101
|
Algate K, Haynes DR, Bartold PM, Crotti TN, Cantley MD. The effects of tumour necrosis factor-α on bone cells involved in periodontal alveolar bone loss; osteoclasts, osteoblasts and osteocytes. J Periodontal Res 2015; 51:549-66. [DOI: 10.1111/jre.12339] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2015] [Indexed: 12/22/2022]
Affiliation(s)
- K. Algate
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - D. R. Haynes
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - P. M. Bartold
- School of Dentistry; University of Adelaide; Adelaide SA Australia
| | - T. N. Crotti
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - M. D. Cantley
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
- Myeloma Research Laboratory; University of Adelaide; Adelaide SA Australia
| |
Collapse
|
102
|
Noguchi S, Mori T, Igase M, Mizuno T. A novel apoptosis-inducing mechanism of 5-aza-2′-deoxycitidine in melanoma cells: Demethylation of TNF-α and activation of FOXO1. Cancer Lett 2015; 369:344-53. [DOI: 10.1016/j.canlet.2015.08.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/19/2015] [Accepted: 08/24/2015] [Indexed: 10/23/2022]
|
103
|
Pozniak PD, Darbinyan A, Khalili K. TNF-α/TNFR2 Regulatory Axis Stimulates EphB2-Mediated Neuroregeneration Via Activation of NF-κB. J Cell Physiol 2015; 231:1237-48. [PMID: 26492598 DOI: 10.1002/jcp.25219] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/20/2015] [Indexed: 12/11/2022]
Abstract
HIV-1 infected individuals are at high risk of developing HIV-associated neurocognitive disorders (HAND) as HIV infection leads to neuronal injury and synaptic loss in the central nervous system (CNS). The neurotoxic effects of HIV-1 are primarily a result of viral replication leading to the production of inflammatory chemokines and cytokines, including TNF-α. Given an important role of TNF-α in regulating synaptic plasticity, we investigated the effects of TNF-α on the development of neuronal processes after mechanical injury, and we showed that TNF-α treatment stimulates the regrowth of neuronal processes. To investigate transcriptional effects of TNF-α on synaptic plasticity, we analyzed both human neurosphere and isolated neuronal cultures for the regulation of genes central to synaptic alterations during learning and memory. TNF-α treatment upregulated Ephrin receptor B2 (EphB2), which is strongly involved in dendritic arborization and synaptic integrity. TNF-α strongly activates the NF-κB pathway, therefore, we propose that TNF-α-induced neurite regrowth occurs primarily through EphB2 signaling via stimulation of NF-κB. EphB2 promoter activity increased with TNF-α treatment and overexpression of NF-κB. Direct binding of NF-κB to the EphB2 promoter occurred in the ChIP assay, and site-directed mutagenesis identified binding sites involved in TNF-α-induced EphB2 activation. TNF-α induction of EphB2 was determined to occur specifically through TNF-α receptor 2 (TNFR2) activation in human primary fetal neurons. Our observations provide a new avenue for the investigation on the impact of TNF-α in the context of HIV-1 neuronal cell damage as well as providing a potential therapeutic target in TNFR2 activation of EphB2.
Collapse
Affiliation(s)
- Paul D Pozniak
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Armine Darbinyan
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania.,Division of Neuropathology, Department of Pathology, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
104
|
Ham B, Wang N, D'Costa Z, Fernandez MC, Bourdeau F, Auguste P, Illemann M, Eefsen RL, Høyer-Hansen G, Vainer B, Evrard M, Gao ZH, Brodt P. TNF Receptor-2 Facilitates an Immunosuppressive Microenvironment in the Liver to Promote the Colonization and Growth of Hepatic Metastases. Cancer Res 2015; 75:5235-47. [DOI: 10.1158/0008-5472.can-14-3173] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 10/07/2015] [Indexed: 11/16/2022]
|
105
|
Webb KC, Tung R, Winterfield LS, Gottlieb AB, Eby JM, Henning SW, Le Poole IC. Tumour necrosis factor-α inhibition can stabilize disease in progressive vitiligo. Br J Dermatol 2015; 173:641-50. [PMID: 26149498 PMCID: PMC4583813 DOI: 10.1111/bjd.14016] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2015] [Indexed: 12/21/2022]
Abstract
Tumour necrosis factor (TNF)-α, a proinflammatory cytokine central to many autoimmune diseases, has been implicated in the depigmentation process in vitiligo. We review its role in vitiligo by exploring its pro- and anti-inflammatory properties and examine the effects of blocking its actions with TNF-α antagonist therapeutics in reports available in the literature. We found that TNF-α inhibition halts disease progression in patients with progressive vitiligo but that, paradoxically, treatment can be associated with de novo vitiligo development in some patients when used for other autoimmune conditions, particularly when using adalimumab and infliximab. These studies reinforce the importance of stating appropriate outcomes measures, as most pilot trials propose to measure repigmentation, whereas halting depigmentation is commonly overlooked as a measure of success. We conclude that TNF-α inhibition has proven useful for patients with progressive vitiligo, where TNF-α inhibition is able to quash cytotoxic T-cell-mediated melanocyte destruction. However, a lingering concern for initiating de novo disease will likely prevent more widespread application of TNF inhibitors to treat vitiligo.
Collapse
Affiliation(s)
- K C Webb
- Department of Dermatology, Loyola University Stritch School of Medicine, 2160 South First Ave, Maywood, IL, U.S.A
| | - R Tung
- Department of Dermatology, Loyola University Stritch School of Medicine, 2160 South First Ave, Maywood, IL, U.S.A
| | - L S Winterfield
- Department of Dermatology, Loyola University Stritch School of Medicine, 2160 South First Ave, Maywood, IL, U.S.A
| | - A B Gottlieb
- Department of Dermatology, Tufts University Medical Center, Boston, MA, U.S.A
| | - J M Eby
- Oncology Research Institute, Loyola University Chicago, IL, U.S.A
| | - S W Henning
- Oncology Research Institute, Loyola University Chicago, IL, U.S.A
| | - I C Le Poole
- Departments of Pathology, Microbiology and Immunology, Loyola University Stritch School of Medicine, 2160 South First Ave, Maywood, IL, U.S.A
| |
Collapse
|
106
|
Ban L, Kuhtreiber W, Butterworth J, Okubo Y, Vanamee ÉS, Faustman DL. Strategic internal covalent cross-linking of TNF produces a stable TNF trimer with improved TNFR2 signaling. MOLECULAR AND CELLULAR THERAPIES 2015; 3:7. [PMID: 26266038 PMCID: PMC4531505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/17/2015] [Indexed: 11/21/2023]
Abstract
BACKGROUND Soluble TNF superfamily (TNFSF) ligands are less stable and less active than their transmembrane (tm) analogues. This is a problem for the therapeutic use of recombinant TNFSF ligands in diverse diseases including cancer and autoimmunity. Creating TNFSF ligand analogues with improved targeting of their respective receptors is important for research and therapeutic purposes. FINDINGS Covalent internal cross-linking of TNF monomers by double mutations, S95C/G148C, results in stable trimers with improved TNFR2 function. The resulting mutein induced the selective death of autoreactive CD8 T cells in type-1 diabetic patients and demonstrates targeted proliferation and expansion of human CD4 Tregs. CONCLUSIONS Stable TNF trimers, created by internal covalent cross-linking, show improved signaling. The high structural homology within the TNF superfamily provides an opportunity to extend internal cross-linking to other TNF superfamily proteins to produce active trimers with improved stability and receptor signaling, and with potential applications for cancer, autoimmunity, infections, and transplantation.
Collapse
Affiliation(s)
- Liqin Ban
- Immunobiology Laboratory, Harvard Medical School and Massachusetts General Hospital, Boston, MA USA
| | - Willem Kuhtreiber
- Immunobiology Laboratory, Harvard Medical School and Massachusetts General Hospital, Boston, MA USA
| | - John Butterworth
- Immunobiology Laboratory, Harvard Medical School and Massachusetts General Hospital, Boston, MA USA
| | - Yoshiaki Okubo
- Immunobiology Laboratory, Harvard Medical School and Massachusetts General Hospital, Boston, MA USA
| | - Éva S. Vanamee
- Immunobiology Laboratory, Harvard Medical School and Massachusetts General Hospital, Boston, MA USA
| | - Denise L. Faustman
- Immunobiology Laboratory, Harvard Medical School and Massachusetts General Hospital, Boston, MA USA
| |
Collapse
|
107
|
Ban L, Kuhtreiber W, Butterworth J, Okubo Y, Vanamee ÉS, Faustman DL. Strategic internal covalent cross-linking of TNF produces a stable TNF trimer with improved TNFR2 signaling. MOLECULAR AND CELLULAR THERAPIES 2015; 3:7. [PMID: 26266038 PMCID: PMC4531505 DOI: 10.1186/s40591-015-0044-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/17/2015] [Indexed: 11/10/2022]
Abstract
Background Soluble TNF superfamily (TNFSF) ligands are less stable and less active than their transmembrane (tm) analogues. This is a problem for the therapeutic use of recombinant TNFSF ligands in diverse diseases including cancer and autoimmunity. Creating TNFSF ligand analogues with improved targeting of their respective receptors is important for research and therapeutic purposes. Findings Covalent internal cross-linking of TNF monomers by double mutations, S95C/G148C, results in stable trimers with improved TNFR2 function. The resulting mutein induced the selective death of autoreactive CD8 T cells in type-1 diabetic patients and demonstrates targeted proliferation and expansion of human CD4 Tregs. Conclusions Stable TNF trimers, created by internal covalent cross-linking, show improved signaling. The high structural homology within the TNF superfamily provides an opportunity to extend internal cross-linking to other TNF superfamily proteins to produce active trimers with improved stability and receptor signaling, and with potential applications for cancer, autoimmunity, infections, and transplantation. Electronic supplementary material The online version of this article (doi:10.1186/s40591-015-0044-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liqin Ban
- Immunobiology Laboratory, Harvard Medical School and Massachusetts General Hospital, Boston, MA USA
| | - Willem Kuhtreiber
- Immunobiology Laboratory, Harvard Medical School and Massachusetts General Hospital, Boston, MA USA
| | - John Butterworth
- Immunobiology Laboratory, Harvard Medical School and Massachusetts General Hospital, Boston, MA USA
| | - Yoshiaki Okubo
- Immunobiology Laboratory, Harvard Medical School and Massachusetts General Hospital, Boston, MA USA
| | - Éva S Vanamee
- Immunobiology Laboratory, Harvard Medical School and Massachusetts General Hospital, Boston, MA USA
| | - Denise L Faustman
- Immunobiology Laboratory, Harvard Medical School and Massachusetts General Hospital, Boston, MA USA
| |
Collapse
|
108
|
Peterson MJ, Thompson DK, Pieper CF, Morey MC, Kraus VB, Kraus WE, Sullivan P, Fillenbaum G, Cohen HJ. A Novel Analytic Technique to Measure Associations Between Circulating Biomarkers and Physical Performance Across the Adult Life Span. J Gerontol A Biol Sci Med Sci 2015; 71:196-202. [PMID: 25745025 DOI: 10.1093/gerona/glv007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 01/16/2015] [Indexed: 12/20/2022] Open
Abstract
Understanding associations between circulating biomarkers and physical performance across the adult life span could aid in better describing mechanistic pathways leading to disability. We hypothesized that high concentrations of circulating biomarkers would be associated with lower functioning across study populations representing the adult life span. The data were from four intervention and two observational studies with ages ranging 22-89 years. Biomarkers assayed included inflammatory, coagulation, and endothelial function markers. Physical performance was measured either by VO2peak (studies of young and middle-aged adults) or usual gait speed (studies of older adults). Partialled (by age, body mass index, race, and sex) and weighted common correlations were calculated between biomarkers and physical performance. Homogeneity of the associations was also assessed. Interleukin-6 (weighted r = -.22), tumor necrosis factor receptor 2 (weighted r = -.19), D-dimer (weighted r = -.16), tumor necrosis factor receptor 1 (weighted r = -.15), granulocyte colony-stimulating factor (weighted r = -.14), and tumor necrosis factor alpha (weighted r = -.10) were all significantly inversely correlated with physical performance (p < .05). All significant correlations were homogeneous across studies. In summary, we observed consistent inverse associations between six circulating biomarkers and objective measures of physical performance. These results suggest that these serum biomarkers may be broadly applicable for detection, trajectory, and treatment monitoring of physical function across the life span or possibly for midlife predictors of functionally deleterious conditions.
Collapse
Affiliation(s)
| | - Dana K Thompson
- Department of Medicine, Duke University, Durham, North Carolina
| | - Carl F Pieper
- Center for the Study of Aging and Human Development/Claude D. Pepper Older Adults Independence Center and Department of Biometry and Bioinformatics, Duke University Medical Center, Durham, North Carolina
| | | | - Virginia B Kraus
- Center for the Study of Aging and Human Development/Claude D. Pepper Older Adults Independence Center and Department of Medicine, Duke University, Durham, North Carolina
| | - William E Kraus
- Center for the Study of Aging and Human Development/Claude D. Pepper Older Adults Independence Center and Department of Medicine, Duke University, Durham, North Carolina
| | - Patrick Sullivan
- Department of Medicine, Duke University, Durham, North Carolina. Geriatric Research, Education and Clinical Center, Veteran's Affairs Medical Center, Durham, North Carolina
| | - Gerda Fillenbaum
- Center for the Study of Aging and Human Development/Claude D. Pepper Older Adults Independence Center and
| | | |
Collapse
|
109
|
TNFR1 and TNFR2 Expression and Induction on Human Treg Cells from Type 1 Diabetic Subjects. Antibodies (Basel) 2015. [DOI: 10.3390/antib4010034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
110
|
Burkly LC. Regulation of Tissue Responses: The TWEAK/Fn14 Pathway and Other TNF/TNFR Superfamily Members That Activate Non-Canonical NFκB Signaling. Front Immunol 2015; 6:92. [PMID: 25784914 PMCID: PMC4345838 DOI: 10.3389/fimmu.2015.00092] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 02/17/2015] [Indexed: 12/29/2022] Open
Affiliation(s)
- Linda C Burkly
- Department of Immunology, Biogen Idec, Inc. , Cambridge, MA , USA
| |
Collapse
|
111
|
Kitagawa M, Shiozaki A, Ichikawa D, Nakashima S, Kosuga T, Konishi H, Komatsu S, Fujiwara H, Okamoto K, Otsuji E. Tumor necrosis factor-α-induced apoptosis of gastric cancer MKN28 cells: accelerated degradation of the inhibitor of apoptosis family members. Arch Biochem Biophys 2014; 566:43-8. [PMID: 25513960 DOI: 10.1016/j.abb.2014.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 11/09/2014] [Accepted: 12/04/2014] [Indexed: 11/28/2022]
Abstract
The role of the inhibitor of apoptosis (IAP) family members in tumor necrosis factor-α (TNF-α)-induced apoptosis of human gastric cancer MKN28 cells was explored. TNF-α induced up-regulation of cIAP2, whereas cycloheximide (CHX) induced down-regulation of XIAP and survivin. Degradation of cIAP1 and XIAP, but not survivin, was accelerated by co-treatment of cells with TNF-α and CHX, and TNF-α-induced up-regulation of cIAP2 was inhibited by BMS-345541 (NF-κB inhibitor). Treatment of MKN28 cells with TNF-α plus CHX induced degradation of survivin and activation of caspase-8 and -3, followed by degradation of cIAP1 and XIAP and apoptosis. Proteasome inhibitors (MG132 and epoxomicin) suppressed TNF-α plus CHX-induced degradation of survivin, cIAP1, and XIAP as well as apoptosis. A caspase inhibitor (z-VAD-fmk) suppressed TNF-α plus CHX-induced apoptosis, but allowed degradation of survivin, cIAP1 and XIAP. TNF-α receptor 1 and 2 were expressed on MKN28 cells. The magnitude of apoptosis induced by TNF-α plus BMS-345541 was much less than that induced by TNF-α plus CHX. These findings suggest that TNF-α plus CHX-induced apoptosis of gastric cancer MKN28 cells may be caused by accelerated degradation of the IAP family members (survivin, cIAP1, and XIAP), in addition to inhibition of NF-κB-dependent synthesis of anti-apoptotic molecules.
Collapse
Affiliation(s)
- Maki Kitagawa
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan.
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan.
| | - Daisuke Ichikawa
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Shingo Nakashima
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Toshiyuki Kosuga
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Kazuma Okamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
112
|
Sasi SP, Rahimi L, Yan X, Silver M, Qin G, Losordo DW, Kishore R, Goukassian DA. Genetic deletion of TNFR2 augments inflammatory response and blunts satellite-cell-mediated recovery response in a hind limb ischemia model. FASEB J 2014; 29:1208-19. [PMID: 25466901 DOI: 10.1096/fj.14-249813] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 11/12/2014] [Indexed: 01/09/2023]
Abstract
We have previously shown that TNF-tumor necrosis factor receptor-2/p75 (TNFR2/p75) signaling plays a critical role in ischemia-induced neovascularization in skeletal muscle and heart tissues. To determine the role of TNF-TNFR2/p75 signaling in ischemia-induced inflammation and muscle regeneration, we subjected wild-type (WT) and TNFR2/p75 knockout (p75KO) mice to hind limb ischemia (HLI) surgery. Ischemia induced significant and long-lasting inflammation associated with considerable decrease in satellite-cell activation in p75KO muscle tissue up to 10 d after HLI surgery. To determine the possible additive negative roles of tissue aging and the absence of TNFR2/p75, either in the tissue or in the bone marrow (BM), we generated 2 chimeric BM transplantation (BMT) models where both young green fluorescent protein (GFP)-positive p75KO and WT BM-derived cells were transplanted into adult p75KO mice. HLI surgery was performed 1 mo after BMT, after confirming complete engraftment of the recipient BM with GFP donor cells. In adult p75KO with the WT-BMT, proliferative (Ki67(+)) cells were detected only by d 28 and were exclusively GFP(+), suggesting significantly delayed contribution of young WT-BM cell to adult p75KO ischemic tissue recovery. No GFP(+) young p75KO BM cells survived in adult p75KO tissue, signifying the additive negative roles of tissue aging combined with decreased/absent TNFR2/p75 signaling in postischemic recovery.
Collapse
Affiliation(s)
- Sharath P Sasi
- Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts, USA
| | - Layla Rahimi
- Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts, USA
| | - Xinhua Yan
- Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts, USA; Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Marcy Silver
- Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts, USA
| | - Gangjian Qin
- Feinberg Cardiovascular Institute, Feinberg School of Medicine Northwestern University, Chicago, Illinois, USA; and
| | - Douglas W Losordo
- Feinberg Cardiovascular Institute, Feinberg School of Medicine Northwestern University, Chicago, Illinois, USA; and
| | - Raj Kishore
- Center for Translational Medicine, Temple University School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - David A Goukassian
- Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts, USA; Tufts University School of Medicine, Boston, Massachusetts, USA;
| |
Collapse
|
113
|
Regulation of ceramide synthase 6 in a spontaneous experimental autoimmune encephalomyelitis model is sex dependent. Biochem Pharmacol 2014; 92:326-35. [DOI: 10.1016/j.bcp.2014.08.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 08/12/2014] [Accepted: 08/13/2014] [Indexed: 12/16/2022]
|
114
|
Gasperi V, Evangelista D, Oddi S, Florenzano F, Chiurchiù V, Avigliano L, Catani MV, Maccarrone M. Regulation of inflammation and proliferation of human bladder carcinoma cells by type-1 and type-2 cannabinoid receptors. Life Sci 2014; 138:41-51. [PMID: 25445433 DOI: 10.1016/j.lfs.2014.09.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/08/2014] [Accepted: 09/29/2014] [Indexed: 01/14/2023]
Abstract
AIMS Pro-inflammatory cytokines, growth and angiogenic factors released by leukocytes are involved in carcinogenesis and cancer progression, but they are also crucial for fighting tumour growth and spreading. We have previously demonstrated that endocannabinoids modulate cell-to-cell crosstalk during inflammation. Here, we investigated the inflammatory and tumourigenic properties of endocannabinoids in a human urinary bladder carcinoma cell line. MAIN METHODS Endocannabinoid-treated ECV304 cells were checked for tumour necrosis factor (TNF)-α secretion (by ELISA assay) and surface exposure of selectins (by in situ ELISA and FACS analysis). ECV304/Jurkat T cell interaction was assessed by adhesion and live imaging experiments. Proliferation rate, cell death and cell cycle were determined by FACS analysis. KEY FINDINGS By binding to type-1 (CB1) and type-2 (CB2) cannabinoid receptors, the endocannabinoid 2-arachidonoylglycerol (2-AG) exacerbates the pro-inflammatory status surrounding bladder carcinoma ECV304 cells, by: (i) enhancing TNF-α release, (ii) increasing surface exposure of P- and E-selectins, and (iii) allowing Jurkat T lymphocytes to adhere to treated cancer cells. We also found that the CB1 inverse agonist AM281, unlike 2-AG, decreases cancer proliferation by delaying cell cycle progression. SIGNIFICANCE Our data suggest that 2-AG modulates the inflammatory milieu of cancer cells in vitro, while AM281 plays a more specific role in proliferation. Collectively, these findings suggest that CB receptors may play distinct roles in cancer biology, depending on the specific ligand employed. CONCLUSIONS The in vivo assessment of the role of CB receptors in inflammation and cancer might be instrumental in broadening the understanding about bladder cancer biology.
Collapse
Affiliation(s)
- Valeria Gasperi
- Department of Experimental Medicine & Surgery, Tor Vergata University of Rome, Rome, Italy
| | - Daniela Evangelista
- Department of Experimental Medicine & Surgery, Tor Vergata University of Rome, Rome, Italy
| | - Sergio Oddi
- Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy; European Center for Brain Research (CERC)/IRCCS S. Lucia Foundation, Rome, Italy
| | | | - Valerio Chiurchiù
- European Center for Brain Research (CERC)/IRCCS S. Lucia Foundation, Rome, Italy; Center of Integrated Research, Campus Bio-Medico University of Rome, Rome, Italy
| | - Luciana Avigliano
- Department of Experimental Medicine & Surgery, Tor Vergata University of Rome, Rome, Italy
| | - M Valeria Catani
- Department of Experimental Medicine & Surgery, Tor Vergata University of Rome, Rome, Italy.
| | - Mauro Maccarrone
- European Center for Brain Research (CERC)/IRCCS S. Lucia Foundation, Rome, Italy; Center of Integrated Research, Campus Bio-Medico University of Rome, Rome, Italy.
| |
Collapse
|
115
|
Zhong H, Bussel J, Yazdanbakhsh K. In vitro TNF blockade enhances ex vivo expansion of regulatory T cells in patients with immune thrombocytopenia. Br J Haematol 2014; 168:274-83. [PMID: 25252160 DOI: 10.1111/bjh.13126] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 07/29/2014] [Indexed: 01/09/2023]
Abstract
Tumour necrosis factor-α (TNF) is an inflammatory cytokine that is elevated in a number of autoimmune diseases including immune thrombocytopenia (ITP), a bleeding disorder characterized by low platelet counts. In vitro TNF blockade increases expansion of the regulatory T cell (Treg) IKZF2 (also termed Helios) subset in T cell-monocyte cocultures from healthy donors, but its role on proliferative responses of Tregs in ITP patients, who have altered immunoregulatory compartment, remains unclear. TNF in CD4+ T cells from patients with chronic ITP were elevated and negatively correlated with peripheral Treg frequencies, suggesting a possible inhibitory effect of TNF on ITP Tregs. In vitro antibody neutralization with anti-TNF in T cell-monocyte cocultures resulted in a robust expansion of pre-existing ITP Tregs, higher than in healthy controls. Similar to the effects of anti-TNF antibodies, TNF blockade with antibodies against TNFRSF1B (anti-TNFRSF1B, previously termed anti-TNFRII) almost doubled ITP Treg expansion whereas neutralization with anti-TNFRSF1A (anti-TNFRI) antibodies had no effect on proliferative responses of Tregs. In addition, TNFRSF1B levels on ITP Tregs were significantly elevated, which may explain the increased susceptibility of patient Tregs to the actions of TNF blockade. Altogether, these data raise the possibility that TNF blockers, through their ability to increase Treg proliferation, may be efficacious in ITP patients.
Collapse
Affiliation(s)
- Hui Zhong
- Laboratory of Complement Biology, New York Blood Center, New York, NY, USA
| | | | | |
Collapse
|
116
|
Osta B, Lavocat F, Eljaafari A, Miossec P. Effects of Interleukin-17A on Osteogenic Differentiation of Isolated Human Mesenchymal Stem Cells. Front Immunol 2014; 5:425. [PMID: 25228904 PMCID: PMC4151036 DOI: 10.3389/fimmu.2014.00425] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 08/19/2014] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is characterized by defective bone repair and excessive destruction and ankylosing spondylitis (AS) by increased ectopic bone formation with syndesmophytes. Since TNF-α and IL-17A are involved in both diseases, this study investigated their effects on the osteogenic differentiation of isolated human bone marrow-derived mesenchymal stem cells (hMSCs). METHODS Differentiation of hMSCs into osteoblasts was induced in the presence or absence of IL-17A and/or TNF-α. Matrix mineralization (MM) was evaluated by alizarin red staining and alkaline phosphatase (ALP) activity. mRNA expression was measured by qRT-PCR for bone morphogenetic protein (BMP)-2 and Runx2, genes associated with osteogenesis, DKK-1, a negative regulator of osteogenesis, Schnurri-3 and receptor activator of nuclear factor kappa B ligand (RANKL), associated with the cross talk with osteoclasts, and TNF-α receptor type I and TNF-α receptor type II (TNFRII). RESULTS TNF-α alone increased both MM and ALP activity. IL-17A alone increased ALP but not MM. Their combination was more potent. TNF-α alone increased BMP2 mRNA expression at 6 and 12 h. These levels decreased in combination with IL-17A at 6 h only. DKK-1 mRNA expression was inhibited by TNF-α and IL-17A either alone or combined. Supporting an imbalance toward osteoblastogenesis, RANKL expression was inhibited by TNF-α and IL-17A. However, TNF-α but not IL-17 alone decreased Runx2 mRNA expression at 6 h. In parallel, TNF-α but not IL-17 alone increased Schnurri-3 expression with a synergistic effect with their combination. This may be related to an increase of TNFRII overexpression. CONCLUSION IL-17 increased the effects of TNF-α on bone matrix formation by hMSCs. However, IL-17 decreased the TNF-α-induced BMP2 inhibition. Synergistic interactions between TNF-α and IL-17 were seen for RANKL inhibition and Schnurri-3 induction. Such increase of Schnurri-3 may in turn activate osteoclasts leading to bone destruction as in RA. Conversely, in the absence of osteoclasts, this could promote ectopic bone formation as in AS.
Collapse
Affiliation(s)
- Bilal Osta
- Immunogenomics and Inflammation Research Unit EA 4130, Department of Clinical Immunology and Rheumatology, Edouard Herriot Hospital, University of Lyon 1 , Lyon , France
| | - Fabien Lavocat
- Immunogenomics and Inflammation Research Unit EA 4130, Department of Clinical Immunology and Rheumatology, Edouard Herriot Hospital, University of Lyon 1 , Lyon , France
| | - Assia Eljaafari
- Immunogenomics and Inflammation Research Unit EA 4130, Department of Clinical Immunology and Rheumatology, Edouard Herriot Hospital, University of Lyon 1 , Lyon , France
| | - Pierre Miossec
- Immunogenomics and Inflammation Research Unit EA 4130, Department of Clinical Immunology and Rheumatology, Edouard Herriot Hospital, University of Lyon 1 , Lyon , France
| |
Collapse
|
117
|
Joedicke JJ, Myers L, Carmody AB, Messer RJ, Wajant H, Lang KS, Lang PA, Mak TW, Hasenkrug KJ, Dittmer U. Activated CD8+ T cells induce expansion of Vβ5+ regulatory T cells via TNFR2 signaling. THE JOURNAL OF IMMUNOLOGY 2014; 193:2952-60. [PMID: 25098294 DOI: 10.4049/jimmunol.1400649] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Vβ5(+) regulatory T cells (Tregs), which are specific for a mouse endogenous retroviral superantigen, become activated and proliferate in response to Friend virus (FV) infection. We previously reported that FV-induced expansion of this Treg subset was dependent on CD8(+) T cells and TNF-α, but independent of IL-2. We now show that the inflammatory milieu associated with FV infection is not necessary for induction of Vβ5(+) Treg expansion. Rather, it is the presence of activated CD8(+) T cells that is critical for their expansion. The data indicate that the mechanism involves signaling between the membrane-bound form of TNF-α on activated CD8(+) T cells and TNFR2 on Tregs. CD8(+) T cells expressing membrane-bound TNF-α but no soluble TNF-α remained competent to induce strong Vβ5(+) Treg expansion in vivo. In addition, Vβ5(+) Tregs expressing only TNFR2 but no TNFR1 were still responsive to expansion. Finally, treatment of naive mice with soluble TNF-α did not induce Vβ5(+) Treg expansion, but treatment with a TNFR2-specific agonist did. These results reveal a new mechanism of intercellular communication between activated CD8(+) T cell effectors and Tregs that results in the activation and expansion of a Treg subset that subsequently suppresses CD8(+) T cell functions.
Collapse
Affiliation(s)
- Jara J Joedicke
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Lara Myers
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840
| | - Aaron B Carmody
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840
| | - Ronald J Messer
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840
| | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg 97080, Germany
| | - Karl S Lang
- Institute for Immunology, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Philipp A Lang
- Department of Gastroenterology, Hepatology, and Infectious Diseases, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany; Department of Molecular Medicine II, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany; and
| | - Tak W Mak
- Department of Medical Biophysics and Immunology, The Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, University of Toronto, Toronto, Ontario M5G 2M9, Canada
| | - Kim J Hasenkrug
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840;
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany;
| |
Collapse
|
118
|
Ślebioda TJ, Kmieć Z. Tumour necrosis factor superfamily members in the pathogenesis of inflammatory bowel disease. Mediators Inflamm 2014; 2014:325129. [PMID: 25045210 PMCID: PMC4087264 DOI: 10.1155/2014/325129] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 05/29/2014] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a group of inflammatory conditions of the gastrointestinal tract of unclear aetiology of which two major forms are Crohn's disease (CD) and ulcerative colitis (UC). CD and UC are immunologically distinct, although they both result from hyperactivation of proinflammatory pathways in intestines and disruption of intestinal epithelial barrier. Members of the tumour necrosis factor superfamily (TNFSF) are molecules of broad spectrum of activity, including direct disruption of intestinal epithelial barrier integrity and costimulation of proinflammatory functions of lymphocytes. Tumour necrosis factor (TNF) has a well-established pathological role in IBD which also serves as a target in IBD treatment. In this review we discuss the role of TNF and other TNFSF members, notably, TL1A, FasL, LIGHT, TRAIL, and TWEAK, in the pathogenesis of IBD.
Collapse
Affiliation(s)
- Tomasz J. Ślebioda
- Department of Histology, Medical University of Gdańsk, Dębinki 1, 80-211 Gdańsk, Poland
| | - Zbigniew Kmieć
- Department of Histology, Medical University of Gdańsk, Dębinki 1, 80-211 Gdańsk, Poland
| |
Collapse
|
119
|
2-Arachidonoylglycerol modulates human endothelial cell/leukocyte interactions by controlling selectin expression through CB1 and CB2 receptors. Int J Biochem Cell Biol 2014; 51:79-88. [DOI: 10.1016/j.biocel.2014.03.028] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 03/10/2014] [Accepted: 03/28/2014] [Indexed: 12/17/2022]
|