1
|
Oranger A, Colaianni G, Ingravallo G, Scarcella VS, Faienza MF, Grano M, Colucci S, Brunetti G. LIGHT/TNFSF14 Affects Adipose Tissue Phenotype. Int J Mol Sci 2024; 25:716. [PMID: 38255789 PMCID: PMC10815871 DOI: 10.3390/ijms25020716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/22/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
LIGHT/TNFSF14 is linked to several signaling pathways as a crucial member of a larger immunoregulatory network. It is primarily expressed in inflammatory effector cells, and high levels of LIGHT have been reported in obesity. Thus, with the aim of deepening the knowledge of the role of LIGHT on adipose tissue phenotype, we studied wild-type (WT), Tnfsf14-/-, Rag-/- and Rag-/Tnfsf14- (DKO) mice fed a normal diet (ND) or high-fat diet (HFD). Our results show that, although there is no significant weight gain between the mice with different genotypes, it is significant within each of them. We also detected an increase in visceral White Adipose Tissue (vWAT) weight in all mice fed HFD, together with the lowest levels of vWAT weight in Tnfsf14-/- and DKO mice fed ND with respect to the other strain. Inguinal WAT (iWAT) weight is significantly affected by genotype and HFD. The least amount of iWAT was detected in DKO mice fed ND. Histological analysis of vWAT showed that both the genotype and the diet significantly affect the adipocyte area, whereas the number is affected only by the genotype. In iWAT, the genotype and the diet significantly affect mean adipocyte area and number; interestingly, the area with the least adipocyte was detected in DKO mice fed ND, suggesting a potential browning effect due to the simultaneous lack of mature lymphocytes and LIGHT. Consistently, Uncoupling Protein 1 (UCP1) staining of iWAT demonstrated that few positive brown adipocytes appeared in DKO mice. Furthermore, LIGHT deficiency is associated with greater levels of UCP1, highlighting the lack of its expression in Rag-/- mice. Liver examination showed that all mice fed HFD had a steatotic liver, but it was particularly evident for DKO mice. In conclusion, our study demonstrates that the adipose tissue phenotype is affected by LIGHT levels but also much more by mature lymphocytes.
Collapse
Affiliation(s)
- Angela Oranger
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (A.O.); (G.C.); (M.G.)
| | - Graziana Colaianni
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (A.O.); (G.C.); (M.G.)
| | - Giuseppe Ingravallo
- Section of Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy; (G.I.); (V.S.S.)
| | - Vincenza Sara Scarcella
- Section of Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy; (G.I.); (V.S.S.)
| | - Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Maria Grano
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (A.O.); (G.C.); (M.G.)
| | - Silvia Colucci
- Department of Translational Biomedicine and Neuroscience, University of Bari, 70124 Bari, Italy;
| | - Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy
| |
Collapse
|
2
|
Lu C, Zhang X, Schardey J, Wirth U, Heinrich K, Massiminio L, Cavestro GM, Neumann J, Bazhin AV, Werner J, Kühn F. Molecular characteristics of microsatellite stable early-onset colorectal cancer as predictors of prognosis and immunotherapeutic response. NPJ Precis Oncol 2023; 7:63. [PMID: 37393364 DOI: 10.1038/s41698-023-00414-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 06/15/2023] [Indexed: 07/03/2023] Open
Abstract
The incidence of early-onset colorectal cancer (EO-CRC, in patients younger than 50) is increasing worldwide. The specific gene signatures in EO-CRC patients are largely unknown. Since EO-CRC with microsatellite instability is frequently associated with Lynch syndrome, we aimed to comprehensively characterize the tumor microenvironment (TME) and gene expression profiles of EO-CRC with microsatellite stable (MSS-EO-CRC). Here, we demonstrated that MSS-EO-CRC has a similar pattern of tumor-infiltrating immune cells, immunotherapeutic responses, consensus molecular subtypes, and prognosis as late-onset CRC with MSS (MSS-LO-CRC). 133 differential expressed genes were identified as unique gene signatures of MSS-EO-CRC. Moreover, we established a risk score, which was positively associated with PD-L1 expression and could reflect both the level of tumor-infiltrating immune cells and the prognosis of MSS-EO-CRC patients. Application of this score on the anti-PD-L1 treatment cohort demonstrated that the low-risk score group has significant therapeutic advantages and clinical benefits. In addition, candidate driver genes were identified in the different-sidedness of MSS-EO-CRC patients. Altogether, MSS-EO-CRC exhibits distinct molecular profiles that differ from MSS-LO-CRC even though they have a similar TME characterization and survival pattern. Our risk score appears to be robust enough to predict prognosis and immunotherapeutic response and therefore could help to optimize the treatment of MSS-EO-CRC.
Collapse
Affiliation(s)
- Can Lu
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377, Munich, Germany
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention (Ministry of Education), The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for CANCER & Cancer Center of Zhejiang University, Hangzhou, China
| | - Xiaopeng Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital and Institute, Beijing, China
- Institute of Laboratory Medicine, University Hospital of LMU Munich, Munich, Germany
| | - Josefine Schardey
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377, Munich, Germany
| | - Ulrich Wirth
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377, Munich, Germany
| | - Kathrin Heinrich
- Department of Medicine III, University Hospital, Ludwig-Maximilians-University Munich, 81377, Munich, Germany
| | - Luca Massiminio
- Experimental Gastroenterology Laboratory, Gastroenterology and Endoscopy Department, San Raffaele Scientific Institute, Milano, Italy
| | - Giulia Martina Cavestro
- Experimental Gastroenterology Laboratory, Gastroenterology and Endoscopy Department, San Raffaele Scientific Institute, Milano, Italy
| | - Jens Neumann
- Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Partner Site Munich, Munich, Germany
| | - Alexandr V Bazhin
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Partner Site Munich, Munich, Germany
| | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Partner Site Munich, Munich, Germany
| | - Florian Kühn
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, 81377, Munich, Germany.
- Bavarian Cancer Research Center (BZKF), Partner Site Munich, Munich, Germany.
| |
Collapse
|
3
|
Mekchay S, Pothakam N, Norseeda W, Supakankul P, Teltathum T, Liu G, Naraballobh W, Khamlor T, Sringarm K, Krutmuang P. Association of IFNA16 and TNFRSF19 Polymorphisms with Intramuscular Fat Content and Fatty Acid Composition in Pigs. BIOLOGY 2022; 11:109. [PMID: 35053107 PMCID: PMC8773020 DOI: 10.3390/biology11010109] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/18/2021] [Accepted: 01/07/2022] [Indexed: 06/14/2023]
Abstract
Interferon-alpha-16 (IFNA16) and tumor necrosis factor receptor superfamily member 19 (TNFRSF19) are cytokines that may play a role in adipogenesis and fatness. Single nucleotide polymorphisms (SNPs) of the porcine IFNA16 and TNFRSF19 genes were verified and their association with intramuscular fat (IMF) content and fatty acid (FA) composition were evaluated in commercial crossbred pigs. Two non-synonymous SNPs of the porcine IFNA16 c.413G > A and TNFRSF19 c.860G > C loci were detected in commercial crossbred pigs. The porcine IFNA16 c.413G >A polymorphism was significantly associated with stearic acid, total saturated FAs (SFAs), and the ratio of monounsaturated FAs (MUFAs) to SFAs (p < 0.05). Furthermore, the porcine TNFRSF19 c.860G > C polymorphism was found to be significantly associated with IMF content and arachidic acid levels (p < 0.05). The results revealed that porcine IFNA16 and TNFRSF19 polymorphisms are related to IMF content and/or FA composition and affirmed the importance of these cytokine genes as potential candidate genes for lipid deposition and FA composition in the muscle tissue of pigs.
Collapse
Affiliation(s)
- Supamit Mekchay
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand; (N.P.); (W.N.); (T.K.); (K.S.)
- Center of Excellence on Agricultural Biotechnology: (AG-BIO/MHESI), Bangkok 10900, Thailand
- Cluster of Research and Development of Pharmaceutical and Natural Products Innovation for Human or Animal, Chiang Mai University, Chiang Mai 50200, Thailand
- Innovative Agriculture Research Center, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Nanthana Pothakam
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand; (N.P.); (W.N.); (T.K.); (K.S.)
- Center of Excellence on Agricultural Biotechnology: (AG-BIO/MHESI), Bangkok 10900, Thailand
- Veterinary, Conservation and Research Section, Animal Management Division, Chiang Mai Night Safari, Chiang Mai 50230, Thailand
| | - Worrarak Norseeda
- Department of Agriculture, Faculty of Agricultural Technology, Lampang Rajabhat University, Lampang 52100, Thailand;
| | - Pantaporn Supakankul
- Division of Animal Science, School of Agriculture and Natural Resources, University of Phayao, Phayao 56000, Thailand;
| | - Tawatchai Teltathum
- Mae Hong Son Livestock Research and Breeding Center, Mae Hong Son 58000, Thailand;
| | - Guisheng Liu
- Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan 430064, China;
- Hubei Key Lab for Animal Embryo Engineering and Molecular Breeding, Wuhan 430064, China
| | - Watcharapong Naraballobh
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand; (N.P.); (W.N.); (T.K.); (K.S.)
- Innovative Agriculture Research Center, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Trisadee Khamlor
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand; (N.P.); (W.N.); (T.K.); (K.S.)
| | - Korawan Sringarm
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand; (N.P.); (W.N.); (T.K.); (K.S.)
- Cluster of Research and Development of Pharmaceutical and Natural Products Innovation for Human or Animal, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Patcharin Krutmuang
- Innovative Agriculture Research Center, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand;
- Department of Entomology and Plant Pathology, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
4
|
Tumor Necrosis Receptor Superfamily Interact with Fusion and Fission of Mitochondria of Adipose Tissue in Obese Patients without Type 2 Diabetes. Biomedicines 2021; 9:biomedicines9091260. [PMID: 34572446 PMCID: PMC8470627 DOI: 10.3390/biomedicines9091260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 11/17/2022] Open
Abstract
Interactions between receptors and ligands of the tumor necrosis factor superfamily (TNFSF) provide costimulatory signals that control the survival, proliferation, differentiation, and effector function of immune cells. All components of the TNF superfamily are associated with NF-kB functions that are not limited to cell death and may promote survival in the face of adipose tissue inflammation in obesity. Inflammation dysfunction of mitochondria is a key factor associated with insulin resistance in obesity. The aim of the study was to analyze the relationship of soluble forms of receptors and ligands of the TNF superfamily in blood plasma with mitochondrial dynamics in adipose tissue (greater omentum (GO) and subcutaneous adipose tissue (Sat)) of obese patients with and without type 2 diabetes mellitus (T2DM). Increased plasma sTNF-R1, sTNF-R2, sTNFRSF8 receptors, and ligands TNFSF12, TNFSF13, TNFSF13B are characteristic of obese patients without T2DM. The TNF-a levels in blood plasma were associated with a decrease in MFN2 gene expression in GO and IL-10 in blood plasma. The TNFSF12 levels contributed to a decrease in glucose levels, a decrease in BMI, and an increase in IL-10 levels by influencing the MFN2 gene expression in GO, which supports mitochondrial fusion.
Collapse
|
5
|
Song J, Deng T. The Adipocyte and Adaptive Immunity. Front Immunol 2020; 11:593058. [PMID: 33329579 PMCID: PMC7728694 DOI: 10.3389/fimmu.2020.593058] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 10/27/2020] [Indexed: 12/21/2022] Open
Abstract
Not only do Adipocytes have energy storage and endocrine functions, but they also play an immunological role. Adipocytes are involved in adaptive immunity to mediate the pathological processes of a variety of chronic inflammatory diseases and autoimmune syndromes. The adaptive immune response consists of T cell-mediated cellular immunity and B cell-mediated humoral immunity. Obese adipocytes overexpress MHC class II molecules and costimulators to act as antigen-presenting cells (APCs) and promote the activation of CD4+ T cells. In addition, various adipokines secreted by adipocytes regulate the proliferation and differentiation of T cells. Adipokines are also involved in B cell generation, development, activation, and antibody production. Therefore, adipocytes play an important role in B cell-mediated adaptive immunity. This review describes how adipocytes participate in adaptive immunity from the perspective of T cells and B cells, and discusses their role in the pathogenesis of various diseases.
Collapse
Affiliation(s)
- Jianfeng Song
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Tuo Deng
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.,Clinical Immunology Center, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
6
|
Szymczak M, Ziętkiewicz S, Kuncewicz K, Rodziewicz-Motowidło S, Orlikowska M. Expression, purification, and efficient refolding of the extracellular domain of Escherichia coli-expressed signaling receptor herpesvirus entry mediator. Protein Expr Purif 2019; 164:105450. [PMID: 31299214 DOI: 10.1016/j.pep.2019.105450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 07/05/2019] [Accepted: 07/08/2019] [Indexed: 01/21/2023]
Abstract
Herpesvirus entry mediator (HVEM), a member of the TNF-receptor superfamily, plays an important role in the regulation of the immune system. It forms a complex with ligands and can either activate or inhibit the response of the immune system. Furthermore, HVEM can exhibit pro-inflammatory or anti-inflammatory effects in many human diseases. Therefore, understanding the mechanism underlying the interaction of HVEM with other receptors is extremely important to design small therapeutic molecules that can stimulate the response of the immune system. In this study, we attempted to develop the most efficient method for the expression and purification of the extracellular domain of HVEM using Escherichia coli. The soluble fraction constituted only a small portion of the E. coli-expressed protein, whereas majority of the protein was found to be accumulated in the insoluble fraction. Three different protein refolding methods were analyzed: dialysis, dilution, and using chromatographic column. The oligomeric state of the protein was determined by characterizing the obtained fractions using analytical size exclusion chromatography. All the obtained fractions were tested for their ability to form a complex with B- and T-lymphocyte attenuator using enzyme-linked immunosorbent assay. The results of this study provide crucial information regarding the production of HVEM protein in a robust, well-established, and convenient heterologous expression system using E. coli as a host. In addition, it allows for the selection of the most effective method for appropriate refolding of HVEM protein, which gets accumulated in the insoluble fraction.
Collapse
Affiliation(s)
- Marta Szymczak
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, 80-308, Gdansk, Wita Stwosza 63, Poland
| | - Szymon Ziętkiewicz
- Department of Molecular and Cellular Biology, Intercollegiate Faculty of Biotechnology, University of Gdansk, 80-822, Gdansk, Kladki 24, Poland
| | - Katarzyna Kuncewicz
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, 80-308, Gdansk, Wita Stwosza 63, Poland
| | - Sylwia Rodziewicz-Motowidło
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, 80-308, Gdansk, Wita Stwosza 63, Poland
| | - Marta Orlikowska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, 80-308, Gdansk, Wita Stwosza 63, Poland.
| |
Collapse
|
7
|
|
8
|
Is early life exposure to polyomaviruses and herpesviruses associated with obesity indices and metabolic traits in childhood? Int J Obes (Lond) 2018; 42:1590-1601. [PMID: 29445241 DOI: 10.1038/s41366-018-0017-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 12/20/2017] [Accepted: 12/27/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Evidence for an infectious origin of obesity is emerging. We explored whether common viruses were associated with obesity and metabolic traits. METHODS We used cross-sectional (n = 674) and prospective (n = 440) data from children participating at the 4 and 6 years of age follow-up in the Rhea birth cohort. Presence of IgG antibodies to ten polyomaviruses (BKPyV, JCPyV, KIPyV, WUPyV, HPyV6, HPyV7, TSPyV, MCPyV, HPyV9, and HPyV10) and four herpesviruses (EBV, CMV, HSV-1, and HSV-2) were measured at age 4. Body mass index, waist circumference, and skinfold thickness were measured at age 4 and 6. Data on serum lipids, leptin, and adiponectin were also available. Multivariable linear regression models were used to explore the associations. RESULTS At 4 years of age, seroprevalence to polyomaviruses ranged from 21.0% for HPyV9 to 82.0% for HPyV10. Seroprevalence for EBV, CMV, HSV-1, and HSV-2 was 53.0%, 26.0%, 3.6%, and 1.5% respectively. BKPyV seropositivity was associated with lower BMI SD score at age 4 [-0.21 (95% CI: -0.39, -0.03)] and 6 [-0.27 (95% CI:-0.48, -0.05)], waist circumference at age 4 [-1.12 cm (95% CI: -2.10, -0.15)] and 6 [-1.73 cm (95% CI: -3.33, -0.12)], sum of four skinfolds [-2.97 mm (95% CI: -5.70, -0.24)], and leptin levels at age 4 [ratio of geometric means, 0.83 (95% CI: 0.70, 0.98)]. CMV seropositivity was associated with higher BMI SD score at age 4 [0.28 (95% CI: 0.11, 0.45)] and 6 [0.24 (95% CI: 0.03, 0.45)] and sum of four skinfolds at age 6 [4.75 mm (95% CI: 0.67, 8.83)]. Having "2-3 herpesviruses infections" (versus "0 herpesvirus infections") was associated with higher BMI SD score [0.32, (95% CI: 0.12, 0.53)], waist circumference [1.22 cm (95% CI: 0.13, 2.31)], and sum of four skinfolds [3.26 mm (95% CI: 0.18, 6.35)] at age 4. Polyomaviruses burden was not associated with outcomes. CONCLUSIONS A higher herpesviruses burden and CMV seropositivity were associated with obesity traits in childhood.
Collapse
|
9
|
Saunders BM, Rudnicka C, Filipovska A, Davies S, Ward N, Hricova J, Schlaich MP, Matthews VB. Shining LIGHT on the metabolic role of the cytokine TNFSF14 and the implications on hepatic IL-6 production. Immunol Cell Biol 2017; 96:41-53. [DOI: 10.1111/imcb.1002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/20/2017] [Accepted: 08/24/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Bernadette M Saunders
- School of Life Sciences; Faculty of Science; University of Technology Sydney; New South Wales Australia
- Tuberculosis Research Program; Centenary Institute; Newtown New South Wales Australia
| | - Caroline Rudnicka
- Research Centre; Royal Perth Hospital; Perth Western Australia Australia
| | - Aleksandra Filipovska
- Harry Perkins Institute of Medical Research; Nedlands Western Australia Australia
- School of Molecular Sciences; University of Western Australia; Nedlands Western Australia Australia
| | - Stefan Davies
- Harry Perkins Institute of Medical Research; Nedlands Western Australia Australia
| | - Natalie Ward
- School of Medicine; University of Western Australia; Perth Western Australia Australia
- Curtin Health and Innovation Research Institute; Curtin University; Perth Western Australia Australia
| | - Jana Hricova
- Royal Perth Hospital Unit; Dobney Hypertension Centre; School of Biomedical Sciences; University of Western Australia; Perth Western Australia Australia
| | - Markus P Schlaich
- Royal Perth Hospital Unit; Dobney Hypertension Centre; School of Biomedical Sciences; University of Western Australia; Perth Western Australia Australia
- Department of Cardiology and Department of Nephrology; Royal Perth Hospital; Perth Western Australia Australia
| | - Vance B Matthews
- Royal Perth Hospital Unit; Dobney Hypertension Centre; School of Biomedical Sciences; University of Western Australia; Perth Western Australia Australia
| |
Collapse
|
10
|
Yuan X, Gu Y, Lai X, Gu Q. LIGHT is increased in patients with coronary disease and regulates inflammatory response and lipid metabolism in oxLDL-induced THP-1 macrophages. Biochem Biophys Res Commun 2017. [PMID: 28642135 DOI: 10.1016/j.bbrc.2017.06.110] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Inflammation is critical for the progression of hyperlipidemia. Although the exact mechanism through which inflammation affects hyperlipidemia is not very clear, evidence suggests that the tumor necrosis factor superfamily member 14 (TNFSF14/LIGHT)LIGHT might regulate lipid metabolism. In this study we investigated the expression of LIGHT in patients with different stages of coronary disease. The expression of lipid metabolism-related enzymes and inflammation-related proteins were further explored in oxidized low-density lipoproteins (oxLDL)-induced THP-1 macrophages. We found that LIGHT is highly expressed and companied with severe inflammations in patients with coronary disease. LIGHT significantly enhanced inflammation response in oxLDL-induced THP-1 macrophages. We further demonstrated that LIGHT markedly decreased the levels of lipolytic genes and increased the expressions of lipogenic genes in oxLDL-induced THP-1 macrophages. In addition, our results showed that LIGHT exerts its pro-inflammatory and pro-lipogenesis roles through activating nuclear factor-kappa B (NF-κB) signaling pathway. Taken together our study has demonstrated that LIGHT NF-κB-dependently exacerbates inflammation response and promotes lipid accumulation, and provided a new potential target for treatment of hyperlipidemia-related disease.
Collapse
Affiliation(s)
- Xiaomei Yuan
- Heart Failure Center, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Yonglin Gu
- Department of Cardiology, Guang'an People's Hospital, Guang'an, 638099, China
| | - Xiaoyu Lai
- Department of Cardiology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Qing Gu
- Department of Geriatric Medicine, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, 610072, China.
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Obesity is a multifactorial disease that is now endemic throughout most of the world. Although addressing proximate causes of obesity (excess energy intake and reduced energy expenditure) have been longstanding global health priorities, the problem has continued to worsen at the global level. RECENT FINDINGS Numerous microbial agents cause obesity in various experimental models-a phenomena known as infectobesity. Several of the same agents alter metabolic function in human cells and are associated with human obesity or metabolic dysfunction in humans. We address the evidence for a role in the genesis of obesity for viral agents in five broad categories: adenoviridae, herpesviridae, phages, transmissible spongiform encephalopathies (slow virus), and other encephalitides and hepatitides. Despite the importance of this topic area, there are many persistent knowledge gaps that need to be resolved. We discuss factors motivating further research and recommend that future infectobesity investigation should be more comprehensive, leveraged, interventional, and patient-centered.
Collapse
Affiliation(s)
- Jameson D Voss
- United States Air Force School of Aerospace Medicine, Epidemiology Consult Service Division, 2510 Fifth Street, Building 840, Wright-Patterson AFB, OH, 45433, USA
| | - Nikhil V Dhurandhar
- Department of Nutritional Sciences, Texas Tech University, Box 41270, Lubbock, TX, 79409-1240, USA.
| |
Collapse
|
12
|
Halvorsen B, Santilli F, Scholz H, Sahraoui A, Gulseth HL, Wium C, Lattanzio S, Formoso G, Di Fulvio P, Otterdal K, Retterstøl K, Holven KB, Gregersen I, Stavik B, Bjerkeli V, Michelsen AE, Ueland T, Liani R, Davi G, Aukrust P. LIGHT/TNFSF14 is increased in patients with type 2 diabetes mellitus and promotes islet cell dysfunction and endothelial cell inflammation in vitro. Diabetologia 2016; 59:2134-44. [PMID: 27421726 PMCID: PMC5016561 DOI: 10.1007/s00125-016-4036-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 06/08/2016] [Indexed: 12/02/2022]
Abstract
AIMS/HYPOTHESIS Activation of inflammatory pathways is involved in the pathogenesis of type 2 diabetes mellitus. On the basis of its role in vascular inflammation and in metabolic disorders, we hypothesised that the TNF superfamily (TNFSF) member 14 (LIGHT/TNFSF14) could be involved in the pathogenesis of type 2 diabetes mellitus. METHODS Plasma levels of LIGHT were measured in two cohorts of type 2 diabetes mellitus patients (191 Italian and 40 Norwegian). Human pancreatic islet cells and arterial endothelial cells were used to explore regulation and relevant effects of LIGHT in vitro. RESULTS Our major findings were: (1) in both diabetic cohorts, plasma levels of LIGHT were significantly raised compared with sex- and age-matched healthy controls (n = 32); (2) enhanced release from activated platelets seems to be an important contributor to the raised LIGHT levels in type 2 diabetes mellitus; (3) in human pancreatic islet cells, inflammatory cytokines increased the release of LIGHT and upregulated mRNA and protein levels of the LIGHT receptors lymphotoxin β receptor (LTβR) and TNF receptor superfamily member 14 (HVEM/TNFRSF14); (4) in these cells, LIGHT attenuated the insulin release in response to high glucose at least partly via pro-apoptotic effects; and (5) in human arterial endothelial cells, glucose boosted inflammatory response to LIGHT, accompanied by an upregulation of mRNA levels of HVEM (also known as TNFRSF14) and LTβR (also known as LTBR). CONCLUSIONS/INTERPRETATION Our findings show that patients with type 2 diabetes mellitus are characterised by increased plasma LIGHT levels. Our in vitro findings suggest that LIGHT may contribute to the progression of type 2 diabetes mellitus by attenuating insulin secretion in pancreatic islet cells and by contributing to vascular inflammation.
Collapse
Affiliation(s)
- Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway.
- K.G. Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | | | - Hanne Scholz
- Institute for Surgical Research, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section for Transplantation Surgery, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Afaf Sahraoui
- Institute for Surgical Research, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section for Transplantation Surgery, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Hanne L Gulseth
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Cecilie Wium
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Lipid Clinic, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | | | - Gloria Formoso
- Center of Excellence on Aging, University of Chieti, Chieti, Italy
| | | | - Kari Otterdal
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kjetil Retterstøl
- Lipid Clinic, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Kirsten B Holven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- National Advisory Unit on Familial Hypercholesterolemia, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ida Gregersen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Benedicte Stavik
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
| | - Vigdis Bjerkeli
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
- K.G. Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Rossella Liani
- Center of Excellence on Aging, University of Chieti, Chieti, Italy
| | - Giovanni Davi
- Center of Excellence on Aging, University of Chieti, Chieti, Italy
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
- K.G. Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
- K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway
| |
Collapse
|
13
|
Voss JD, Atkinson RL, Dhurandhar NV. Role of adenoviruses in obesity. Rev Med Virol 2015; 25:379-87. [PMID: 26352001 DOI: 10.1002/rmv.1852] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/13/2015] [Accepted: 07/15/2015] [Indexed: 12/30/2022]
Abstract
UNLABELLED Five human adenovirus subtypes, Ad5, Ad9, Ad31, Ad36, and Ad37, and a non-human adenovirus, SMAM1, are linked to increased adiposity in vitro or in vivo. Experimental infection with Ad5, Ad36, and Ad37 produced excess adiposity or weight gain in animals. Ad9 and Ad31 increase fat storage in tissue culture but are not associated with animal or human obesity. Ad36 is the most extensively studied adipogenic adenovirus and is correlated with some measure of overweight/obesity in humans from multiple countries. The correlation is strongest and most consistent in children, but some studies have been negative in both children and adults. About 30% of overweight/obese children and adults and about 15-20% of lean individuals have Ad36 antibodies in epidemiologic studies. The mechanisms of action of Ad36 are due to the early gene 4, open reading frame 1 (E4-ORF1). Blocking E4-ORF1 with siRNA prevents the effects of Ad36, and transfection of lentivirus with E4-ORF1 reproduces the Ad36 effects. Increased adiposity is caused by stimulation of at least three pathways by Ad36. Cell membrane glucose receptors are increased via the Ras pathway, leading to increased intracellular glucose. Fatty acid synthase is increased, which converts the glucose to fatty acids. Finally, peroxisome proliferator-activated receptor-γ is increased, resulting in differentiation of adult stem cells into adipocytes. CONCLUSIONS several adenoviruses increase adiposity in animals and are associated with obesity in humans. There are critical gaps in the literature needing further investigation including evaluation of other adenovirus subtypes and better research designs to improve the strength of causal inferences.
Collapse
Affiliation(s)
- Jameson D Voss
- Epidemiology Consult Division, United States Air Force School of Aerospace Medicine, Wright-Patterson AFB, OH, USA
| | - Richard L Atkinson
- Virginia Obesity Research Institute, Richmond, VA, USA.,Virginia Commonwealth University, Richmond, VA, USA
| | | |
Collapse
|
14
|
|
15
|
Otterdal K, Haukeland JW, Yndestad A, Dahl TB, Holm S, Segers FM, Gladhaug IP, Konopski Z, Damås JK, Halvorsen B, Aukrust P. Increased Serum Levels of LIGHT/TNFSF14 in Nonalcoholic Fatty Liver Disease: Possible Role in Hepatic Inflammation. Clin Transl Gastroenterol 2015; 6:e95. [PMID: 26133108 PMCID: PMC4816254 DOI: 10.1038/ctg.2015.23] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 03/18/2015] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES The tumor necrosis factor superfamily member 14, LIGHT (homologous to lymphotoxin, exhibits inducible expression, and competes with HSV glycoprotein D for herpes virus entry mediator (HVEM), a receptor expressed by T lymphocytes), has been involved in various autoimmune disorders and has been shown to influence hepatic lipid metabolism. We hypothesized that LIGHT could also have a pathogenic role in nonalcoholic fatty liver disease (NAFLD). METHODS Serum levels of LIGHT in NAFLD patients and control subjects, as well as LIGHT and interleukin (IL)-8 released from Huh7 (human hepatoma cell line) hepatocytes, were determined by enzyme-linked immunosorbent assay. The mRNA expression of LIGHT in the liver tissue and mRNA levels of LIGHT and IL-8 in Huh7 hepatocytes were assessed by real-time quantitative reverse transcription-PCR. RESULTS (i) Serum levels of LIGHT were significantly elevated in NAFLD patients (n=66) as compared with healthy controls (n=16), with no differences between simple steatosis (n=34) and nonalcoholic steatohepatitis (NASH) (n=32). (ii) Within the liver, NAFLD patients (n=14) had significantly increased mRNA levels of the two LIGHT receptors, herpes virus entry mediator and lymphotoxin β receptor (LTβR), as compared with controls (n=7), with no difference between simple steatosis (n=8) and NASH (n=6). (iii) LIGHT markedly increased the release of IL-8 in Huh7 hepatocytes in a time- and dose-dependent manner. (iv) The reactive oxygen species (ROS) H2O2 (hydrogen peroxide) enhanced the LIGHT-mediated release of IL-8 in Huh7 hepatocytes. CONCLUSION We show increased levels of LIGHT and its two membrane-bound receptors in NAFLD, potentially promoting hepatic inflammation through ROS interaction. Our findings should encourage further studies on the role of LIGHT in NAFLD development and progression.
Collapse
Affiliation(s)
- Kari Otterdal
- 1] Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway [2] Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Arne Yndestad
- 1] Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway [2] Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway [3] K.G. Jebsen Inflammatory Research Center, Oslo, Norway
| | - Tuva B Dahl
- 1] Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway [2] Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sverre Holm
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Filip M Segers
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ivar P Gladhaug
- 1] Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway [2] Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Zbigniew Konopski
- Department of Gastroenterology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Jan Kristian Damås
- 1] Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, St Olavs Hospital, Trondheim, Norway [2] Department of Infectious Diseases, St Olavs Hospital, Trondheim, Norway
| | - Bente Halvorsen
- 1] Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway [2] Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway [3] K.G. Jebsen Inflammatory Research Center, Oslo, Norway
| | - Pål Aukrust
- 1] Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway [2] Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway [3] K.G. Jebsen Inflammatory Research Center, Oslo, Norway [4] Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
16
|
Šedý J, Bekiaris V, Ware CF. Tumor necrosis factor superfamily in innate immunity and inflammation. Cold Spring Harb Perspect Biol 2014; 7:a016279. [PMID: 25524549 DOI: 10.1101/cshperspect.a016279] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The tumor necrosis factor superfamily (TNFSF) and its corresponding receptor superfamily (TNFRSF) form communication pathways required for developmental, homeostatic, and stimulus-responsive processes in vivo. Although this receptor-ligand system operates between many different cell types and organ systems, many of these proteins play specific roles in immune system function. The TNFSF and TNFRSF proteins lymphotoxins, LIGHT (homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for herpes virus entry mediator [HVEM], a receptor expressed by T lymphocytes), lymphotoxin-β receptor (LT-βR), and HVEM are used by embryonic and adult innate lymphocytes to promote the development and homeostasis of lymphoid organs. Lymphotoxin-expressing innate-acting B cells construct microenvironments in lymphoid organs that restrict pathogen spread and initiate interferon defenses. Recent results illustrate how the communication networks formed among these cytokines and the coreceptors B and T lymphocyte attenuator (BTLA) and CD160 both inhibit and activate innate lymphoid cells (ILCs), innate γδ T cells, and natural killer (NK) cells. Understanding the role of TNFSF/TNFRSF and interacting proteins in innate cells will likely reveal avenues for future therapeutics for human disease.
Collapse
Affiliation(s)
- John Šedý
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| | - Vasileios Bekiaris
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| | - Carl F Ware
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| |
Collapse
|
17
|
Choi EK, Kim WK, Sul OJ, Park YK, Kim ES, Suh JH, Yu R, Choi HS. TNFRSF14 deficiency protects against ovariectomy-induced adipose tissue inflammation. J Endocrinol 2014; 220:25-33. [PMID: 24287621 DOI: 10.1530/joe-13-0341] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
To elucidate the role of tumor necrosis factor receptor superfamily member 14 (TNFRSF14) in metabolic disturbance due to loss of ovarian function, ovariectomy (OVX) was performed in TNFRSF 14-knockout mice. OVX increased fat mass and infiltration of highly inflammatory CD11c cells in the adipose tissue (AT), which was analyzed by flow cytometry, and resulted in disturbance of glucose metabolism, whereas TNFRSF14 deficiency attenuated these effects. TNFRSF14 deficiency decreased recruitment of CD11c-expressing cells in AT and reduced the polarization of bone marrow-derived macrophages to M1. Upon engagement of LIGHT, a TNFRSF14 ligand, TNFRSF14 enhanced the expression of CD11c via generation of reactive oxygen species, suggesting a role of TNFRSF14 as a redox modulator. TNFRSF14 participated in OVX-induced AT inflammation via upregulation of CD11c, resulting in metabolic perturbation. TNFRSF14 could be used as a therapeutic target for the treatment of postmenopausal syndrome by reducing AT inflammation.
Collapse
Affiliation(s)
- Eun-Kyung Choi
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea Department of Endocrinology, Ulsan University Hospital, Ulsan 682-714, Korea Department of Pathology, Ulsan University Hospital, Ulsan 682-714, Korea Department of Food Science and Nutrition, University of Ulsan, Ulsan 680-749, Korea
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Bjordahl RL, Steidl C, Gascoyne RD, Ware CF. Lymphotoxin network pathways shape the tumor microenvironment. Curr Opin Immunol 2013; 25:222-9. [PMID: 23339845 DOI: 10.1016/j.coi.2013.01.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Accepted: 01/02/2013] [Indexed: 12/13/2022]
Abstract
Accumulating evidence indicates that Lymphotoxin (LT)-β related cytokines directly contribute to the phenotype of cancer cells and alter the tumor microenvironment. Lymphotoxins are part of a cytokine network well known in controlling the development and homeostasis of secondary lymphoid organs. In the adult, the LT network takes on the responsibility of generating inflammatory microenvironments that control innate and adaptive immune responses involved in host defense. This review provides a perspective of the emerging evidence implicating the LT Network in the development and progression of various cancers including lymphoma. Redirecting the LT Network to alter tumor microenvironments may provide a specific approach to therapeutically target tumor-permissive microenvironments and cancer progression.
Collapse
Affiliation(s)
- Ryan L Bjordahl
- Infectious and Inflammatory Diseases Center, Sanford Burnham Medical Research Institute, 10901N. Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
19
|
Ware CF. Protein therapeutics targeted at the TNF superfamily. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2013; 66:51-80. [PMID: 23433455 DOI: 10.1016/b978-0-12-404717-4.00002-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Protein-based drugs with their unequivocal specificity achieved the long sought milestone of selectively disrupting cytokine pathways to alleviate ongoing inflammation. Tumor necrosis factor (TNF), a member of the superfamily of cytokines involved in regulating immune and inflammatory processes, provides an exemplary model of protein therapeutics. Antibody and receptor-based inhibitors of TNF modify inflammation leading to dramatic improvement in patients with certain autoimmune diseases. Collectively, the structure, specificity and valence of these protein-based drugs provide direct evidence that the essential mechanism of action is antagonism of the ligand-receptor interaction. Accumulating clinical knowledge regarding TNF inhibitors also provide insights into the mechanisms involved in different autoimmune diseases. Experience in the development of an arsenal of biologics directed at TNF has additionally contributed to knowledge toward overcoming the challenges of protein drugs, which include production, delivery, antigenicity and pharmacodynamics. Dramatic clinical outcomes with TNF inhibitors are driving investigation and development of biologics toward other members of the TNF superfamily to selectively alter functional properties of the immune system.
Collapse
Affiliation(s)
- Carl F Ware
- Laboratory of Molecular Immunology, Infectious and Inflammatory Diseases Center, Sanford Burnham Medical Research Institute, La Jolla, CA, USA.
| |
Collapse
|
20
|
Croft M, Duan W, Choi H, Eun SY, Madireddi S, Mehta A. TNF superfamily in inflammatory disease: translating basic insights. Trends Immunol 2011; 33:144-52. [PMID: 22169337 DOI: 10.1016/j.it.2011.10.004] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 10/20/2011] [Accepted: 10/20/2011] [Indexed: 12/17/2022]
Abstract
The tumor necrosis factor (TNF) and TNF receptor superfamilies (TNFSF and TNFRSF) consist of approximately 50 membrane and soluble proteins that can modulate cellular function. Most of these molecules are expressed by or can target cells of the immune system, and they have a wide range of actions including promoting cellular differentiation, survival, and production of inflammatory cytokines and chemokines. Emerging data show that TNFSF ligand-receptor signaling pathways are active in inflammatory and autoimmune disease. Furthermore, several genetic polymorphisms in TNFSF and TNFRSF associate with susceptibility to developing disease. Here, we examine recent data regarding the potential of these molecules as targets for therapy of autoimmune and inflammatory disease.
Collapse
Affiliation(s)
- Michael Croft
- La Jolla Institute for Allergy and Immunology, Division of Immune Regulation, 9420 Athena Circle, La Jolla, CA 92037, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Steinberg M, Cheung TC, Ware CF. The signaling networks of the herpesvirus entry mediator (TNFRSF14) in immune regulation. Immunol Rev 2011; 244:169-87. [PMID: 22017438 PMCID: PMC3381650 DOI: 10.1111/j.1600-065x.2011.01064.x] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The tumor necrosis factor (TNF) receptor superfamily member herpesvirus entry mediator (HVEM) (TNFRSF14) regulates T-cell immune responses by activating both inflammatory and inhibitory signaling pathways. HVEM acts as both a receptor for the canonical TNF-related ligands, LIGHT [lymphotoxin-like, exhibits inducible expression, and competes with herpes simplex virus glycoprotein D for HVEM, a receptor expressed on T lymphocytes] and lymphotoxin-α, and as a ligand for the immunoglobulin superfamily proteins BTLA (B and T lymphocyte attenuator) and CD160, a feature distinguishing HVEM from other immune regulatory molecules. The ability of HVEM to interact with multiple ligands in distinct configurations creates a functionally diverse set of intrinsic and bidirectional signaling pathways that control both inflammatory and inhibitory responses. The HVEM system is integrated into the larger LTβR and TNFR network through extensive shared ligand and receptor usage. Experimental mouse models and human diseases indicate that dysregulation of HVEM network may contribute to autoimmune pathogenesis, making it an attractive target for drug intervention.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Autoimmunity
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/immunology
- GPI-Linked Proteins/metabolism
- Gene Expression/immunology
- Herpes Simplex/immunology
- Herpes Simplex/metabolism
- Herpes Simplex/virology
- Herpesvirus 1, Human/immunology
- Humans
- Immunity, Innate
- Lymphocyte Activation
- Lymphotoxin beta Receptor/genetics
- Lymphotoxin beta Receptor/immunology
- Lymphotoxin beta Receptor/metabolism
- Lymphotoxin-alpha/genetics
- Lymphotoxin-alpha/immunology
- Lymphotoxin-alpha/metabolism
- Mice
- Mice, Knockout
- Protein Binding/immunology
- Receptor Cross-Talk/immunology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Receptors, Tumor Necrosis Factor, Member 14/genetics
- Receptors, Tumor Necrosis Factor, Member 14/immunology
- Receptors, Tumor Necrosis Factor, Member 14/metabolism
- Signal Transduction/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
- Tumor Necrosis Factor Ligand Superfamily Member 14/immunology
- Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
- Tumor Necrosis Factor-alpha/metabolism
- Viral Envelope Proteins/immunology
- Viral Envelope Proteins/metabolism
Collapse
Affiliation(s)
| | | | - Carl F. Ware
- Laboratory of Molecular Immunology, Center for Infectious and Inflammatory Diseases, Sanford|Burnham Medical Research Institute, La Jolla, CA, USA
| |
Collapse
|
22
|
Kim HJ, Kim HM, Kim CS, Jeong CS, Choi HS, Kawada T, Kim BS, Yu R. HVEM-deficient mice fed a high-fat diet are protected from adipose tissue inflammation and glucose intolerance. FEBS Lett 2011; 585:2285-90. [PMID: 21679708 DOI: 10.1016/j.febslet.2011.05.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 05/12/2011] [Accepted: 05/26/2011] [Indexed: 01/13/2023]
Abstract
HVEM is a member of the TNF receptor superfamily that plays a role in the development of various inflammatory diseases. In this study, we show that HVEM deficiency attenuates adipose tissue inflammatory responses and glucose intolerance in diet-induced obesity. Feeding a high-fat diet (HFD) to HVEM-deficient mice elicited a reduction in the number of macrophages and T cells infiltrated into adipose tissue. Proinflammatory cytokine levels in the adipose tissue decreased in HFD-fed HVEM-deficient mice, while levels of the anti-inflammatory cytokine IL-10 increased. Moreover, glucose intolerance and insulin sensitivity were markedly improved in the HFD-fed HVEM-deficient mice. These findings indicate that HVEM may be a useful target for combating obesity-induced inflammatory responses and insulin resistance.
Collapse
Affiliation(s)
- Ha-Jung Kim
- Department of Food Science and Nutrition, University of Ulsan, Ulsan, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Karjala Z, Neal D, Rohrer J. Association between HSV1 seropositivity and obesity: data from the National Health and Nutritional Examination Survey, 2007-2008. PLoS One 2011; 6:e19092. [PMID: 21589933 PMCID: PMC3092767 DOI: 10.1371/journal.pone.0019092] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 03/28/2011] [Indexed: 12/18/2022] Open
Abstract
Background Herpes simplex virus (HSV) is among the most common sexually transmitted pathogens in the United States and worldwide. HSV has a high incidence of undetected cases. In addition, there is no treatment, and there is a lack of knowledge why disparities among populations exist. Research studies suggest that fat tissue may participate in body's immune responses, and the impact of obesity on susceptibility to HSV1 infection is not clear. The purpose of this study was to examine whether obesity is a risk factor for HSV1 infection using a large sample from the general population. Methods/Principal Findings This cross-sectional study used data from the National Health and Examination and Nutritional Examination Survey (NHANES) from 2007–2008. Variables, gender, age, race/ethnicity, marital status, education, poverty level, and diabetes represented potential confounders and were included in analyses. The two-tailed Pearson's chi square, student's t test, and a multiple logistic regression analysis were applied to evaluate associations using a significance value of p≤0.05. Adjusted odds ratios with 95% confidence interval represented the degree of these associations. The prevalence of HSV1 infection in US population between 20 and 49 years old was 60.3% (n = 1,536). In this study, having a BMI classified as the obese group (BMI 30–39.9) was significantly associated with HSV1 infection before [unadjusted OR = 1.74 (95% CI 1.20–2.51), p = 0.006] and after controlling for socio-demographic factors [adjusted OR = 1.50 (95%CI 1.06–2.13)], p = 0.026]. This association was stronger than three already established risk factors of age, female gender, and poverty level. Conclusions/Significance This study provides evidence that obesity may play a role in the susceptibility to HSV1 infection. Findings from this study suggest that obesity should be considered when designing preventive measures for HSV1 infection. These results may also explain why some people acquire HSV1 infections and some do not. Further, these findings may justify an increased emphasis on the control and prevention of HSV1 transmission and other pathogens in overweight and obese populations.
Collapse
Affiliation(s)
- Zuzana Karjala
- Walden University, Minneapolis, Minnesota, United States of America.
| | | | | |
Collapse
|
24
|
Kim HM, Jeong CS, Choi HS, Kawada T, Yu R. LIGHT/TNFSF14 enhances adipose tissue inflammatory responses through its interaction with HVEM. FEBS Lett 2011; 585:579-84. [PMID: 21236258 DOI: 10.1016/j.febslet.2011.01.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Revised: 01/05/2011] [Accepted: 01/06/2011] [Indexed: 11/19/2022]
Abstract
Obesity-induced adipose tissue inflammation is characterized by increased macrophage infiltration and cytokine production, and is associated with metabolic disorders. LIGHT/TNFSF14, a member of the TNF superfamily, plays a role in the development of various inflammatory diseases. The purpose of this study was to examine the involvement of soluble LIGHT (sLIGHT) in obesity-induced adipose tissue inflammatory responses. LIGHT gene expression on macrophages/adipocytes was upregulated by treatment with obesity-related factors. sLIGHT displayed chemotactic activity for macrophages and T cells, and enhanced inflammatory cytokine release from macrophages, adipocytes, and adipose tissue-derived SVF cells. The sLIGHT-induced inflammatory responses were blunted by neutralizing anti-HVEM antibody or knockout of HVEM, a receptor for sLIGHT. These findings indicate that sLIGHT enhances adipose tissue inflammatory responses through its interaction with HVEM.
Collapse
MESH Headings
- Adipocytes/metabolism
- Adipose Tissue, White/cytology
- Adipose Tissue, White/metabolism
- Animals
- Cell Line
- Cells, Cultured
- Chemotaxis
- Culture Media, Conditioned
- Cytokines/metabolism
- Dietary Fats/administration & dosage
- Gene Expression Regulation
- Inflammation Mediators/metabolism
- Macrophages/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Obesity/metabolism
- Oxidative Stress
- RNA, Messenger/metabolism
- Receptors, Tumor Necrosis Factor, Member 14/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Member 14/genetics
- Receptors, Tumor Necrosis Factor, Member 14/metabolism
- Stromal Cells/metabolism
- Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
- Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
Collapse
Affiliation(s)
- Hong-Min Kim
- Department of Biological Science, University of Ulsan, Nam-ku, Ulsan, South Korea
| | | | | | | | | |
Collapse
|