999210120205607] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/05/2020] [Accepted: 08/27/2020] [Indexed: 11/22/2022]
Abstract
Inflammatory bowel diseases (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), are chronic inflammatory diseases of the gastrointestinal tract. In the last few years, the development of biological agents targeting cytokines and receptors involved in IBD pathogenesis has led to better outcomes and has improved the course of the disease. Despite their effectiveness, drugs such as tumor necrosis factor (TNF) inhibitors, anti-Interleukin-12/23 and anti-integrins, do not induce a response in about one-third of patients, and 40% of patients lose response over time. Therefore, more efficient therapies are required. Recent studies showed that TL1A (Tumor necrosis factor-like cytokine 1A) acts as a regulator of mucosal immunity and participates in immunological pathways involved in the IBD pathogenesis. In this review article, we analyze the role of TL1A as a new potential target therapy in IBD patients.
Collapse
Affiliation(s)
- Federica Furfaro
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Ludovico Alfarone
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Daniela Gilardi
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Carmen Correale
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Mariangela Allocca
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Gionata Fiorino
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | | | - Alessandra Zilli
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Eirini Zacharopoulou
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Laura Loy
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Giulia Roda
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Silvio Danese
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| |
Collapse
|
12
|
Zhao C, Wang D, Wu M, Luo Y, Yang M, Guo J, Zhang H, Zhang X. Tumor necrosis factor ligand-related molecule 1A affects the intestinal mucosal barrier function by promoting Th9/interleukin-9 expression. J Int Med Res 2021; 48:300060520926011. [PMID: 32567429 PMCID: PMC7309405 DOI: 10.1177/0300060520926011] [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] [Indexed: 01/15/2023] Open
Abstract
Objectives To investigate the effect of tumor necrosis factor ligand-related molecule 1A (TL1A) on the intestinal mucosal barrier in mice with chronic colitis. Methods Male TL1A-overexpressing transgenic mice and male C57BL/6 wild-type mice were used to establish a dextran sodium sulfate (DSS)-induced colitis model. The expression of occludin and claudin-1 was observed. Bacterial distribution in the intestinal mucosa and Th9/interleukin (IL)-9 expression were detected. In vitro co-culture systems of naive CD4+ T cells and Caco-2 cells were established and TL1A was added. Changes in transepithelial electrical resistance and IL-9 expression were measured. CD4+IL-9 cells were detected by flow cytometry. Results DSS mice showed a significant down-regulation of occludin and claudin-1 compared with controls. Expression levels of occludin, zonulin-1, and claudin-1 in the Caco-2+TGF-β+IL-4+TL1A group were significantly lower than in the Caco-2+TGF-β+IL-4 group. Bacterial distribution was clearly disordered in the DSS group. Transmembrane resistance of the Caco-2+TGF-β+IL-4+TL1A group was significantly lower and IL-9 expression significantly higher than in the Caco-2+TGF-β+IL-4 group. Conclusions TL1A overexpression promotes destruction of the intestinal mucosal barrier in mice with chronic colitis. The underlying mechanism may be associated with the promoting role of TL1A in Th9/IL-9 expression, which further destroys the mucosal barrier.
Collapse
Affiliation(s)
- Caihong Zhao
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China.,Department of Gastroenterology, Harrison International Peace Hospital, Hengshui, Hebei, China
| | - Dong Wang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Mengyao Wu
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Yuxin Luo
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Mingyue Yang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Jinbo Guo
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Hong Zhang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Xiaolan Zhang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China
| |
Collapse
|
13
|
Ming J, Cronin SJF, Penninger JM. Targeting the RANKL/RANK/OPG Axis for Cancer Therapy. Front Oncol 2020; 10:1283. [PMID: 32850393 PMCID: PMC7426519 DOI: 10.3389/fonc.2020.01283] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
RANKL and RANK are expressed in different cell types and tissues throughout the body. They were originally described for their essential roles in bone remodeling and the immune system but have subsequently been shown to provide essential signals from regulating mammary gland homeostasis during pregnancy to modulating tumorigenesis. The success of RANKL/RANK research serves as a paragon for translational research from the laboratory to the bedside. The case in point has been the development of Denosumab, a RANKL-blocking monoclonal antibody which has already helped millions of patients suffering from post-menopausal osteoporosis and skeletal related events in cancer. Here we will provide an overview of the pathway from its origins to its clinical relevance in disease, with a special focus on emerging evidence demonstrating the therapeutic value of targeting the RANKL/RANK/OPG axis not only in breast cancer but also as an addition to the cancer immunotherapy arsenal.
Collapse
Affiliation(s)
- Jie Ming
- Department of Breast and Thyroid Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shane J F Cronin
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna Biocenter, Vienna, Austria
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna Biocenter, Vienna, Austria.,Department of Medical Genetics, Life Science Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
14
|
Wang K, Hackney JR, Siegal GP, Wei S. RANKLed by the Complexity of Signaling in Breast Cancer Metastasis to the Brain. Clin Breast Cancer 2020; 20:e569-e575. [PMID: 32381383 DOI: 10.1016/j.clbc.2020.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 03/17/2020] [Accepted: 04/01/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND Receptor activator of nuclear factor κB (RANK) and its ligand, RANKL, are essential for mammary gland development and play a vital role in breast carcinogenesis. RANKL-RANK signaling also drives thermoregulation and modulates inflammatory activation in the brain. The expression of RANKL in primary breast cancer (BC) has been negatively associated with brain metastases, while significantly higher levels of RANK are seen in BC with brain metastases. We examined the expression of RANK and RANKL in BC metastasis to the brain. PATIENTS AND METHODS We examined the expression of RANK and RANKL in 40 cases of BC metastasis to the brain. RESULTS RANK was variably expressed in BC cells but minimally expressed in the adjacent brain parenchyma. In contrast, the expression of RANKL was minimal in metastatic BC but highly variable in tumoral stroma. RANKL expression in normal brain stroma obtained during autopsy was negligible. Histologic grade and BC subtypes were not significantly associated with RANK expression in metastatic BC. A significant negative correlation between RANK in metastatic BC and RANKL in tumoral stroma was identified (P < .001). CONCLUSION RANK expressed by primary BC and RANKL detected in the tumor microenvironment together participate in cancer development, while the same principle may operate at distant sites. Further investigation is necessary to provide additional insight into the role of the RANKL-RANK pathway in BC progression and to investigate the potential efficacy of therapeutic strategies targeting these molecules in BC metastasis to the brain.
Collapse
Affiliation(s)
- Kai Wang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - James R Hackney
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - Gene P Siegal
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - Shi Wei
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL.
| |
Collapse
|
15
|
Vascular Endothelial Growth Inhibitor, a Cytokine of the Tumor Necrosis Factor Family, is Associated With Epithelial-Mesenchymal Transition in Renal Cell Carcinoma. Appl Immunohistochem Mol Morphol 2019; 26:727-733. [PMID: 28362712 DOI: 10.1097/pai.0000000000000517] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Previous studies have revealed that the activation of the epithelial-mesenchymal transition (EMT) endows metastatic properties upon cancer cells to promote invasion and migration. In this study, immunohistochemical analysis was performed in 50 cases of clear cell renal cell carcinoma (RCC) and paired normal kidney tissues. We detected the expression of vascular endothelial growth inhibitor (VEGI) and EMT markers (E-cadherin, fibronectin, and Slug) and recorded the clinical, pathologic, and follow-up (median follow-up: 79.0 mo) information. The expression of VEGI and E-cadherin was significantly lower in RCC tissues compared with normal kidney tissues (P<0.001). However, the expression of fibronectin and Slug was higher in RCC tissues (P<0.05). VEGI and EMT marker expression marginally differed in tumor size and stage. Significant differences were found in the pathologic grade (P<0.05). The Spearman correlation analysis suggested a positive correlation between VEGI and E-cadherin (r=0.451, P<0.01). A negative correlation was shown between VEGI and fibronectin (r=-0.465, P<0.01). There was also a negative correlation between VEGI and Slug (r=-0.758, P<0.01). During the 79.0 months (range, 7 to 119 mo) of follow-up, 6 patients died due to RCC, and the tumor-free survival rate was 88% (44/50). We did not find a significant correlation between VEGI/EMT markers (E-cadherin, fibronectin, and Slug) and overall survival (P>0.05). Our findings indicate that VEGI plays an important role in EMT in RCC. It suggests that VEGI may be investigated as a disease biomarker and therapeutic target in RCC.
Collapse
|
16
|
Kumanishi S, Yamanegi K, Nishiura H, Fujihara Y, Kobayashi K, Nakasho K, Futani H, Yoshiya S. Epigenetic modulators hydralazine and sodium valproate act synergistically in VEGI-mediated anti-angiogenesis and VEGF interference in human osteosarcoma and vascular endothelial cells. Int J Oncol 2019; 55:167-178. [PMID: 31180533 DOI: 10.3892/ijo.2019.4811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 05/14/2019] [Indexed: 11/06/2022] Open
Abstract
Vascular endothelial growth inhibitor (VEGI; also referred to as TNFSF15 or TL1A) is involved in the modulation of vascular homeostasis. VEGI is known to operate via two receptors: Death receptor‑3 (DR3) and decoy receptor‑3 (DcR3). DR3, which is thus far the only known functional receptor for VEGI, contains a death domain and induces cell apoptosis. DcR3 is secreted as a soluble protein and antagonizes VEGI/DR3 interaction. Overexpression of DcR3 and downregulation of VEGI have been detected in a number of cancers. The aim of the present study was to investigate the effects of sodium valproate (VPA), a histone deacetylase inhibitor, in combination with hydralazine hydrochloride (Hy), a DNA methylation inhibitor, on the expression of VEGI and its related receptors in human osteosarcoma (OS) cell lines and human microvascular endothelial (HMVE) cells. Combination treatment with Hy and VPA synergistically induced the expression of VEGI and DR3 in both OS and HMVE cells, without inducing DcR3 secretion. In addition, it was observed that the combination of VPA and Hy significantly enhanced the inhibitory effect on vascular tube formation by VEGI/DR3 autocrine and paracrine pathways. Furthermore, the VEGI/VEGF‑A immune complex was pulled down by immunoprecipitation. Taken together, these findings suggest that DNA methyltransferase and histone deacetylase inhibitors not only have the potential to induce the re‑expression of tumor suppressor genes in cancer cells, but also exert anti‑angiogenic effects, via enhancement of the VEGI/DR3 pathway and VEGI/VEGF‑A interference.
Collapse
Affiliation(s)
- Shunsuke Kumanishi
- Department of Orthopedic Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Koji Yamanegi
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Hiroshi Nishiura
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Yuki Fujihara
- Department of Orthopedic Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Kenta Kobayashi
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Keiji Nakasho
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Hiroyuki Futani
- Department of Orthopedic Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Shinichi Yoshiya
- Department of Orthopedic Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| |
Collapse
|
17
|
TRAIL, OPG, and TWEAK in kidney disease: biomarkers or therapeutic targets? Clin Sci (Lond) 2019; 133:1145-1166. [PMID: 31097613 PMCID: PMC6526163 DOI: 10.1042/cs20181116] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 04/19/2019] [Accepted: 04/29/2019] [Indexed: 12/11/2022]
Abstract
Ligands and receptors of the tumor necrosis factor (TNF) superfamily regulate immune responses and homeostatic functions with potential diagnostic and therapeutic implications. Kidney disease represents a global public health problem, whose prevalence is rising worldwide, due to the aging of the population and the increasing prevalence of diabetes, hypertension, obesity, and immune disorders. In addition, chronic kidney disease is an independent risk factor for the development of cardiovascular disease, which further increases kidney-related morbidity and mortality. Recently, it has been shown that some TNF superfamily members are actively implicated in renal pathophysiology. These members include TNF-related apoptosis-inducing ligand (TRAIL), its decoy receptor osteoprotegerin (OPG), and TNF-like weaker inducer of apoptosis (TWEAK). All of them have shown the ability to activate crucial pathways involved in kidney disease development and progression (e.g. canonical and non-canonical pathways of the transcription factor nuclear factor-kappa B), as well as the ability to regulate cell proliferation, differentiation, apoptosis, necrosis, inflammation, angiogenesis, and fibrosis with double-edged effects depending on the type and stage of kidney injury. Here we will review the actions of TRAIL, OPG, and TWEAK on diabetic and non-diabetic kidney disease, in order to provide insights into their full clinical potential as biomarkers and/or therapeutic options against kidney disease.
Collapse
|
18
|
Wang K, Li Y, Dai Y, Han L, Zhu Y, Xue C, Wang P, Wang J. Peptides from Antarctic Krill ( Euphausia superba) Improve Osteoarthritis via Inhibiting HIF-2α-Mediated Death Receptor Apoptosis and Metabolism Regulation in Osteoarthritic Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:3125-3133. [PMID: 30798606 DOI: 10.1021/acs.jafc.8b05841] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Osteoarthritis (OA) is a prevalent debilitating disease which is predominantly characterized by cartilage degeneration. In the current study, destabilization of the medial meniscus (DMM) mouse model was used to investigate the effects of Antarctic krill peptides (AKP) on cartilage protection. As observed, AKP clearly ameliorate cartilage degeneration as evidenced by increased cartilage thickness and cartilage area and decreased histological Osteoarthritis Research Society International (OARSI) scores. Toluidine blue staining showed that AKO remarkably inhibited the loss of cartilage matrix in mice with OA. Hypoxia-inducible factor-2α (HIF-2α) has a key role in catabolic regulation and inflammation cascades which are the main causes of OA. AKP can down-regulate the expression of HIF-2α and its downstream genes such as MMP-13, Adamts-5, IL-1β, iNOS, CXCL-1, and NOS2. Consistent with this, anabolic genes such as Acan and Col2α1 were restored after treatment with AKP. Chondrocyte apoptosis and the reduction in cartilage cell viability are also involved in the process of OA. The HIF-2α-mediated death receptor apoptosis signaling pathway has been involved in the regulation of chondrocyte apoptosis. AKP can reduce the expressions of key pro-apoptosis genes in Fas-FasL and DR3-DR3L signaling pathways such as Fas, FasL, FADD, caspase8, caspase3, DR3, DR3L, RIP, and NF-κB. In addition, expressions of antiapoptosis genes such as c-AIP and c-FLIP were increased significantly. These findings indicate that AKP can be used as a new functional factor in the development of functional foods and chondroprotective drugs.
Collapse
Affiliation(s)
- Kai Wang
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| | - Yuanyuan Li
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| | - Yufeng Dai
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| | - Lihau Han
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| | - Yujie Zhu
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| | - Changhu Xue
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| | - Peng Wang
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| | - Jingfeng Wang
- College of Food Science and Engineering , Ocean University of China , Qingdao , Shandong Province 266003 , China
| |
Collapse
|
19
|
Infante M, Fabi A, Cognetti F, Gorini S, Caprio M, Fabbri A. RANKL/RANK/OPG system beyond bone remodeling: involvement in breast cancer and clinical perspectives. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:12. [PMID: 30621730 PMCID: PMC6325760 DOI: 10.1186/s13046-018-1001-2] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022]
Abstract
RANKL/RANK/OPG system consists of three essential signaling molecules: i) the receptor activator of nuclear factor (NF)-kB-ligand (RANKL), ii) the receptor activator of NF-kB (RANK), and iii) the soluble decoy receptor osteoprotegerin (OPG). Although this system is critical for the regulation of osteoclast differentiation/activation and calcium release from the skeleton, different studies have elucidated its specific role in mammary gland physiology and hormone-driven epithelial proliferation during pregnancy. Of note, several data suggest that progesterone induces mammary RANKL expression in mice and humans. In turn, RANKL controls cell proliferation in breast epithelium under physiological conditions typically associated with higher serum progesterone levels, such as luteal phase of the menstrual cycle and pregnancy. Hence, RANKL/RANK system can be regarded as a major downstream mediator of progesterone-driven mammary epithelial cells proliferation, potentially contributing to breast cancer initiation and progression. Expression of RANKL, RANK, and OPG has been detected in breast cancer cell lines and in human primary breast cancers. To date, dysregulation of RANKL/RANK/OPG system at the skeletal level has been widely documented in the context of metastatic bone disease. In fact, RANKL inhibition through the RANKL-blocking human monoclonal antibody denosumab represents a well-established therapeutic option to prevent skeletal-related events in metastatic bone disease and adjuvant therapy-induced bone loss in breast cancer. On the other hand, the exact role of OPG in breast tumorigenesis is still unclear. This review focuses on molecular mechanisms linking RANKL/RANK/OPG system to mammary tumorigenesis, highlighting pre-clinical and clinical evidence for the potential efficacy of RANKL inhibition as a prevention strategy and adjuvant therapy in breast cancer settings.
Collapse
Affiliation(s)
- Marco Infante
- Unit of Endocrinology and Metabolic Diseases, Department of Systems Medicine, CTO A. Alesini Hospital, ASL Roma 2, University Tor Vergata, Via San Nemesio, 21, 00145, Rome, Italy
| | - Alessandra Fabi
- Division of Medical Oncology 1, Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy
| | - Francesco Cognetti
- Division of Medical Oncology 1, Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy
| | - Stefania Gorini
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta, 247, 00166, Rome, Italy
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta, 247, 00166, Rome, Italy.,Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Via di Val Cannuta, 247, 00166, Rome, Italy
| | - Andrea Fabbri
- Unit of Endocrinology and Metabolic Diseases, Department of Systems Medicine, CTO A. Alesini Hospital, ASL Roma 2, University Tor Vergata, Via San Nemesio, 21, 00145, Rome, Italy.
| |
Collapse
|
20
|
Jiang W, Wu DB, Fu SY, Chen EQ, Tang H, Zhou TY. Insight into the role of TRAIL in liver diseases. Biomed Pharmacother 2018; 110:641-645. [PMID: 30544063 DOI: 10.1016/j.biopha.2018.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 11/19/2018] [Accepted: 12/02/2018] [Indexed: 02/05/2023] Open
Abstract
TNF-related apoptosis inducing ligand (TRAIL) is a potential antitumor protein known for its ability to selectively eliminate various types of tumor cells without exerting toxic effects in normal cells and tissues. TRAIL has recently been suggested as a potential therapeutic target in hepatocellular carcinoma (HCC) because it promotes apoptosis in cancer cells. Furthermore, studies on the role of TRAIL in liver injury have reported that TRAIL plays an essential role in viral hepatitis, fatty liver diseases, etc. However, several contradictory and confounding effects of TRAIL in these liver diseases have not been fully elucidated or placed into perspective. Hence, this review summarizes recent progress in studies on TRAIL, including its role in apoptotic signaling, potential therapeutic applications of TRAIL in HCC, hepatitis virus infection, and liver fibrosis and cirrhosis.
Collapse
Affiliation(s)
- Wei Jiang
- Center of Infectious Diseases, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Wuhou District, Chengdu, Sichuan Province, China
| | - Dong-Bo Wu
- Center of Infectious Diseases, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Wuhou District, Chengdu, Sichuan Province, China
| | - Si-Yu Fu
- Center of Infectious Diseases, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Wuhou District, Chengdu, Sichuan Province, China
| | - En-Qiang Chen
- Center of Infectious Diseases, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Wuhou District, Chengdu, Sichuan Province, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Wuhou District, Chengdu, Sichuan Province, China
| | - Tao-You Zhou
- Center of Infectious Diseases, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Wuhou District, Chengdu, Sichuan Province, China.
| |
Collapse
|
21
|
Peng Y, Sheng X, Xue F, Qian Y. The genetic association between osteoprotegerin (OPG) gene polymorphisms and bone mineral density (BMD) in postmenopausal women: A meta-analysis. Medicine (Baltimore) 2018; 97:e13507. [PMID: 30572450 PMCID: PMC6320146 DOI: 10.1097/md.0000000000013507] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Osteoporosis is a common skeletal disorder in eldest people, especially in postmenopausal women. The osteoprotegerin (OPG) gene has been reported to be associated with the BMD and pathogenesis of osteoporosis. However, the results were inconsistent and inconclusive in previous studies. METHODS A meta-analysis was performed to investigate the effect of four common OPG gene polymorphisms (A163G, G1181C, T245G, and T950C) on BMD in postmenopausal women. RESULTS A total of 23 eligible studies with 12,973 postmenopausal women were enrolled in present study. Individuals who with AA genotype of A163G were found to have slightly higher femoral hip (P = .03, SMD = 0.49, [95% CI] = [0.06, 0.91]) and total hip BMD (P = .002, SMD = -0.25, [95% CI] = [-0.42, -0.09]) than those with AG genotype. Subjects with GG genotype of G1181C was found to have lower BMD than those with CC or GC genotypes in lumbar spine (GG vs GC: P = .0002, SMD = -0.85, [95% CI] = [-1.29, -0.41]; GG vs CC: P = .02, SMD = -0.21, [-0.39, -0.03]) and total hip BMD (GG vs GC: P = .002, SMD = -0.25, [95% CI] = [-0.42, -0.09]; GG vs CC: P = .01, SMD = -0.15, [95% CI] = [-0.26, -0.03]). In addition, the subjects with GC genotype of G1181C was detected to have lower BMD than those with CC genotype in lumbar spine BMD (P < .05). Furthermore, individuals with TT genotype of T950C were shown to have significant lower lumbar spine BMD compared with those with genotype CC in Caucasian (P < .05). The lumbar spine BMD was lower for subjects with TC genotype of T950C than those with CC genotype in both Caucasian and Asian populations (P < .05). In contrast to A163G, G1181C, and T950G, no association was detected between T245G polymorphism and BMD (P > .05). CONCLUSION The present meta-analysis demonstrated the OPG A163G, G1181C, and T950G, but not T245G, might influence the BMD in postmenopausal women.
Collapse
Affiliation(s)
- Yuqin Peng
- Department of Orthopedics, The First Affiliated Hospital of Suzhou University, Suzhou
| | - Xiaowen Sheng
- Department of Orthopedics, The First Affiliated Hospital of Suzhou University, Suzhou
| | - Feng Xue
- Department of Orthopedics, The First Affiliated Hospital of Suzhou University, Suzhou
| | - Yufeng Qian
- Department of Orthopedics, The First Affiliated Hospital of Suzhou University, Suzhou
- Department of Orthopedics, Changshu First People's Hospital, Changshu, People's Republic of China
| |
Collapse
|
22
|
Zhao Q, Hong B, Liu T, Ji Y, Tang X, Gong K, Ye L, Yang Y, Zhang N. VEGI174 protein and its functional domain peptides exert antitumour effects on renal cell carcinoma. Int J Oncol 2018; 54:390-398. [PMID: 30431089 DOI: 10.3892/ijo.2018.4632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/24/2018] [Indexed: 11/06/2022] Open
Abstract
Vascular endothelial growth inhibitor (VEGI) has been identified as an anti‑angiogenic cytokine. However, the effects of VEGI174 protein, and its functional domain peptides V7 and V8, on renal cell carcinoma (RCC) remain unknown. In the present study, the protein and peptides were biosynthesised as experimental agents. The A498 and 786‑O RCC cell lines, and an established mouse xenograft model, were separately treated with VEGI174, V7 or V8. Cellular functions, including proliferation, migration and invasion, were subsequently detected. Cell migration and invasion were monitored using the xCELLigence system. Furthermore, tumour growth and mouse behaviours, including mobility, appetite and body weight, were assessed. The results demonstrated that VEGI174, V7 and V8 inhibited the proliferation, migration and invasion of A498 and 786‑O cell lines when administered at concentrations of 1 and 100 pM, 10 nM and 1 µM. The inhibitory effects exhibited dose‑ and time‑dependent antitumour activity. Furthermore, VEGI174, V7 and V8 inhibited tumour growth in A498 and 786‑O xenograft mice. In the A498 xenografts, the tumour growth inhibition (TGI) rates in the VEGI174‑, V7‑ and V8‑treated groups were 71, 20 and 31%, respectively. In the 786‑O xenografts, the TGI rates in the VEGI174‑, V7‑ and V8‑treated groups were 34, 26 and 31%, respectively. There was no significant loss in body weight and no cases of mortality were observed for all treated mice. In conclusion, VEGI174, V7 and V8 exhibited potential antitumour effects and were well tolerated in vivo. V7 and V8, as functional domain peptides of the VEGI174 protein, may be studied for the future treatment of RCC.
Collapse
Affiliation(s)
- Qiang Zhao
- Department of Urology, Beijing Institute for Cancer Research, Beijing Cancer Hospital, Beijing 100142, P.R. China
| | - Baoan Hong
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, Beijing 100034, P.R. China
| | - Tiezhu Liu
- Department of Urology, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163001, P.R. China
| | - Yongpeng Ji
- Department of Urology, Beijing Institute for Cancer Research, Beijing Cancer Hospital, Beijing 100142, P.R. China
| | - Xinxin Tang
- Department of Urology, Beijing Institute for Cancer Research, Beijing Cancer Hospital, Beijing 100142, P.R. China
| | - Kan Gong
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, Beijing 100034, P.R. China
| | - Lin Ye
- Metastasis and Angiogenesis Research Group, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | - Yong Yang
- Department of Urology, Beijing Institute for Cancer Research, Beijing Cancer Hospital, Beijing 100142, P.R. China
| | - Ning Zhang
- Department of Urology, Beijing Institute for Cancer Research, Beijing Cancer Hospital, Beijing 100142, P.R. China
| |
Collapse
|
23
|
Sawicka-Powierza J, Jablonska E, Ratajczak-Wrona W, Rogowska-Szadkowska D, Garley M, Oltarzewska AM, Chlabicz S, Konstantynowicz J. Bone Metabolism Markers and Bone Mineral Density in Patients on Long-Term Acenocoumarol Treatment: A Cross-Sectional Study. J Clin Med 2018; 7:jcm7100372. [PMID: 30347817 PMCID: PMC6209941 DOI: 10.3390/jcm7100372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 10/17/2018] [Accepted: 10/18/2018] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to evaluate levels of osteocalcin (OC), osteoprotegerin (OPG) and total soluble receptor activator of nuclear factor-κB ligand (RANKL), and bone mineral density (BMD) in patients on long-term acenocoumarol (AC) treatment. The cross-sectional study was carried out in 42 patients treated long-term with AC and 28 control subjects. Serum concentrations of OC, OPG, and sRANKL were measured using enzyme linked immunosorbent assay (ELISA) kits, and BMD at the femoral neck and lumbar spine were assessed by dual energy X-ray absorptiometry. A significantly decreased concentration of OC was found in AC users compared to control subjects (4.94 ± 2.22 vs. 10.68 ± 4.5; p < 0.001). Levels of OPG, sRANKL logarithm (log), sRANKL/OPG log ratio, and BMD were comparable between. In female AC users, positive correlations between OC and RANKL log, and between OC and RANKL/OPG log ratio (p = 0.017; p = 0.005, respectively), and a negative correlation between OC and OPG (p = 0.027) were found. Long-term AC anticoagulation significantly decreases OC concentration, but does not affect other bone metabolism markers or BMD. Our results also suggest the possibility that long-term treatment with AC may alleviate bone resorption in postmenopausal women.
Collapse
Affiliation(s)
- Jolanta Sawicka-Powierza
- Department of Family Medicine, Medical University of Bialystok, Bialystok 15-054, Poland.
- Department of Haematology, Medical University of Bialystok, Bialystok 15-276, Poland.
| | - Ewa Jablonska
- Department of Immunology, Medical University of Bialystok, Bialystok 15-269, Poland.
| | | | | | - Marzena Garley
- Department of Immunology, Medical University of Bialystok, Bialystok 15-269, Poland.
| | - Alicja M Oltarzewska
- Department of Family Medicine, Medical University of Bialystok, Bialystok 15-054, Poland.
| | - Slawomir Chlabicz
- Department of Family Medicine, Medical University of Bialystok, Bialystok 15-054, Poland.
| | - Jerzy Konstantynowicz
- Department of Pediatric Rheumatology, Immunology, and Metabolic Bone Diseases, Medical University of Bialystok, Bialystok 15-274, Poland.
| |
Collapse
|
24
|
Inflammation-independent TL1A-mediated intestinal fibrosis is dependent on the gut microbiome. Mucosal Immunol 2018; 11:1466-1476. [PMID: 29988118 PMCID: PMC6162160 DOI: 10.1038/s41385-018-0055-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 05/16/2018] [Accepted: 06/04/2018] [Indexed: 02/08/2023]
Abstract
Tumor necrosis factor-like cytokine 1A (TL1A, TNFSF15) is implicated in inflammatory bowel disease (IBD), modulating the location and severity of intestinal inflammation and fibrosis. TL1A expression is increased in inflamed gut mucosa and associated with fibrostenosing Crohn's disease. Tl1a-overexpression in mice lead to spontaneous ileitis, and exacerbated induced proximal colitis and fibrosis. IBD is associated with shifts in the gut microbiome, but the effect of differing microbial populations and their interaction with TL1A on fibrosis has not been investigated. We demonstrate that the pro-fibrotic and inflammatory phenotype resulting from Tl1a-overexpression is abrogated in the absence of resident microbiota. To evaluate if this is due to the absence of a unique bacterial population, as opposed to any bacteria per se, we gavaged germ-free (GF) wild-type and Tl1a-transgenic (Tl1a-Tg) mice with stool from specific pathogen free (SPF) mice and a healthy human donor (Hu). Reconstitution with SPF, but not Hu microbiota, resulted in increased intestinal collagen deposition and fibroblast activation in Tl1a-Tg mice. Notably, there was reduced fibroblast migration and activation under GF conditions compared to native conditions. We then identified several candidate organisms that correlated directly with increased fibrosis in reconstituted mice and showed that these organisms directly impact fibroblast function in vitro. Thus, Tl1a-mediated intestinal fibrosis and fibroblast activation are dependent on specific microbial populations.
Collapse
|
25
|
Han Y, You X, Xing W, Zhang Z, Zou W. Paracrine and endocrine actions of bone-the functions of secretory proteins from osteoblasts, osteocytes, and osteoclasts. Bone Res 2018; 6:16. [PMID: 29844945 PMCID: PMC5967329 DOI: 10.1038/s41413-018-0019-6] [Citation(s) in RCA: 323] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/21/2018] [Accepted: 04/16/2018] [Indexed: 12/17/2022] Open
Abstract
The skeleton is a dynamic organ that is constantly remodeled. Proteins secreted from bone cells, namely osteoblasts, osteocytes, and osteoclasts exert regulation on osteoblastogenesis, osteclastogenesis, and angiogenesis in a paracrine manner. Osteoblasts secrete a range of different molecules including RANKL/OPG, M-CSF, SEMA3A, WNT5A, and WNT16 that regulate osteoclastogenesis. Osteoblasts also produce VEGFA that stimulates osteoblastogenesis and angiogenesis. Osteocytes produce sclerostin (SOST) that inhibits osteoblast differentiation and promotes osteoclast differentiation. Osteoclasts secrete factors including BMP6, CTHRC1, EFNB2, S1P, WNT10B, SEMA4D, and CT-1 that act on osteoblasts and osteocytes, and thereby influenceaA osteogenesis. Osteoclast precursors produce the angiogenic factor PDGF-BB to promote the formation of Type H vessels, which then stimulate osteoblastogenesis. Besides, the evidences over the past decades show that at least three hormones or "osteokines" from bone cells have endocrine functions. FGF23 is produced by osteoblasts and osteocytes and can regulate phosphate metabolism. Osteocalcin (OCN) secreted by osteoblasts regulates systemic glucose and energy metabolism, reproduction, and cognition. Lipocalin-2 (LCN2) is secreted by osteoblasts and can influence energy metabolism by suppressing appetite in the brain. We review the recent progresses in the paracrine and endocrine functions of the secretory proteins of osteoblasts, osteocytes, and osteoclasts, revealing connections of the skeleton with other tissues and providing added insights into the pathogenesis of degenerative diseases affecting multiple organs and the drug discovery process.
Collapse
Affiliation(s)
- Yujiao Han
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031 China
| | - Xiuling You
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031 China
| | - Wenhui Xing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031 China
| | - Zhong Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031 China
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031 China
| |
Collapse
|
26
|
Goswami S, Sharma-Walia N. Osteoprotegerin rich tumor microenvironment: implications in breast cancer. Oncotarget 2018; 7:42777-42791. [PMID: 27072583 PMCID: PMC5173171 DOI: 10.18632/oncotarget.8658] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 03/31/2016] [Indexed: 12/18/2022] Open
Abstract
Osteoprotegerin (OPG) is a soluble decoy receptor for tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL). It belongs to the tumor necrosis factor receptor superfamily (TNFRSF). OPG was initially discovered to contribute to homeostasis of bone turnover due to its capability of binding to receptor activator of nuclear factor-kappaB (NF-kB). However, apart from bone turnover, OPG plays important and diverse role(s) in many biological functions. Besides having anti-osteoclastic activity, OPG is thought to exert a protective anti-apoptotic action in OPG-expressing tumors by overcoming the physiologic mechanism of tumor surveillance exerted by TRAIL. Along with inhibiting TRAIL induced apoptosis, it can induce proliferation by binding to various cell surface receptors and thus turning on the canonical cell survival and proliferative pathways. OPG also induces angiogenesis, one of the hallmarks of cancer, thus facilitating tumor growth. Recently, the understanding of OPG and its different roles has been augmented substantially. This review is aimed at providing a very informative overview as to how OPG affects cancer progression especially breast cancer.
Collapse
Affiliation(s)
- Sudeshna Goswami
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Neelam Sharma-Walia
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| |
Collapse
|
27
|
Langdahl BL, Ralston SH. How Basic Science Discoveries Have Shaped the Treatment of Bone and Mineral Disorders. J Bone Miner Res 2017; 32:2324-2330. [PMID: 29194750 DOI: 10.1002/jbmr.3316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Bente L Langdahl
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Stuart H Ralston
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
28
|
Hashiramoto A, Konishi Y, Murayama K, Kawasaki H, Yoshida K, Tsumiyama K, Tanaka K, Mizuhara M, Shiotsuki T, Kitamura H, Komai K, Kimura T, Yagita H, Shiozawa K, Shiozawa S. A variant of death-receptor 3 associated with rheumatoid arthritis interferes with apoptosis-induction of T cell. J Biol Chem 2017; 293:1933-1943. [PMID: 29180447 PMCID: PMC5808757 DOI: 10.1074/jbc.m117.798884] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 11/14/2017] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic polyarthritis of unknown etiology. To unravel the molecular mechanisms in RA, we performed targeted DNA sequencing analysis of patients with RA. This analysis identified a variant of the death receptor 3 (DR3) gene, a member of the family of apoptosis-inducing Fas genes, which contains four single-nucleotide polymorphisms (SNPs) and a 14-nucleotide deletion within exon 5 and intron 5. We found that the deletion causes the binding of splicing regulatory proteins to DR3 pre-mRNA intron 5, resulting in a portion of intron 5 becoming part of the coding sequence, thereby generating a premature stop codon. We also found that this truncated DR3 protein product lacks the death domain and forms a heterotrimer complex with wildtype DR3 that dominant-negatively inhibits ligand-induced apoptosis in lymphocytes. Myelocytes from transgenic mice expressing the human DR3 variant produced soluble truncated DR3, forming a complex with TNF-like ligand 1A (TL1A), which inhibited apoptosis induction. In summary, our results reveal that a DR3 splice variant that interferes with ligand-induced T cell responses and apoptosis may contribute to RA pathogenesis.
Collapse
Affiliation(s)
- Akira Hashiramoto
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Yoshitake Konishi
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Koichi Murayama
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Hiroki Kawasaki
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Kohsuke Yoshida
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Ken Tsumiyama
- the Department of Medicine, Rheumatic Diseases Unit, Kyushu University Beppu Hospital, Beppu 874-0838
| | - Kimie Tanaka
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Masaru Mizuhara
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Toshio Shiotsuki
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Hitomi Kitamura
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Koichiro Komai
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Tomoatsu Kimura
- the Department of Orthopedic Surgery, Faculty of Medicine, University of Toyama, 3190 Gofuku, 930-0194 Toyama
| | - Hideo Yagita
- the Department of Immunology, Juntendo University School of Medicine, Tokyo 113-8431, and
| | - Kazuko Shiozawa
- the Department of Rheumatology, Hyogo Prefectural Kakogawa Medical Center, Kakogawa 675-8555, Japan
| | - Shunichi Shiozawa
- the Department of Medicine, Rheumatic Diseases Unit, Kyushu University Beppu Hospital, Beppu 874-0838,
| |
Collapse
|
29
|
Zhang N, Hong B, Lian W, Zhou C, Chen S, Du X, Deng X, Duoerkun S, Li Q, Yang Y, Gong K. Vascular endothelial growth inhibitor 174 and its functional domains inhibit epithelial-mesenchymal transition in renal cell carcinoma cells in vitro. Int J Mol Med 2017; 40:569-575. [PMID: 28656288 DOI: 10.3892/ijmm.2017.3033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 06/08/2017] [Indexed: 11/06/2022] Open
Abstract
The present study was carried out to investigate the effects of vascular endothelial growth inhibitor 174 (VEGI174) and its functional domains (V7 and V8) on epithelial‑mesenchymal transition (EMT) in renal cell carcinoma (RCC) cells in vitro. The RCC cell lines A498 and 786‑O were used in this study. Based on our preliminary study, we selected full‑length VEGI174 and its functional domains (V7 and V8) as the target genes in this study. Plasmids containing VEGI174, V7 or V8 transgenes were constructed and transfected into A498 and 786‑O cell lines. Cytological activity was tested during cell culture. Quantitative PCR and western blot analysis were performed to determine the expression levels of EMT markers (E‑cadherin, vimentin, β‑catenin and Slug). Overexpression of VEGI174, V7 or V8 did not have a significant influence on cell viability (P>0.05). The mRNA level of E‑cadherin was significantly upregulated, while that of vimentin was downregulated in A498VEGIexp, A498V7exp, A498V8exp, 786‑OVEGIexp, 786‑OV7exp and 786‑OV8exp cells compared with the cells containing the empty plasmid controls (P<0.05). The western blot results showed that changes in protein expression levels were consistent with the changes in mRNA expression. Both the mRNA and protein expression levels of β‑catenin and Slug were downregulated in the A498VEGIexp, A498V7exp, A498V8exp, 786‑OVEGIexp, 786‑OV7exp and 786‑OV8exp cells. In conclusion, overexpression of VEGI174, V7 or V8 inhibited EMT in A498 and 786‑O cells. Notably, V7 and V8 are two effective functional domains of VEGI174 that have the potential to be studied for peptide synthesis and the treatment of RCC.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Urology, Peking University Cancer Hospital, Beijing Institute for Cancer Research, Beijing 100142, P.R. China
| | - Baoan Hong
- Department of Urology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Wenyong Lian
- Department of Urology, Xinjiang Production and Construction Corps First Division Hospital, Aksu, Xinjiang 843000, P.R. China
| | - Changhua Zhou
- School of Pharmaceutical Sciences, Center for Cellular and Structural Biology, Sun Yat-Sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Siqi Chen
- School of Pharmaceutical Sciences, Center for Cellular and Structural Biology, Sun Yat-Sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Xin Du
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Xiaohu Deng
- Department of Urology, Karamay People's Hospital, Karamay, Xinjiang 834000, P.R. China
| | - Shayiremu Duoerkun
- Department of Urology, Hami District Central Hospital, Hami, Xinjiang 839000, P.R. China
| | - Qing Li
- School of Pharmaceutical Sciences, Center for Cellular and Structural Biology, Sun Yat-Sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Yong Yang
- Department of Urology, Peking University Cancer Hospital, Beijing Institute for Cancer Research, Beijing 100142, P.R. China
| | - Kan Gong
- Department of Urology, Peking University First Hospital, Beijing 100034, P.R. China
| |
Collapse
|
30
|
Bittner S, Knoll G, Ehrenschwender M. Death receptor 3 signaling enhances proliferation of human regulatory T cells. FEBS Lett 2017; 591:1187-1195. [DOI: 10.1002/1873-3468.12632] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/10/2017] [Accepted: 03/20/2017] [Indexed: 01/18/2023]
Affiliation(s)
- Sebastian Bittner
- Institute of Clinical Microbiology and Hygiene; University Hospital Regensburg; Germany
| | - Gertrud Knoll
- Institute of Clinical Microbiology and Hygiene; University Hospital Regensburg; Germany
| | - Martin Ehrenschwender
- Institute of Clinical Microbiology and Hygiene; University Hospital Regensburg; Germany
| |
Collapse
|
31
|
Fortner RT, Sarink D, Schock H, Johnson T, Tjønneland A, Olsen A, Overvad K, Affret A, His M, Boutron-Ruault MC, Boeing H, Trichopoulou A, Naska A, Orfanos P, Palli D, Sieri S, Mattiello A, Tumino R, Ricceri F, Bueno-de-Mesquita HB, Peeters PHM, Van Gils CH, Weiderpass E, Lund E, Quirós JR, Agudo A, Sánchez MJ, Chirlaque MD, Ardanaz E, Dorronsoro M, Key T, Khaw KT, Rinaldi S, Dossus L, Gunter M, Merritt MA, Riboli E, Kaaks R. Osteoprotegerin and breast cancer risk by hormone receptor subtype: a nested case-control study in the EPIC cohort. BMC Med 2017; 15:26. [PMID: 28173834 PMCID: PMC5297136 DOI: 10.1186/s12916-017-0786-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 01/10/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Circulating osteoprotegerin (OPG), a member of the receptor activator of nuclear factor kappa-B (RANK) axis, may influence breast cancer risk via its role as the decoy receptor for both the RANK ligand (RANKL) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Circulating OPG and breast cancer risk has been examined in only one prior study. METHODS A case-control study was nested in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. A total of 2008 incident invasive breast cancer cases (estrogen receptor (ER)+, n = 1622; ER-, n = 386), matched 1:1 to controls, were included in the analysis. Women were predominantly postmenopausal at blood collection (77%); postmenopausal women included users and non-users of postmenopausal hormone therapy (HT). Serum OPG was quantified with an electrochemiluminescence assay. Relative risks (RRs) and 95% confidence intervals (CIs) were calculated using conditional logistic regression. RESULTS The associations between OPG and ER+ and ER- breast cancer differed significantly. Higher concentrations of OPG were associated with increased risk of ER- breast cancer (top vs. bottom tertile RR = 1.93 [95% CI 1.24-3.02]; p trend = 0.03). We observed a suggestive inverse association for ER+ disease overall and among women premenopausal at blood collection. Results for ER- disease did not differ by menopausal status at blood collection (p het = 0.97), and we observed no heterogeneity by HT use at blood collection (p het ≥ 0.43) or age at breast cancer diagnosis (p het ≥ 0.30). CONCLUSIONS This study provides the first prospective data on OPG and breast cancer risk by hormone receptor subtype. High circulating OPG may represent a novel risk factor for ER- breast cancer.
Collapse
Affiliation(s)
- Renée T. Fortner
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | - Danja Sarink
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | - Helena Schock
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | - Theron Johnson
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | - Anne Tjønneland
- Diet, Genes and Environment, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Anja Olsen
- Diet, Genes and Environment, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Kim Overvad
- Section for Epidemiology, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Aurélie Affret
- Université Paris-Saclay, Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
- Gustave Roussy, F-94805 Villejuif, France
| | - Mathilde His
- Université Paris-Saclay, Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
- Gustave Roussy, F-94805 Villejuif, France
| | - Marie-Christine Boutron-Ruault
- Université Paris-Saclay, Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
- Gustave Roussy, F-94805 Villejuif, France
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Antonia Trichopoulou
- Hellenic Health Foundation, Athens, Greece
- WHO Collaborating Center for Nutrition and Health, Unit of Nutritional Epidemiology and Nutrition in Public Health, Department of Hygiene, Epidemiology and Medical Statistics, University of Athens Medical School, Athens, Greece
| | - Androniki Naska
- Hellenic Health Foundation, Athens, Greece
- WHO Collaborating Center for Nutrition and Health, Unit of Nutritional Epidemiology and Nutrition in Public Health, Department of Hygiene, Epidemiology and Medical Statistics, University of Athens Medical School, Athens, Greece
| | - Philippos Orfanos
- Hellenic Health Foundation, Athens, Greece
- WHO Collaborating Center for Nutrition and Health, Unit of Nutritional Epidemiology and Nutrition in Public Health, Department of Hygiene, Epidemiology and Medical Statistics, University of Athens Medical School, Athens, Greece
| | - Domenico Palli
- Cancer Risk Factors and Life-Style Epidemiology Unit, Cancer Research and Prevention Institute – ISPO, Florence, Italy
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Department of Preventive & Predictive Medicine Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Amalia Mattiello
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Rosario Tumino
- Cancer Registry and Histopathology Unit, “Civic - M.p.Arezzo” Hospital, ASP Ragusa, Italy
| | - Fulvio Ricceri
- Unit of Epidemiology, Regional Health Service ASL TO3, Grugliasco (TO), Italy
- Unit of Cancer Epidemiology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - H. Bas Bueno-de-Mesquita
- Department for Determinants of Chronic Diseases (DCD), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Social & Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Petra H. M. Peeters
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, UK
| | - Carla H. Van Gils
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Elisabete Weiderpass
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, Tromsø, Norway
- Department of Research, Cancer Registry of Norway, Institute of Population-Based Cancer Research, Oslo, Norway
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Genetic Epidemiology Group, Folkhälsan Research Center, Helsinki, Finland
| | - Eiliv Lund
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, Tromsø, Norway
| | | | - Antonio Agudo
- Unit of Nutrition and Cancer. Cancer Epidemiology Research Program. Catalan Institute of Oncology-IDIBELL. L’Hospitalet de Llobregat, Barcelona, Spain
| | - Maria-José Sánchez
- Escuela Andaluza de Salud Pública. Instituto de Investigación Biosanitaria ibs. GRANADA. Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - María-Dolores Chirlaque
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Epidemiology, Regional Health Council, IMIB-Arrixaca, Murcia, Spain
- Department of Health and Social Sciences, Universidad de Murcia, Murcia, Spain
| | - Eva Ardanaz
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Navarra Public Health Institute, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Miren Dorronsoro
- Public Health Direction and Biodonostia Research Institute CIBERESP, Basque Regional Health Department, San Sebastian, Spain
| | - Tim Key
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Kay-Tee Khaw
- Cancer Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Sabina Rinaldi
- International Agency for Research on Cancer, Lyon, France
| | - Laure Dossus
- International Agency for Research on Cancer, Lyon, France
| | - Marc Gunter
- International Agency for Research on Cancer, Lyon, France
| | - Melissa A. Merritt
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| |
Collapse
|
32
|
Abstract
Inflammatory bowel diseases (IBDs) are thought to develop as a result of complex interactions between host genetics, the immune system and the environment including the gut microbiome. Although an improved knowledge of the immunopathogenesis of IBDs has led to great advances in therapy such as the highly effective anti-tumor necrosis factor class of medications, a significant proportion of patients with Crohn's disease and ulcerative colitis do not respond to anti-tumor necrosis factor antibodies. Further understanding of the different immune pathways involved in the genesis of chronic intestinal inflammation is required to help find effective treatments for IBDs. In this review, the role of the mucosal innate and adaptive immune system in IBD is summarized, highlighting new areas of discovery which may hold the key to identifying novel predictive or prognostic biomarkers and new avenues of therapeutic discovery.
Collapse
|
33
|
Gorczynski RM, Sadozai H, Zhu F, Khatri I. Effect of infusion of monoclonal antibodies to tumour necrosis factor-receptor super family 25 on graft rejection in allo-immune mice receiving autologous marrow transplantation. Immunology 2016; 150:418-431. [PMID: 27859243 DOI: 10.1111/imm.12693] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/07/2016] [Accepted: 11/14/2016] [Indexed: 02/06/2023] Open
Abstract
Significant barriers to transplantation exist for individuals who are pre-sensitized to donor antigen and have high titres of donor-reactive antibody. We report the effect of autologous bone marrow transplantation (BMTx) after myeloablation in pre-sensitized mice along with the use of monoclonal antibodies (mAbs) to tumour necrosis factor-receptor super family 25 (TNFRSF25), expressed on regulatory T (Treg) cells. C57BL/6 mice, which had been sensitized earlier with BALB/c skin allografts, received secondary BALB/c grafts after the primary grafts had been rejected. Subsequently, recipient mice underwent myeloablation with cyclophosphamide and busulphan and were injected with T-cell-depleted bone marrow from CD45.1 congenic donors (BMTx). Recipient mice underwent immunosuppressive treatment with rapamycin. A subgroup of mice was also treated with mAbs to TNFRSF25. Control mice were pre-sensitized mice that received cyclophosphamide and busulphan followed by rapamycin. BMTx-treated mice had significantly prolonged skin graft survival versus control mice. These mice also showed attenuated donor-specific mixed lymphocyte co-culture responses relative to controls, increased splenic Treg cells and markedly diminished serum anti-donor IgG. Infusion of anti-TNFRSF25 mAbs further augmented graft survival and increased graft-infiltrating Treg cells. These mAbs also expanded murine and human Treg cells in vitro with the capacity to attenuate mixed lymphocyte co-cultures using fresh peripheral blood mononuclear cells. Overall, this study delineates the roles of autologous BMTx and anti-TNFRSF25 mAbs in expanding Treg cells and attenuating alloimmune responses in pre-sensitized mice.
Collapse
Affiliation(s)
- Reginald M Gorczynski
- Transplant Research Division, University Health Network and Toronto General Hospital, Toronto, ON, Canada.,Department of Immunology and Surgery, University of Toronto, Toronto, ON, Canada
| | - Hassan Sadozai
- Transplant Research Division, University Health Network and Toronto General Hospital, Toronto, ON, Canada
| | - Fang Zhu
- Transplant Research Division, University Health Network and Toronto General Hospital, Toronto, ON, Canada
| | - Ismat Khatri
- Transplant Research Division, University Health Network and Toronto General Hospital, Toronto, ON, Canada
| |
Collapse
|
34
|
B Virus (Macacine Herpesvirus 1) Divergence: Variations in Glycoprotein D from Clinical and Laboratory Isolates Diversify Virus Entry Strategies. J Virol 2016; 90:9420-32. [PMID: 27512063 DOI: 10.1128/jvi.00799-16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/03/2016] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED B virus (Macacine herpesvirus 1) can cause deadly zoonotic disease in humans. Molecular mechanisms of B virus cell entry are poorly understood for both macaques and humans. Here we investigated the abilities of clinical B virus isolates to use entry receptors of herpes simplex viruses (HSV). We showed that resistant B78H1 cells became susceptible to B virus clinical strains upon expression of either human nectin-2 or nectin-1. Antibody against glycoprotein D (gD) protected these nectin-bearing cells from B virus infection, and a gD-negative recombinant B virus failed to enter these cells, indicating that the nectin-mediated B virus entry depends on gD. We observed that the infectivity of B virus isolates with a single amino acid substitution (D122N) in the IgV-core of the gD ectodomain was impaired on nectin-1-bearing cells. Computational homology-based modeling of the B virus gD-nectin-1 complex revealed conformational differences between the structures of the gD-122N and gD-122D variants that affected the gD-nectin-1 protein-protein interface and binding affinity. Unlike HSV, B virus clinical strains were unable to use herpesvirus entry mediator (HVEM) as a receptor, regardless of conservation of the gD amino acid residues essential for HSV-1 entry via HVEM. Based on the model of the B virus gD-HVEM interface, we predict that residues R7, R11, and G15 are largely responsible for the inability of B virus to utilize HVEM for entry. The ability of B virus to enter cells of a human host by using a combination of receptors distinct from those for HSV-1 or HSV-2 suggests a possible mechanism of enhanced neuropathogenicity associated with zoonotic infections. IMPORTANCE B virus causes brainstem destruction in infected humans in the absence of timely diagnosis and intervention. Nectins are cell adhesion molecules that are widely expressed in human tissues, including neurons and neuronal synapses. Here we report that human nectin-2 is a target receptor for B virus entry, in addition to the reported receptor human nectin-1. Similar to a B virus lab strain, B virus clinical strains can effectively use both nectin-1 and nectin-2 as cellular receptors for entry into human cells, but unlike HSV-1 and HSV-2, none of the clinical strains uses an HVEM-mediated entry pathway. Ultimately, these differences between B virus and HSV-1 and -2 may provide insight into the neuropathogenicity of B virus during zoonotic infections.
Collapse
|
35
|
Death Receptor 3 Promotes Chemokine-Directed Leukocyte Recruitment in Acute Resolving Inflammation and Is Essential for Pathological Development of Mesothelial Fibrosis in Chronic Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2813-2823. [PMID: 27664471 DOI: 10.1016/j.ajpath.2016.07.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 06/20/2016] [Accepted: 07/19/2016] [Indexed: 12/19/2022]
Abstract
Death receptor 3 (DR3; TNFRSF25) and its tumor necrosis factor-like ligand TL1A (TNFSF15) control several processes in inflammatory diseases through the expansion of effector T cells and the induction of proinflammatory cytokines from myeloid and innate lymphoid cells. Using wild-type (DR3+/+) and DR3-knockout (DR3-/-) mice, we show that the DR3/TL1A pathway triggers the release of multiple chemokines after acute peritoneal inflammation initiated by a single application of Staphylococcus epidermidis supernatant, correlating with the infiltration of multiple leukocyte subsets. In contrast, leukocyte infiltration was not DR3 dependent after viral challenge with murine cytomegalovirus. DR3 expression was recorded on connective tissue stroma, which provided DR3-dependent release of chemokine (C-C motif) ligand (CCL) 2, CCL7, CXCL1, and CXCL13. CCL3, CCL4, and CXCL10 production was also DR3 dependent, but quantitative RT-PCR showed that their derivation was not stromal. In vitro cultures identified resident macrophages as a DR3-dependent source of CCL3. Whether DR3 signaling could contribute to a related peritoneal pathology was then tested using multiple applications of S. epidermidis supernatant, the repetitive inflammatory episodes of which lead to peritoneal membrane thickening and collagen deposition. Unlike their DR3+/+ counterparts, DR3-/- mice did not develop fibrosis of the mesothelial layer. Thus, this work describes both a novel function and essential requirement for the DR3/TL1A pathway in acute, resolving, and chronic inflammation in the peritoneal cavity.
Collapse
|
36
|
Cavallini C, Lovato O, Bertolaso A, Zoratti E, Malpeli G, Mimiola E, Tinelli M, Aprili F, Tecchio C, Perbellini O, Scarpa A, Zamò A, Cassatella MA, Pizzolo G, Scupoli MT. Expression and function of the TL1A/DR3 axis in chronic lymphocytic leukemia. Oncotarget 2016; 6:32061-74. [PMID: 26393680 PMCID: PMC4741659 DOI: 10.18632/oncotarget.5201] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/04/2015] [Indexed: 01/15/2023] Open
Abstract
TNF-like ligand 1A (TL1A) and its unique receptor death receptor 3 (DR3) acts as broad T-cell costimulator involved in regulatory mechanisms of adaptive immune response under physiological and pathological settings. Moreover, we have recently shown that TL1A negatively regulates B-cell proliferation. Despite increasing interest on the TL1A/DR3-axis functions, very little is known on its expression and role in leukemia. In this study, we investigated the expression and function of TL1A/DR3 axis in chronic lymphocytic leukemia (CLL). DR3 was differentially expressed in activated CLL cells and predominantly detected in patients with early clinical stage disease. Soluble TL1A has been revealed in the sera of CLL patients where higher TL1A levels were associated with early stage disease. T cells, monocytes and leukemic B cells have been identified as major sources of TL1A in CLL. The relevance of these findings has been sustained by functional data showing that exogenous TL1A reduces CLL proliferation induced by stimulation of the B cell receptor. Overall, these data document the expression of the TL1A/DR3 axis in early-stage CLL. They also identify a novel function for TL1A as a negative regulator of leukemic cell proliferation that may influence the CLL physiopathology and clinical outcome at an early-stage disease.
Collapse
Affiliation(s)
- Chiara Cavallini
- Interdepartmental Laboratory of Medical Research (LURM), University of Verona, Verona, Italy
| | - Ornella Lovato
- Interdepartmental Laboratory of Medical Research (LURM), University of Verona, Verona, Italy
| | - Anna Bertolaso
- Department of Pathology and Diagnostics, Section of Pathological Anatomy, University of Verona, Verona, Italy
| | - Elisa Zoratti
- Interdepartmental Laboratory of Medical Research (LURM), University of Verona, Verona, Italy.,Applied Research on Cancer-Network (ARC-NET), University of Verona, Verona, Italy
| | - Giorgio Malpeli
- Department of Pathology and Diagnostics, Section of Pathological Anatomy, University of Verona, Verona, Italy.,Applied Research on Cancer-Network (ARC-NET), University of Verona, Verona, Italy
| | - Elda Mimiola
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Martina Tinelli
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Fiorenza Aprili
- Department of Pathology and Diagnostics, Laboratory of Cytogenetics, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Cristina Tecchio
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Omar Perbellini
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Aldo Scarpa
- Department of Pathology and Diagnostics, Section of Pathological Anatomy, University of Verona, Verona, Italy.,Applied Research on Cancer-Network (ARC-NET), University of Verona, Verona, Italy
| | - Alberto Zamò
- Department of Pathology and Diagnostics, Section of Pathological Anatomy, University of Verona, Verona, Italy
| | - Marco Antonio Cassatella
- Department of Pathology and Diagnostics, Section of General Pathology, University of Verona, Verona, Italy
| | - Giovanni Pizzolo
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Maria Teresa Scupoli
- Interdepartmental Laboratory of Medical Research (LURM), University of Verona, Verona, Italy.,Applied Research on Cancer-Network (ARC-NET), University of Verona, Verona, Italy.,Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| |
Collapse
|
37
|
Jacob N, Targan SR, Shih DQ. Cytokine and anti-cytokine therapies in prevention or treatment of fibrosis in IBD. United European Gastroenterol J 2016; 4:531-40. [PMID: 27536363 DOI: 10.1177/2050640616649356] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 04/19/2016] [Indexed: 12/18/2022] Open
Abstract
The frequency of fibrosing Crohn's disease (CD) is significant, with approximately 40% of CD patients with ileal disease developing clinically apparent strictures throughout their lifetime. Although strictures may be subdivided into fibrotic, inflammatory, or mixed forms, despite immunosuppressive therapy in CD patients in the form of steroids or immunomodulators, the frequency of fibrostenosing complications has still remained significant. A vast number of genetic and epigenetic variables are thought to contribute to fibrostenosing disease, including those that affect cytokine biology, and therefore highlight the complexity of disease, but also shed light on targetable pathways. Exclusively targeting fibrosis may be difficult, however, because of the relatively slow evolution of fibrosis in CD, and the potential adverse effects of inhibiting pathways involved in tissue repair and mucosal healing. Acknowledging these caveats, cytokine-targeted therapy has become the mainstay of treatment for many inflammatory conditions and is being evaluated for fibrotic disorders. The question of whether anti-cytokine therapy will prove useful for intestinal fibrosis is, therefore, acutely relevant. This review will highlight some of the current therapeutics targeting cytokines involved in fibrosis.
Collapse
Affiliation(s)
- Noam Jacob
- F. Widjaja Foundation, Inflammatory Bowel & Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Digestive Diseases, Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Stephan R Targan
- F. Widjaja Foundation, Inflammatory Bowel & Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - David Q Shih
- F. Widjaja Foundation, Inflammatory Bowel & Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
38
|
Jiang F, Chen Q, Huang L, Wang Y, Zhang Z, Meng X, Liu Y, Mao C, Zheng F, Zhang J, Yan H. TNFSF15 Inhibits Blood Retinal Barrier Breakdown Induced by Diabetes. Int J Mol Sci 2016; 17:ijms17050615. [PMID: 27120595 PMCID: PMC4881442 DOI: 10.3390/ijms17050615] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 12/21/2022] Open
Abstract
Tumor necrosis factor superfamily 15 (TNFSF15) is an endogenous neovascularization inhibitor and an important negative regulator of vascular homeostasis. This study aimed to explore the potential role of TNFSF15 in diabetic retinopathy. Vitreous TNFSF15 and VEGF levels in proliferative diabetic retinopathy (PDR) patients were detected by ELISA. Retinal expression of TNFSF15 and the content of tight junction proteins (TJPs) in rats were detected by immunohistochemistry and Western blot, respectively. The blood retinal barrier (BRB) permeability was evaluated using Evans Blue (EB) dye. The TNFSF15/VEGF ratio was decreased in the vitreous fluid of patients with PDR relative to the controls, even though the expression levels of TNFSF15 were higher. TNFSF15 was dramatically decreased one month later after diabetes induction (p < 0.001), and then increased three months later and thereafter. TNFSF15 treatment significantly protected the BRB in the diabetic animals. Diabetes decreased TJPs levels in the retina, and these changes were inhibited by TNFSF15 treatment. Moreover, TNFSF15 decreased activation of VEGF both in mRNA and protein levels caused by diabetes. These results indicate that TNFSF15 is an important inhibitor in the progression of DR and suggest that the regulation of TNFSF15 shows promise for the development of diabetic retinopathy treatment strategies.
Collapse
Affiliation(s)
- Feng Jiang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Qingzhong Chen
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Liming Huang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Ying Wang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Zhuhong Zhang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Xiangda Meng
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Yuanyuan Liu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Chunjie Mao
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Fang Zheng
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Jingkai Zhang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
39
|
Bernardi S, Bossi F, Toffoli B, Fabris B. Roles and Clinical Applications of OPG and TRAIL as Biomarkers in Cardiovascular Disease. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1752854. [PMID: 27200369 PMCID: PMC4856888 DOI: 10.1155/2016/1752854] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/28/2016] [Accepted: 04/05/2016] [Indexed: 12/19/2022]
Abstract
Cardiovascular diseases (CVD) remain the major cause of death and premature disability in Western societies. Assessing the risk of CVD is an important aspect in clinical decision-making. Among the growing number of molecules that are studied for their potential utility as CVD biomarkers, a lot of attention has been focused on osteoprotegerin (OPG) and its ligands, which are receptor activator of nuclear factor κB ligand (RANKL) and TNF-related apoptosis-inducing ligand. Based on the existing literature and on our experience in this field, here we review what the possible roles of OPG and TRAIL in CVD are and their potential utility as CVD biomarkers.
Collapse
Affiliation(s)
- Stella Bernardi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34149 Trieste, Italy
| | - Fleur Bossi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34149 Trieste, Italy
| | - Barbara Toffoli
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34149 Trieste, Italy
| | - Bruno Fabris
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34149 Trieste, Italy
| |
Collapse
|
40
|
Kim JY, Ahn SJ, Baek JM, Yoon KH, Lee MS, Oh J. Ostericum koreanum Reduces LPS-Induced Bone Loss Through Inhibition of Osteoclastogenesis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 43:495-512. [PMID: 25967665 DOI: 10.1142/s0192415x15500317] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The roots of Ostericum koreanum (OK) Maximowicz have traditionally been used to produce an herbal medicine reported to possess anti-inflammatory, anti-oxidant, antimicrobial, and antitumor activities; however, its effect on bone metabolism has not yet been reported. The present study examined the effects of OK extract on lipopolysaccharide (LPS)-induced bone loss in mice by investigating bone structure and the levels of the receptor activator of nuclear factor kappa-B ligand (RANKL) and osteoprotegerin (OPG) in serum and bone marrow fluid (BMF). The effects of OK extract on osteoclastogenesis were also investigated in mouse bone marrow macrophages by examining the formation of tartrate-resistant acid phosphatase (TRAP)-positive cells, the actin ring, and bone resorption activity. OK reduced LPS-induced bone destruction in vivo via a decrease in the RANKL/OPG ratio. Furthermore, it suppressed the formation of TRAP-positive cells and the actin ring, and reduced the bone-resorbing activity of mature osteoclasts. OK also significantly down-regulated the expression of various osteoclast-specific genes. However, it did not affect osteoblast differentiation, or the expression of genes involved in this process. These results demonstrated that OK prevented LPS-induced bone loss by decreasing the RANKL/OPG ratio in serum and BMF, and inhibited osteoclast differentiation and function, suggesting that OK represents a potential therapeutic drug for the treatment of osteoclast-associated bone diseases.
Collapse
Affiliation(s)
- Ju-Young Kim
- Imaging Science-Based Lung and Bone Diseases Research Center, Iksan, Republic of Korea
| | | | | | | | | | | |
Collapse
|
41
|
Ma W, Shao Y, Yang W, Li G, Zhang Y, Zhang M, Zuo C, Chen K, Wang J. Evaluation of (188)Re-labeled NGR-VEGI protein for radioimaging and radiotherapy in mice bearing human fibrosarcoma HT-1080 xenografts. Tumour Biol 2016; 37:9121-9. [PMID: 26768609 DOI: 10.1007/s13277-016-4810-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/06/2016] [Indexed: 11/25/2022] Open
Abstract
Vascular endothelial growth inhibitor (VEGI) is an anti-angiogenic protein, which includes three isoforms: VEGI-174, VEGI-192, and VEGI-251. The NGR (asparagine-glycine-arginine)-containing peptides can specifically bind to CD13 (Aminopeptidase N) receptor which is overexpressed in angiogenic blood vessels and tumor cells. In this study, a novel NGR-VEGI fusion protein was prepared and labeled with (188)Re for radioimaging and radiotherapy in mice bearing human fibrosarcoma HT-1080 xenografts. Single photon emission computerized tomography (SPECT) imaging results revealed that (188)Re-NGR-VEGI exhibits good tumor-to-background contrast in CD13-positive HT-1080 tumor xenografts. The CD13 specificity of (188)Re-NGR-VEGI was further verified by significant reduction of tumor uptake in HT-1080 tumor xenografts with co-injection of the non-radiolabeled NGR-VEGI protein. The biodistribution results demonstrated good tumor-to-muscle ratio (4.98 ± 0.25) of (188)Re-NGR-VEGI at 24 h, which is consistent with the results from SPECT imaging. For radiotherapy, 18.5 MBq of (188)Re-NGR-VEGI showed excellent tumor inhibition effect in HT-1080 tumor xenografts with no observable toxicity, which was confirmed by the tumor size change and hematoxylin and eosin (H&E) staining of major mouse organs. In conclusion, these data demonstrated that (188)Re-NGR-VEGI has the potential as a theranostic agent for CD13-targeted tumor imaging and therapy.
Collapse
Affiliation(s)
- Wenhui Ma
- Department of Nuclear Medicine, Xijing Hospital, The Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC 103, Los Angeles, CA, 90033-9061, USA
| | - Yahui Shao
- Department of Nuclear Medicine, Xijing Hospital, The Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
- Department of Nuclear Medicine, General Hospital of Jinan Military Region, Jinan, Shandong, China
| | - Weidong Yang
- Department of Nuclear Medicine, Xijing Hospital, The Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Guiyu Li
- Department of Nuclear Medicine, Xijing Hospital, The Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Yingqi Zhang
- The State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Mingru Zhang
- Department of Nuclear Medicine, Xijing Hospital, The Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Changjing Zuo
- Department of Nuclear Medicine, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Kai Chen
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC 103, Los Angeles, CA, 90033-9061, USA.
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, The Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
42
|
Amarapurkar P, Rosenblatt JD, Pereira D. Brentuximab: a major advance in treatment of CD30-positive malignancies. Int J Hematol Oncol 2015. [DOI: 10.2217/ijh.15.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Antibody-directed therapies allow greater selectivity in targeting of tumor associated antigens and spare normal cells. Brentuximab vedotin is an anti-CD30 antibody–drug conjugate. It has demonstrated impressive activity in the treatment of refractory and or relapsed Hodgkin's lymphoma, anaplastic large cell lymphoma and other CD30+ lymphoid malignancies. Several ongoing trials are testing the potential use of brentuximab vedotin for treatment of various CD30+ and CD30- malignancies in the setting of high-risk untreated disease. It is being tested in combination with chemotherapy, and testing in combination with immune therapy is also planned. CD30 plays a pivotal role in immune regulation and is also an attractive new target for intervention in the setting of select auto-immune diseases, as well as graft versus host disease.
Collapse
Affiliation(s)
- Pooja Amarapurkar
- Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Joseph D Rosenblatt
- Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, 1475 NW 12th Avenue Miami, FL 33136, USA
| | - Denise Pereira
- Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, 1475 NW 12th Avenue Miami, FL 33136, USA
| |
Collapse
|
43
|
Albarbar B, Dunnill C, Georgopoulos NT. Regulation of cell fate by lymphotoxin (LT) receptor signalling: Functional differences and similarities of the LT system to other TNF superfamily (TNFSF) members. Cytokine Growth Factor Rev 2015; 26:659-71. [DOI: 10.1016/j.cytogfr.2015.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 05/10/2015] [Accepted: 05/13/2015] [Indexed: 12/11/2022]
|
44
|
Tumor Necrosis Factor-like Cytokine TL1A and Its Receptors DR3 and DcR3: Important New Factors in Mucosal Homeostasis and Inflammation. Inflamm Bowel Dis 2015; 21:2441-52. [PMID: 26099067 DOI: 10.1097/mib.0000000000000492] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tumor necrosis factor (TNF)-like cytokine 1A (TL1A) is a member of the TNF superfamily of proteins (TNFSF15), which signals through association with death domain receptor 3 (DR3). Decoy receptor 3 (DcR3) competes with DR3 for TL1A binding and inhibits functional signaling. These proteins are significantly upregulated in inflamed intestinal tissues, and their pathogenetic importance for inflammatory bowel disease (IBD) is suggested by accumulating evidence. TL1A/DR3 induce costimulatory signals to activated lymphocytes, including the gut-specific populations of CD4+CD161+ and CD4+CCR9+ cells, affecting all major effector pathways and inducing the mucosal upregulation of Th1, Th2, and Th17 factors. They may also participate in mucosal homeostasis and defense against pathogens through their effects on the development and function of the recently described innate lymphoid cells. T-regulatory lymphocytes highly express DR3, and they respond to TL1A stimulation also. Mechanistic studies by transgenic expression of TL1A, deletion of TL1A or DR3, and therapeutic blockade by anti-TL1A antibodies all support the critical involvement of the corresponding pathways in the pathogenesis of chronic mucosal inflammation. Wide genome association studies have identified IBD-specific polymorphisms in TNFSF15 gene, which have functional implications and serve as poor prognostic factors. Recently, TL1A blockade in mice was presented as a unique pharmacological treatment for the reversal of established intestinal fibrosis. Finally, TL1A/DR3 signaling seems to critically participate in extraintestinal inflammatory conditions that are frequently associated with IBD as part of the gut-joint-skin-eye axis. These converging lines of evidence make TL1A/DR3 a suitable model for personalized approaches to IBD therapy.
Collapse
|
45
|
Sarvothaman S, Undi RB, Pasupuleti SR, Gutti U, Gutti RK. Apoptosis: role in myeloid cell development. Blood Res 2015; 50:73-9. [PMID: 26157776 PMCID: PMC4486162 DOI: 10.5045/br.2015.50.2.73] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 04/06/2015] [Accepted: 04/29/2015] [Indexed: 01/28/2023] Open
Abstract
Hematopoiesis is the process that generates blood cells in an organism from the pluripotent stem cells. Hematopoietic stem cells are characterized by their ability to undergo self-renewal and differentiation. The self-renewing ability ensures that these pluripotent cells are not depleted from the bone marrow niche. A proper balance between cell death and cell survival is necessary to maintain a homeostatic condition, hence, apoptosis, or programmed cell death, is an essential step in hematopoiesis. Recent studies, however, have introduced a new aspect to this process, citing the significance of the apoptosis mediator, caspase, in cell development and differentiation. Extensive research has been carried out to study the possible role of caspases and other apoptosis related factors in the developmental processes. This review focuses on the various apoptotic factors involved in the development and differentiation of myeloid lineage cells: erythrocytes, megakaryocytes, and macrophages.
Collapse
Affiliation(s)
- Shilpa Sarvothaman
- Stem Cells and Haematological Disorders Laboratory, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Ram Babu Undi
- Stem Cells and Haematological Disorders Laboratory, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Satya Ratan Pasupuleti
- Stem Cells and Haematological Disorders Laboratory, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Usha Gutti
- Department of Biotechnology, GITAM Institute of Science, GITAM University, Visakhapatnam, India
| | - Ravi Kumar Gutti
- Stem Cells and Haematological Disorders Laboratory, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
46
|
Weichhaus M, Chung STM, Connelly L. Osteoprotegerin in breast cancer: beyond bone remodeling. Mol Cancer 2015; 14:117. [PMID: 26054853 PMCID: PMC4460694 DOI: 10.1186/s12943-015-0390-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 05/20/2015] [Indexed: 11/28/2022] Open
Abstract
Osteoprotegerin (OPG) is a secreted protein and member of the Tumor Necrosis Factor (TNF) Receptor superfamily. OPG has been well characterized as a regulator of bone metabolism which acts by blocking osteoclast maturation and preventing bone breakdown. Given this role, early studies on OPG in breast cancer focused on the administration of OPG in order to prevent the osteolysis observed with bone metastases. However OPG is also produced by the breast tumor cells themselves. Research focusing on OPG produced by breast tumor cells has revealed actions of OPG which promote tumor progression. In vitro studies into the role of OPG produced by breast tumor cells have demonstrated that OPG can block TNF-related apoptosis inducing ligand (TRAIL)-mediated apoptosis. Furthermore, in vivo studies show that OPG expression by breast tumors can promote tumor growth and metastasis. In addition it has been shown that OPG stimulates endothelial cell survival and tube formation thus it may indirectly promote breast tumor progression through impacting angiogenesis. This article will present a summary of the data concerning the tumor-promoting effects of OPG in breast cancer.
Collapse
Affiliation(s)
- Michael Weichhaus
- Division of Natural Science and Mathematics, Chaminade University of Honolulu, 3140 Waialae Avenue, Honolulu, HI, 96816, USA.
| | - Stephanie Tsang Mui Chung
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI, 96720, USA.
| | - Linda Connelly
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI, 96720, USA.
| |
Collapse
|
47
|
Yamanegi K, Kawabe M, Futani H, Nishiura H, Yamada N, Kato-Kogoe N, Kishimoto H, Yoshiya S, Nakasho K. Sodium valproate, a histone deacetylase inhibitor, modulates the vascular endothelial growth inhibitor-mediated cell death in human osteosarcoma and vascular endothelial cells. Int J Oncol 2015; 46:1994-2002. [PMID: 25778932 DOI: 10.3892/ijo.2015.2924] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 02/12/2015] [Indexed: 11/06/2022] Open
Abstract
The level of vascular endothelial growth inhibitor (VEGI) has been reported to be negatively associated with neovascularization in malignant tumors. The soluble form of VEGI is a potent anti-angiogenic factor due to its effects in inhibiting endothelial cell proliferation. This inhibition is mediated by death receptor 3 (DR3), which contains a death domain in its cytoplasmic tail capable of inducing apoptosis that can be subsequently blocked by decoy receptor 3 (DcR3). We investigated the effects of sodium valproate (VPA) and trichostatin A (TSA), histone deacetylase inhibitors, on the expression of VEGI and its related receptors in human osteosarcoma (OS) cell lines and human microvascular endothelial (HMVE) cells. Consequently, treatment with VPA and TSA increased the VEGI and DR3 expression levels without inducing DcR3 production in the OS cell lines. In contrast, the effect on the HMVE cells was limited, with no evidence of growth inhibition or an increase in the DR3 and DcR3 expression. However, VPA-induced soluble VEGI in the OS cell culture medium markedly inhibited the vascular tube formation of HMVE cells, while VEGI overexpression resulted in enhanced OS cell death. Taken together, the HDAC inhibitor has anti-angiogenesis and antitumor activities that mediate soluble VEGI/DR3-induced apoptosis via both autocrine and paracrine pathways. This study indicates that the HDAC inhibitor may be exploited as a therapeutic strategy modulating the soluble VEGI/DR3 pathway in osteosarcoma patients.
Collapse
Affiliation(s)
- Koji Yamanegi
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Mutsuki Kawabe
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Hiroyuki Futani
- Department of Orthopedic Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Hiroshi Nishiura
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Naoko Yamada
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Nahoko Kato-Kogoe
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Hiromitsu Kishimoto
- Department of Oral and Maxillofacial Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Shinichi Yoshiya
- Department of Orthopedic Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Keiji Nakasho
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| |
Collapse
|
48
|
Siakavellas SI, Sfikakis PP, Bamias G. The TL1A/DR3/DcR3 pathway in autoimmune rheumatic diseases. Semin Arthritis Rheum 2015; 45:1-8. [PMID: 25887448 DOI: 10.1016/j.semarthrit.2015.02.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 01/28/2015] [Accepted: 02/16/2015] [Indexed: 12/13/2022]
Abstract
IMPORTANCE TNF-like cytokine 1A (TL1A) and its receptors, death receptor 3 (DR3) and decoy receptor 3 (DcR3) are members of the TNF and TNF receptor superfamilies of proteins, respectively. They constitute a cytokine system that actively interferes with the regulation of immune responses and may participate in the pathogenesis of autoimmune diseases. OBJECTIVES This review aims to present the current knowledge on the role of the TL1A/DR3/DcR3 system in the pathophysiology of autoimmune rheumatic diseases, with a focus on rheumatoid arthritis (RA). METHODS An extensive literature search was performed in the PubMed database using the following keywords: TL1A, death receptor 3, DR3, decoy receptor 3, DcR3, TNFSF15, TNFRSF25, and TNFSF6B. Studies were assessed and selected in view of their relevance to autoimmune rheumatic diseases. CONCLUSION The TL1A/DR3/DcR3 axis is a novel immune pathway that participates in the pathogenesis of a variety of autoimmune rheumatic diseases. These molecules may be promising therapeutic targets for inflammatory arthritis.
Collapse
Affiliation(s)
- Spyros I Siakavellas
- Academic Department of Gastroenterology, Laikon Hospital, Kapodistrian University of Athens, 17 Agiou Thoma St, Athens 11527, Greece
| | - Petros P Sfikakis
- First Department of Propaedeutic and Internal Medicine, Laikon Hospital, Kapodistrian University of Athens, Athens, Greece
| | - Giorgos Bamias
- Academic Department of Gastroenterology, Laikon Hospital, Kapodistrian University of Athens, 17 Agiou Thoma St, Athens 11527, Greece.
| |
Collapse
|
49
|
Tougaard P, Skov S, Pedersen AE, Krych L, Nielsen DS, Bahl MI, Christensen EG, Licht TR, Poulsen SS, Metzdorff SB, Hansen AK, Hansen CHF. TL1A regulates TCRγδ+intraepithelial lymphocytes and gut microbial composition. Eur J Immunol 2014; 45:865-75. [DOI: 10.1002/eji.201444528] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 09/22/2014] [Accepted: 11/13/2014] [Indexed: 01/03/2023]
Affiliation(s)
- Peter Tougaard
- Department of Veterinary Disease Biology; Section of Experimental Animal Models; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - S. Skov
- Department of Veterinary Disease Biology; Section of Experimental Animal Models; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - A. E. Pedersen
- Department of International Health; Immunology and Microbiology; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - L. Krych
- Department of Food Science; Faculty of Science; University of Copenhagen; Copenhagen Denmark
| | - D. S. Nielsen
- Department of Food Science; Faculty of Science; University of Copenhagen; Copenhagen Denmark
| | - M. I. Bahl
- National Food Institute; Division of Microbiology and Risk Assessment; Technical University of Denmark; Moerkhoej Denmark
| | - E. G. Christensen
- National Food Institute; Division of Microbiology and Risk Assessment; Technical University of Denmark; Moerkhoej Denmark
| | - T. R. Licht
- National Food Institute; Division of Microbiology and Risk Assessment; Technical University of Denmark; Moerkhoej Denmark
| | - S. S. Poulsen
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - S. B. Metzdorff
- Department of Veterinary Disease Biology; Section of Experimental Animal Models; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - A. K. Hansen
- Department of Veterinary Disease Biology; Section of Experimental Animal Models; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - C. H. F. Hansen
- Department of Veterinary Disease Biology; Section of Experimental Animal Models; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
50
|
Kwan JSH, Hsu YH, Cheung CL, Dupuis J, Saint-Pierre A, Eriksson J, Handelman SK, Aragaki A, Karasik D, Pramstaller PP, Kooperberg C, Lacroix AZ, Larson MG, Lau KS, Lorentzon M, Pichler I, Sham PC, Taliun D, Vandenput L, Kiel DP, Hicks AA, Jackson RD, Ohlsson C, Benjamin EJ, Kung AWC. Meta-analysis of genome-wide association studies identifies two loci associated with circulating osteoprotegerin levels. Hum Mol Genet 2014; 23:6684-93. [PMID: 25080503 PMCID: PMC4240210 DOI: 10.1093/hmg/ddu386] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 07/14/2014] [Accepted: 07/21/2014] [Indexed: 01/08/2023] Open
Abstract
Osteoprotegerin (OPG) is involved in bone homeostasis and tumor cell survival. Circulating OPG levels are also important biomarkers of various clinical traits, such as cancers and atherosclerosis. OPG levels were measured in serum or in plasma. In a meta-analysis of genome-wide association studies in up to 10 336 individuals from European and Asian origin, we discovered that variants >100 kb upstream of the TNFRSF11B gene encoding OPG and another new locus on chromosome 17q11.2 were significantly associated with OPG variation. We also identified a suggestive locus on chromosome 14q21.2 associated with the trait. Moreover, we estimated that over half of the heritability of OPG levels could be explained by all variants examined in our study. Our findings provide further insight into the genetic regulation of circulating OPG levels.
Collapse
Affiliation(s)
| | - Yi-Hsiang Hsu
- Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA, Program of Quantitative Genomics, Harvard School of Public Health, Boston, MA, USA, BROAD Institute of the MIT and Harvard, Cambridge, MA, USA
| | | | - Josée Dupuis
- Framingham Heart Study of the National, Heart, Lung, and Blood Institute and Boston University, Framingham, MA, USA, Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Aude Saint-Pierre
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy - Affiliated Institute of the University of Lübeck, Lübeck, Germany, INSERM U1078, Etablissement Français du Sang, Brest, France
| | - Joel Eriksson
- Centre for Bone and Arthritis Research, Departments of Internal Medicine and Geriatrics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Aaron Aragaki
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The Ohio State University, Columbus, OH, USA
| | - David Karasik
- Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA
| | - Peter P Pramstaller
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy - Affiliated Institute of the University of Lübeck, Lübeck, Germany, Department of Neurology, General Central Hospital, Bolzano, Italy, Department of Neurology, University of Lübeck, Lübeck, Germany
| | | | - Andrea Z Lacroix
- Department of Preventive Medicine, University of California San Diego, San Diego, CA, USA
| | - Martin G Larson
- Framingham Heart Study of the National, Heart, Lung, and Blood Institute and Boston University, Framingham, MA, USA, Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | | | - Mattias Lorentzon
- Centre for Bone and Arthritis Research, Departments of Internal Medicine and Geriatrics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Irene Pichler
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy - Affiliated Institute of the University of Lübeck, Lübeck, Germany
| | - Pak C Sham
- Department of Psychiatry and Centre for Genomic Sciences, University of Hong Kong, Pokfulam, Hong Kong
| | - Daniel Taliun
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy - Affiliated Institute of the University of Lübeck, Lübeck, Germany
| | - Liesbeth Vandenput
- Centre for Bone and Arthritis Research, Departments of Internal Medicine and Geriatrics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Douglas P Kiel
- Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA, BROAD Institute of the MIT and Harvard, Cambridge, MA, USA, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Andrew A Hicks
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy - Affiliated Institute of the University of Lübeck, Lübeck, Germany
| | - Rebecca D Jackson
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The Ohio State University, Columbus, OH, USA
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Departments of Internal Medicine and Geriatrics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Emelia J Benjamin
- Framingham Heart Study of the National, Heart, Lung, and Blood Institute and Boston University, Framingham, MA, USA, Department of Medicine, Boston University School of Medicine, Boston, MA, USA and
| | | |
Collapse
|