1
|
Dalvi S, Roll M, Chatterjee A, Kumar LK, Bhogavalli A, Foley N, Arduino C, Spencer W, Reuben-Thomas C, Ortolan D, Pébay A, Bharti K, Anand-Apte B, Singh R. Human iPSC-based disease modeling studies identify a common mechanistic defect and potential therapies for AMD and related macular dystrophies. Dev Cell 2024; 59:3290-3305.e9. [PMID: 39362220 PMCID: PMC11652237 DOI: 10.1016/j.devcel.2024.09.006] [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: 09/15/2023] [Revised: 04/17/2024] [Accepted: 09/05/2024] [Indexed: 10/05/2024]
Abstract
Age-related macular degeneration (AMD) and related macular dystrophies (MDs) primarily affect the retinal pigment epithelium (RPE) in the eye. A hallmark of AMD/MDs that drives later-stage pathologies is drusen. Drusen are sub-RPE lipid-protein-rich extracellular deposits, but how drusen forms and accumulates is not known. We utilized human induced pluripotent stem cell (iPSC)-derived RPE from patients with AMD and three distinct MDs to demonstrate that reduced activity of RPE-secreted matrix metalloproteinase 2 (MMP2) contributes to drusen in multiple maculopathies in a genotype-agnostic manner by instigating sterile inflammation and impaired lipid homeostasis via damage-associated molecular pattern molecule (DAMP)-mediated activation of receptor for advanced glycation end-products (RAGE) and increased secretory phospholipase 2-IIA (sPLA2-IIA) levels. Therapeutically, RPE-specific MMP2 supplementation, RAGE-antagonistic peptide, and a small molecule inhibitor of sPLA2-IIA ameliorated drusen accumulation in AMD/MD iPSC-RPE. Ultimately, this study defines a causal role of the MMP2-DAMP-RAGE-sPLA2-IIA axis in AMD/MDs.
Collapse
Affiliation(s)
- Sonal Dalvi
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Michael Roll
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Amit Chatterjee
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Lal Krishan Kumar
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Akshita Bhogavalli
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Nathaniel Foley
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Cesar Arduino
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Whitney Spencer
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Cheyenne Reuben-Thomas
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Davide Ortolan
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Alice Pébay
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Bela Anand-Apte
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Ruchira Singh
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA.
| |
Collapse
|
2
|
Vicentino ARR, Fraga-Junior VDS, Palazzo M, Tasmo NRA, Rodrigues DAS, Barroso SPC, Ferreira SN, Neves-Borges AC, Allonso D, Fantappié MR, Scharfstein J, Oliveira AC, Vianna-Jorge R, Vale AM, Coutinho-Silva R, Savio LEB, Canetti C, Benjamim CF. High mobility group box 1, ATP, lipid mediators, and tissue factor are elevated in COVID-19 patients: HMGB1 as a biomarker of worst prognosis. Clin Transl Sci 2023; 16:631-646. [PMID: 36631939 PMCID: PMC10087071 DOI: 10.1111/cts.13475] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 11/07/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2, the agent of the ongoing coronavirus disease 2019 (COVID-19) pandemic, has spread worldwide since it was first identified in November 2019 in Wuhan, China. Since then, progress in pathogenesis linked severity of this systemic disease to the hyperactivation of network of cytokine-driven pro-inflammatory cascades. Here, we aimed to identify molecular biomarkers of disease severity by measuring the serum levels of inflammatory mediators in a Brazilian cohort of patients with COVID-19 and healthy controls (HCs). Critically ill patients in the intensive care unit were defined as such by dependence on oxygen supplementation (93% intubated and 7% face mask), and computed tomography profiles showing ground-glass opacity pneumonia associated to and high levels of D-dimer. Our panel of mediators included HMGB1, ATP, tissue factor, PGE2 , LTB4 , and cys-LTs. Follow-up studies showed increased serum levels of every inflammatory mediator in patients with COVID-19 as compared to HCs. Originally acting as a transcription factor, HMGB1 acquires pro-inflammatory functions following secretion by activated leukocytes or necrotic tissues. Serum levels of HMGB1 were positively correlated with cys-LTs, D-dimer, aspartate aminotransferase, and alanine aminotransferase. Notably, the levels of the classical alarmin HMGB1 were higher in deceased patients, allowing their discrimination from patients that had been discharged at the early pulmonary and hyperinflammatory phase of COVID-19. In particular, we verified that HMGB1 levels above 125.4 ng/ml is the cutoff that distinguishes patients that are at higher risk of death. In conclusion, we propose the use of serum levels of HMGB1 as a biomarker of severe prognosis of COVID-19.
Collapse
Affiliation(s)
- Amanda Roberta Revoredo Vicentino
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanderlei da Silva Fraga-Junior
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Matheus Palazzo
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natalia Recardo Amorim Tasmo
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielle A S Rodrigues
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Shana Priscila Coutinho Barroso
- Molecular Biology Laboratory, Laboratório de Biologia Molecular, Instituto de Pesquisas Biomédicas, Hospital Naval Marcílio Dias, Rio de Janeiro, Brazil
| | - Sâmila Natiane Ferreira
- Molecular Biology Laboratory, Laboratório de Biologia Molecular, Instituto de Pesquisas Biomédicas, Hospital Naval Marcílio Dias, Rio de Janeiro, Brazil
| | - Anna Cristina Neves-Borges
- Department of Botanic, Departamento de Botânica, Instituto de Biociências, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diego Allonso
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Rosado Fantappié
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julio Scharfstein
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Carolina Oliveira
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rosane Vianna-Jorge
- Programa de Farmacologia e Inflamação, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - André Macedo Vale
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robson Coutinho-Silva
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiz Eduardo Baggio Savio
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudio Canetti
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia Farias Benjamim
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Borde C, Dillard C, L’Honoré A, Quignon F, Hamon M, Marchand CH, Faccion RS, Costa MGS, Pramil E, Larsen AK, Sabbah M, Lemaire SD, Maréchal V, Escargueil AE. The C-Terminal Acidic Tail Modulates the Anticancer Properties of HMGB1. Int J Mol Sci 2022; 23:ijms23147865. [PMID: 35887213 PMCID: PMC9319070 DOI: 10.3390/ijms23147865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/14/2022] [Accepted: 07/14/2022] [Indexed: 02/04/2023] Open
Abstract
Energy metabolism reprogramming was recently listed as a hallmark of cancer. In this process, the switch from pyruvate kinase isoenzyme type M1 to pyruvate kinase isoenzyme type M2 (PKM2) is believed to play a crucial role. Interestingly, the activity of the active form of PKM2 can efficiently be inhibited by the high-mobility group box 1 (HMGB1) protein, leading to a rapid blockage of glucose-dependent aerobic respiration and cancer cell death. HMGB1 is a member of the HMG protein family. It contains two DNA-binding HMG-box domains and an acidic C-terminal tail capable of positively or negatively modulating its biological properties. In this work, we report that the deletion of the C-terminal tail of HMGB1 increases its activity towards a large panel of cancer cells without affecting the viability of normal immortalized fibroblasts. Moreover, in silico analysis suggests that the truncated form of HMGB1 retains the capacity of the full-length protein to interact with PKM2. However, based on the capacity of the cells to circumvent oxidative phosphorylation inhibition, we were able to identify either a cytotoxic or cytostatic effect of the proteins. Together, our study provides new insights in the characterization of the anticancer activity of HMGB1.
Collapse
Affiliation(s)
- Chloé Borde
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM) U938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; (C.B.); (C.D.); (R.S.F.); (E.P.); (A.K.L.); (M.S.)
| | - Clémentine Dillard
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM) U938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; (C.B.); (C.D.); (R.S.F.); (E.P.); (A.K.L.); (M.S.)
| | - Aurore L’Honoré
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS), INSERM, Institut de Biologie Paris-Seine, Biological Adaptation and Aging, B2A-IBPS, F-75005 Paris, France;
| | - Frédérique Quignon
- Sorbonne Université, CNRS UMR 144, Institut Curie Centre de Recherche, F-75248 Paris, France;
| | - Marion Hamon
- Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Physico-Chimique, Plateforme de Protéomique, FR550, F-75005 Paris, France; (M.H.); (C.H.M.)
| | - Christophe H. Marchand
- Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Physico-Chimique, Plateforme de Protéomique, FR550, F-75005 Paris, France; (M.H.); (C.H.M.)
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Paris-Seine, UMR7238, Laboratory of Computational and Quantitative Biology, F-75005 Paris, France;
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Physico-Chimique, UMR8226, F-75005 Paris, France
| | - Roberta Soares Faccion
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM) U938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; (C.B.); (C.D.); (R.S.F.); (E.P.); (A.K.L.); (M.S.)
- Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Hemato-Oncologia Molecular, Hospital do Câncer I, Centro de Pesquisas do Instituto Nacional de Câncer José Alencar Gomes da Silva (INCA), Praça da Cruz Vermelha 23/6° andar, Rio de Janeiro 20230-130, Brazil
| | - Maurício G. S. Costa
- Fundação Oswaldo Cruz, Programa de Computação Científica, Vice-Presidência de Educação, Informação e Comunicação, Av. Brasil 4365, Manguinhos, Rio de Janeiro 21040-900, Brazil;
| | - Elodie Pramil
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM) U938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; (C.B.); (C.D.); (R.S.F.); (E.P.); (A.K.L.); (M.S.)
- Alliance for Research in Cancerology-APREC, Tenon Hospital, F-75020 Paris, France
| | - Annette K. Larsen
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM) U938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; (C.B.); (C.D.); (R.S.F.); (E.P.); (A.K.L.); (M.S.)
| | - Michèle Sabbah
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM) U938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; (C.B.); (C.D.); (R.S.F.); (E.P.); (A.K.L.); (M.S.)
| | - Stéphane D. Lemaire
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Paris-Seine, UMR7238, Laboratory of Computational and Quantitative Biology, F-75005 Paris, France;
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Physico-Chimique, UMR8226, F-75005 Paris, France
| | - Vincent Maréchal
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM) U938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; (C.B.); (C.D.); (R.S.F.); (E.P.); (A.K.L.); (M.S.)
- Correspondence: (V.M.); (A.E.E.); Tel.: +33-(0)-1-44-27-31-53 (V.M.); +33-(0)-1-49-28-46-44 (A.E.E.)
| | - Alexandre E. Escargueil
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM) U938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; (C.B.); (C.D.); (R.S.F.); (E.P.); (A.K.L.); (M.S.)
- Correspondence: (V.M.); (A.E.E.); Tel.: +33-(0)-1-44-27-31-53 (V.M.); +33-(0)-1-49-28-46-44 (A.E.E.)
| |
Collapse
|
4
|
Jeong J, Cho S, Lee BS, Seo M, Jang Y, Lim S, Park S. Soluble RAGE attenuates Ang II-induced arterial calcification via inhibiting AT1R-HMGB1-RAGE axis. Atherosclerosis 2022; 346:53-62. [DOI: 10.1016/j.atherosclerosis.2022.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 11/30/2022]
|
5
|
Sun C, Zheng W, Liang L, Liu Z, Sun W, Tang R. Ezetimibe Improves Rosuvastatin Effects on Inflammation and Vascular Endothelial Function in Acute Coronary Syndrome Patients Undergoing PCI. J Interv Cardiol 2021; 2021:2995602. [PMID: 34566523 PMCID: PMC8443370 DOI: 10.1155/2021/2995602] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/26/2021] [Accepted: 08/18/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Little is known of the acute effects of ezetimibe in patients with acute coronary syndrome (ACS) undergoing PCI. We investigated whether ezetimibe improves inflammation and vascular endothelial function in patients with ACS undergoing PCI. METHODS We randomized 171 patients with ACS undergoing PCI to receive ezetimibe 10 mg/day plus rosuvastatin 20 mg/day (combination group, n = 81) versus rosuvastatin 20 mg/day (rosuvastatin group, n = 90). Lipid profile, type II secretory phospholipase A2 (sPLA2-IIa), interleukin-1β (IL-1β), vascular cell adhesion molecule-1 (VCAM-1), and intercellular cell adhesion molecule-1 (ICAM-1) were measured at baseline and after 7 days. Three months after PCI, clinical outcomes were examined. RESULT The levels of sPLA2-IIa and IL-1β reduced significantly in both groups, but more when ezetimibe and rosuvastatin were coadministered (sPLA2-IIa: 6.16 ± 2.67 vs. 7.42 ± 3.53 ng/ml, p=0.01; IL-1β: 37.39 ± 26.25 vs. 48.98 ± 32.26 pg/ml, p=0.01). A significant rise of VCAM-1 and ICAM-1 was observed on day 7 after PCI in the both groups, but was less in the combination group (VCAM-1: 918.28 ± 235.31 vs. 988.54 ± 194.41 ng/ml, p=0.03; ICAM-1: 213.01 ± 100.15 vs. 246.88 ± 105.71 ng/ml, p=0.03). Patients in the combination versus rosuvastatin group appeared to suffer from less major adverse events. Periprocedural therapy of ezetimibe improves rosuvastatin effects on proinflammatory responses and endothelial function associated with ACS patients undergoing PCI. This trial is registered with https://clinicaltrials.gov/ct2/show/ChiCTR-IPR-17012219 (Chinese Clinical Trial Registry, http://www.chictr.org.cn on 02/08/2017).
Collapse
Affiliation(s)
- Changqing Sun
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Wuyang Zheng
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Ling Liang
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Zuheng Liu
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Wenchao Sun
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Rong Tang
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| |
Collapse
|
6
|
Behl T, Sharma E, Sehgal A, Kaur I, Kumar A, Arora R, Pal G, Kakkar M, Kumar R, Bungau S. Expatiating the molecular approaches of HMGB1 in diabetes mellitus: Highlighting signalling pathways via RAGE and TLRs. Mol Biol Rep 2021; 48:1869-1881. [PMID: 33479829 DOI: 10.1007/s11033-020-06130-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/24/2020] [Indexed: 12/16/2022]
Abstract
Diabetes mellitus (DM) has become one of the major healthcare challenges worldwide in the recent times and inflammation being one of its key pathogenic process/mechanism affect several body parts including the peripheral and central nervous system. High-mobility group box 1 (HMGB1) is one of the major non-histone proteins that plays a key role in triggering the inflammatory response. Upon its release into the extracellular milieu, HMGB1 acts as an "alarmin" for the immune system to initiate tissue repair as a component of the host defense system. Furthermore, HMGB1 along with its downstream receptors like Toll-like receptors (TLRs) and receptors for advanced glycation end products (RAGE) serve as the suitable target for DM. The forthcoming research in the field of diabetes would potentially focus on the development of alternative approaches to target the centre of inflammation that is primarily mediated by HMGB1 to improve diabetic-related complications. This review covers the therapeutic actions of HMGB1 protein, which acts by activating the RAGE and TLR molecules to constitute a functional tripod system, in turn activating NF-κB pathway that contributes to the production of mediators for pro-inflammatory cytokines associated with DM. The interaction between TLR2 and TLR4 with ligands present in the host and the activation of RAGE stimulates various immune and metabolic responses that contribute to diabetes. This review emphasizes to elucidate the role of HMGB1 in the initiation and progression of DM and control over the inflammatory tripod as a promising therapeutic approach in the management of DM.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Eshita Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Rashmi Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Giridhari Pal
- Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Munish Kakkar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ravinder Kumar
- Cardiovascular Research Institute, Icahn School of Medicine, New York, USA
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
7
|
Song HH, Song TC, Yang T, Sun CS, He BQ, Li H, Wang YJ, Li Y, Wu H, Hu YM, Wang YJ. High mobility group box 1 mediates inflammatory response of astrocytes via cyclooxygenase 2/prostaglandin E2 signaling following spinal cord injury. Neural Regen Res 2021; 16:1848-1855. [PMID: 33510092 PMCID: PMC8328776 DOI: 10.4103/1673-5374.303039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
High mobility group box 1 (HMGB1) interacts with pattern-recognition receptors of immune cells to activate the inflammatory response. Astrocytes play a positive role in the inflammatory response of the central nervous system by expressing a broad range of pattern-recognition receptors. However, the underlying relationship between HMGB1 and the inflammatory reaction of astrocytes remains unclear. In this study, we established rat models of spinal cord injury via laminectomy at the T8–10 level, and the injured spinal cord was subjected to transcriptome sequencing. Our results showed that the HMGB1/Toll-like receptor 4 (TLR4) axis was involved in the activation of astrocyte inflammatory response through regulation of cyclooxygenase 2 (COX2)/prostaglandin E2 (PGE2) signaling. Both TLR4 and COX2 were distributed in astrocytes and showed elevated protein levels following spinal cord injury. Stimulation of primary astrocytes with recombinant HMGB1 showed that COX2 and microsomal PGE synthase (mPGES)-1, rather than COX1, mPGES-2, or cytosolic PGE synthase, were significantly upregulated. Accordingly, PGE2 production in astrocytes was remarkably increased in response to recombinant HMGB1 challenges. Pharmacologic blockade of TLR2/4 attenuated HMGB1-mediated activation of the COX2/PGE2 pathway. Interestingly, HMGB1 did not impact the production of tumor necrosis factor-α or interleukin-1β in astrocytes. Our results suggest that HMGB1 mediates the astrocyte inflammatory response through regulating the COX2/PGE2 signaling pathway. The study was approved by the Laboratory Animal Ethics Committee of Nantong University, China (approval No. 20181204-001) on December 4, 2018.
Collapse
Affiliation(s)
- Hong-Hua Song
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University; Center of Special Inspection, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Tian-Cheng Song
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Ting Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Chun-Shuai Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Bing-Qiang He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Hui Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Ying-Jie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Yu Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hao Wu
- Department of Otolaryngology Head Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Yu-Ming Hu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Yong-Jun Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
8
|
Piotrowski J, Jędrzejewski T, Pawlikowska M, Wrotek S, Kozak W. High mobility group box 1 protein released in the course of aseptic necrosis of tissues sensitizes rats to pyrogenic effects of lipopolysaccharide. J Therm Biol 2019; 84:36-44. [PMID: 31466775 DOI: 10.1016/j.jtherbio.2019.05.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/24/2019] [Accepted: 05/31/2019] [Indexed: 12/16/2022]
Abstract
It is still an open question as to whether or not aseptic injuries affect the generation of fever due to exogenous pyrogens including bacterial products. Therefore, in the present paper we have investigated the course of endotoxin fever in rats induced with lipopolysaccharide (LPS; given intraperitoneally in a dose of 50 μg/kg) 48 h after subcutaneous administration of turpentine oil (TRP; 0.1 mL per rat) that causes aseptic necrosis of tissues. We found that febrile response was significantly augmented in the animals pre-treated with turpentine compared to control rats (pre-treated with saline), and that observed excessive elevation of body temperature (Tb) was accompanied by enhanced release of fever mediators: interleukin-6 (IL-6) and prostaglandin E2 (PGE2) into plasma. Moreover, we found that sensitization to pyrogenic effects of lipopolysaccharide was associated with the increase in plasma level of high mobility group box 1 protein (HMGB1), one of the best-known damage-associated molecular patterns (DAMP), which was recently discovered as inflammatory mediator. Since the injection of anti-HMGB1 antibodies weakened observed hyperpyrexia in the animals pre-treated with turpentine, we conclude that HMGB1 is a plasma-derived factor released in the course of aseptic injury that enhances pyrogenic effects of LPS.
Collapse
Affiliation(s)
- Jakub Piotrowski
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 1 Lwowska Street, 87-100, Torun, Poland.
| | - Tomasz Jędrzejewski
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 1 Lwowska Street, 87-100, Torun, Poland
| | - Małgorzata Pawlikowska
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 1 Lwowska Street, 87-100, Torun, Poland
| | - Sylwia Wrotek
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 1 Lwowska Street, 87-100, Torun, Poland
| | - Wieslaw Kozak
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 1 Lwowska Street, 87-100, Torun, Poland
| |
Collapse
|
9
|
Hwang JH, Chu H, Ahn Y, Kim J, Kim DY. HMGB1 promotes hair growth via the modulation of prostaglandin metabolism. Sci Rep 2019; 9:6660. [PMID: 31040377 PMCID: PMC6491442 DOI: 10.1038/s41598-019-43242-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 04/18/2019] [Indexed: 01/09/2023] Open
Abstract
Unexpected hair growth can occur after tissue injury. The pathogenic mechanism for this phenomenon is unknown but is likely related to inflammatory mediators. One such mediator is high-mobility group box 1 (HMGB1), a ubiquitous nuclear protein that is released from cell nuclei after tissue damage. To elucidate the effect of HMGB1 on hair growth and understand its mechanism of action, we evaluated the effect of HMGB1 treatment on hair shaft elongation and on mRNA and protein expression in cultured human dermal papilla cells (hDPCs). HMGB1 enhanced hair shaft elongation in an ex vivo hair organ culture. In hDPCs, HMGB1 treatment significantly increased mRNA and protein expression levels of prostagladin E synthases. HMGB1 also stimulated prostaglandin E2 (PGE2) secretion from hDPCs. Finally, blocking the receptor for advanced glycation end-products, a canonical HMGB1 receptor, inhibited HMGB1-induced PGE2 production and hair shaft elongation. Our results suggest that HMGB1 promotes hair growth via PGE2 secretion from hDPCs. This mechanism can explain the paradoxical phenomenon of trauma-induced hair growth. Thus, HGMB1 can be a viable therapeutic target for the treatment of alopecia.
Collapse
Affiliation(s)
- Ji-Hye Hwang
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Howard Chu
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Yuri Ahn
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jino Kim
- New Hair Institute, Seoul, Korea
| | - Do-Young Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
10
|
Attiq A, Jalil J, Husain K, Ahmad W. Raging the War Against Inflammation With Natural Products. Front Pharmacol 2018; 9:976. [PMID: 30245627 PMCID: PMC6137277 DOI: 10.3389/fphar.2018.00976] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/08/2018] [Indexed: 12/31/2022] Open
Abstract
Over the last few decade Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) are the drugs of choice for treating numerous inflammatory diseases including rheumatoid arthritis. The NSAIDs produces anti-inflammatory activity via inhibiting cyclooxygenase enzyme, responsible for the conversation of arachidonic acid to prostaglandins. Likewise, cyclooxegenase-2 inhibitors (COX-2) selectively inhibit the COX-2 enzyme and produces significant anti-inflammatory, analgesic, and anti-pyretic activity without producing COX-1 associated gastrointestinal and renal side effects. In last two decades numerous selective COX-2 inhibitors (COXIBs) have been developed and approved for various inflammatory conditions. However, data from clinical trials have suggested that the prolong use of COX-2 inhibitors are also associated with life threatening cardiovascular side effects including ischemic heart failure and myocardial infection. In these scenario secondary metabolites from natural product offers a great hope for the development of novel anti-inflammatory compounds. Although majority of the natural product based compounds exhibit more selectively toward COX-1. However, the data suggest that slight structural modification can be helpful in developing COX-2 selective secondary metabolites with comparative efficacy and limited side effects. This review is an effort to highlight the secondary metabolites from terrestrial and marine source with significant COX-2 and COX-2 mediated PGE2 inhibitory activity, since it is anticipated that isolates with ability to inhibit COX-2 mediated PGE2 production would be useful in suppressing the inflammation and its classical sign and symptoms. Moreover, this review has highlighted the potential lead compounds including berberine, kaurenoic acid, α-cyperone, curcumin, and zedoarondiol for further development with the help of structure-activity relationship (SAR) studies and their current status.
Collapse
Affiliation(s)
- Ali Attiq
- Drug and Herbal Research Centre, Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Juriyati Jalil
- Drug and Herbal Research Centre, Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Khairana Husain
- Drug and Herbal Research Centre, Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Waqas Ahmad
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Malaysia
| |
Collapse
|
11
|
Lipopolysaccharide (LPS)-mediated priming of toll-like receptor 4 enhances oxidant-induced prostaglandin E 2 biosynthesis in primary murine macrophages. Int Immunopharmacol 2017; 54:226-237. [PMID: 29161659 DOI: 10.1016/j.intimp.2017.11.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/13/2017] [Accepted: 11/14/2017] [Indexed: 12/17/2022]
Abstract
Agonists and pseudo-agonists for toll-like receptor 4 (TLR4) are common in our environment. Thus, human exposure to these agents may result in "priming or sensitization" of TLR4. A body of evidence suggests that LPS-mediated sensitization of TLR4 can increase the magnitude of responses to exogenous agents in multiple tissues. We have previously shown that reactive oxygen and nitrogen species (RONS) stimulate TLR4. There is no evidence that LPS-primed TLR4 can influence the magnitude of responses to oxidants from either endogenous or exogenous sources. In the present study, we directly tested the hypothesis that LPS-primed TLR4 will sensitize primary murine peritoneal macrophages (pM) to oxidant-mediated prostaglandin E2 (PGE2) production. We used potassium peroxychromate (PPC) and potassium peroxynitrite (PPN) as direct in vitro sources of exogenous RONS. Our results showed that a direct treatment with PPC or PPN alone as sources of exogenous oxidants had a limited effect on PGE2 biosynthesis. In contrast, pM sensitized by prior incubation with LPS-EK, a TLR4-specific agonist, followed by oxidant stimulation exhibited increased transcriptional and translational expression of cyclooxygenase-2 (COX-2) with enhanced PGE2 biosynthesis/production only in pM derived from TLR4-WT mice but not in TLR4-KO mice. Thus, we have shown a critical role for LPS-primed TLR4 in oxidant-induced inflammatory phenotypes that have the potential to initiate, propagate and maintain many human diseases.
Collapse
|
12
|
Cimmino G, Ciuffreda LP, Ciccarelli G, Calabrò P, Ferraiolo FAV, Rivellino A, De Palma R, Golino P, Rossi F, Cirillo P, Berrino L. Upregulation of TH/IL-17 Pathway-Related Genes in Human Coronary Endothelial Cells Stimulated with Serum of Patients with Acute Coronary Syndromes. Front Cardiovasc Med 2017; 4:1. [PMID: 28224128 PMCID: PMC5293806 DOI: 10.3389/fcvm.2017.00001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/11/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Inflammation plays an essential role in the development and complications of atherosclerosis plaques, including acute coronary syndromes (ACS). Indeed, previous reports have shown that within the coronary circulation of ACS patients, several soluble mediators are released. Moreover, it has been demonstrated that endothelial dysfunction might play an important role in atherosclerosis as well as ACS pathophysiology. However, the mechanisms by which these soluble mediators might affect endothelial functions are still largely unknown. We have evaluated whether soluble mediators contained in serum from coronary circulation of ACS patients might promote changes of gene profile in human coronary endothelial cells (HCAECs). METHODS HCAECs were stimulated in vitro for 12 h with serum obtained from the coronary sinus (CS) and the aorta (Ao) of ACS patients; stable angina (SA) patients served as controls. Gene expression profiles of stimulated cells were evaluated by microarray and real-time PCR. RESULTS HCAECs stimulated with serum from CS of ACS patients showed a significant change (upregulation and downregulation) in gene expression profile as compared with cells stimulated with serum from CS of SA patients. Moreover, ad hoc sub analysis indicated the upregulation of Th-17/IL-17 pathway-related genes. CONCLUSION This study demonstrates that, in ACS patients, the chemical mediators released in the coronary circulation might be able to perturb coronary endothelial cells (ECs) modifying their gene profile. These modified ECs, through downregulation of protective gene and, mainly, through upregulation of gene able to modulate the Th-17/IL-17 pathway, might play a key role in progression of coronary atherosclerosis and in developing future acute events.
Collapse
Affiliation(s)
- Giovanni Cimmino
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Loreta Pia Ciuffreda
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Giovanni Ciccarelli
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Paolo Calabrò
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | | | - Alessia Rivellino
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Raffaele De Palma
- Department of Clinical and Experimental Medicine, Section of Immunology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Paolo Golino
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Plinio Cirillo
- Department of Advanced Biomedical Sciences, Section of Cardiology, University of Naples, “Federico II”, Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
13
|
Xie Q, Zhang D. Effects of Statins and Xuezhikang on the Expression of Secretory Phospholipase A2, Group IIA in Rat Vascular Smooth Muscle Cells. Int Heart J 2017; 58:115-124. [PMID: 28123160 DOI: 10.1536/ihj.16-163] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Atherosclerosis is a multifactorial vascular disease characterized by formation of inflammatory lesions. Secretory phospholipase A2, group IIA (sPLA2-IIA) is involved in this process and plays a critical role. However, the exact role of sPLA2-IIA in cardiovascular inflammation is more complicated and remains unclear. Furthermore, both statins and Xuezhikang (XZK) are widely used in the prevention and treatment of cardiovascular disease risk because of their pleiotropic effects on the cardiovascular system. However, their effects on sPLA2-IIA are still controversial. We investigated the regulation of sPLA2-IIA by rat thoracic aorta smooth muscle cells (VSMCs) in culture. Cells were first incubated with IL-1β alone to induce expression of sPLA2-IIA and then treated with several concentrations of statins or XZK for different times in the absence or presence of IL-1β. We tested the expression of sPLA2-IIA, including sPLA2-IIA mRNA, protein, as well as activity. We found that statins or IL-1β increase the expression of sPLA2-IIA in VSMCs and the effect is based on a synergetic relationship between them. However, for the first time, we observed that XZK effectively reduces sPLA2-IIA expression in IL-1β-treated VSMCs. Our findings may shine a new light on the clinical use of XZK and statins in the prevention and treatment of atherosclerosis-related thrombosis.
Collapse
Affiliation(s)
- Qiang Xie
- Department of Cardiology, The First Hospital of Xiamen University
| | | |
Collapse
|
14
|
Zhu S, Wang Y, Chen W, Li W, Wang A, Wong S, Bao G, Li J, Yang H, Tracey KJ, D’Angelo J, Wang H. High-Density Lipoprotein (HDL) Counter-Regulates Serum Amyloid A (SAA)-Induced sPLA2-IIE and sPLA2-V Expression in Macrophages. PLoS One 2016; 11:e0167468. [PMID: 27898742 PMCID: PMC5127586 DOI: 10.1371/journal.pone.0167468] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/15/2016] [Indexed: 12/20/2022] Open
Abstract
Human serum amyloid A (SAA) has been demonstrated as a chemoattractant and proinflammatory mediator of lethal systemic inflammatory diseases. In the circulation, it can be sequestered by a high-density lipoprotein, HDL, which carries cholesterol, triglycerides, phospholipids and apolipoproteins (Apo-AI). The capture of SAA by HDL results in the displacement of Apo-AI, and the consequent inhibition of SAA’s chemoattractant activities. It was previously unknown whether HDL similarly inhibits SAA-induced sPLA2 expression, as well as the resultant HMGB1 release, nitric oxide (NO) production and autophagy activation. Here we provided compelling evidence that human SAA effectively upregulated the expression and secretion of both sPLA2-IIE and sPLA2-V in murine macrophages, which were attenuated by HDL in a dose-dependent fashion. Similarly, HDL dose-dependently suppressed SAA-induced HMGB1 release, NO production, and autophagy activation. In both RAW 264.7 cells and primary macrophages, HDL inhibited SAA-induced secretion of several cytokines (e.g., IL-6) and chemokines (e.g., MCP-1 and RANTES) that were likely dependent on functional TLR4 signaling. Collectively, these findings suggest that HDL counter-regulates SAA-induced upregulation and secretion of sPLA2-IIE/V in addition to other TLR4-dependent cytokines and chemokines in macrophage cultures.
Collapse
Affiliation(s)
- Shu Zhu
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York, United States of America
- The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Yongjun Wang
- The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Weiqiang Chen
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York, United States of America
- The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Wei Li
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York, United States of America
- The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Angelina Wang
- The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Sarabeth Wong
- The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Guoqiang Bao
- The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
- Department of General Surgery, Tangdu Hospital, The 4 Military Medical University, Xi'an, Shaanxi, China
| | - Jianhua Li
- The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Huan Yang
- The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Kevin J. Tracey
- The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - John D’Angelo
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York, United States of America
| | - Haichao Wang
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York, United States of America
- The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
- * E-mail:
| |
Collapse
|
15
|
Reiss CS. Innate Immunity in Viral Encephalitis. NEUROTROPIC VIRAL INFECTIONS 2016. [PMCID: PMC7153449 DOI: 10.1007/978-3-319-33189-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Carol Shoshkes Reiss
- Departments of Biology and Neural Science, New York University, New York, New York USA
| |
Collapse
|
16
|
El Hadri K, Denoyelle C, Ravaux L, Viollet B, Foretz M, Friguet B, Rouis M, Raymondjean M. AMPK Signaling Involvement for the Repression of the IL-1β-Induced Group IIA Secretory Phospholipase A2 Expression in VSMCs. PLoS One 2015; 10:e0132498. [PMID: 26162096 PMCID: PMC4498592 DOI: 10.1371/journal.pone.0132498] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 06/15/2015] [Indexed: 12/25/2022] Open
Abstract
Secretory Phospholipase A2 of type IIA (sPLA2 IIA) plays a crucial role in the production of lipid mediators by amplifying the neointimal inflammatory context of the vascular smooth muscle cells (VSMCs), especially during atherogenesis. Phenformin, a biguanide family member, by its anti-inflammatory properties presents potential for promoting beneficial effects upon vascular cells, however its impact upon the IL-1β-induced sPLA2 gene expression has not been deeply investigated so far. The present study was designed to determine the relationship between phenformin coupling AMP-activated protein kinase (AMPK) function and the molecular mechanism by which the sPLA2 IIA expression was modulated in VSMCs. Here we find that 5-aminoimidazole-4-carboxamide-1-β-D-ribonucleotide (AICAR) treatment strongly repressed IL-1β-induced sPLA2 expression at least at the transcriptional level. Our study reveals that phenformin elicited a dose-dependent inhibition of the sPLA2 IIA expression and transient overexpression experiments of constitutively active AMPK demonstrate clearly that AMPK signaling is involved in the transcriptional inhibition of sPLA2-IIA gene expression. Furthermore, although the expression of the transcriptional repressor B-cell lymphoma-6 protein (BCL-6) was markedly enhanced by phenformin and AICAR, the repression of sPLA2 gene occurs through a mechanism independent of BCL-6 DNA binding site. In addition we show that activation of AMPK limits IL-1β-induced NF-κB pathway activation. Our results indicate that BCL-6, once activated by AMPK, functions as a competitor of the IL-1β induced NF-κB transcription complex. Our findings provide insights on a new anti-inflammatory pathway linking phenformin, AMPK and molecular control of sPLA2 IIA gene expression in VSMCs.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/metabolism
- Aminoimidazole Carboxamide/analogs & derivatives
- Aminoimidazole Carboxamide/pharmacology
- Animals
- Binding Sites
- Cattle
- Cell Separation
- Cells, Cultured
- Enzyme Activation/drug effects
- Gene Expression Regulation, Enzymologic/drug effects
- Group II Phospholipases A2/genetics
- Group II Phospholipases A2/metabolism
- Interleukin-1beta/pharmacology
- Male
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- NF-kappa B/metabolism
- Phenformin/pharmacology
- Phosphorylation/drug effects
- Promoter Regions, Genetic/genetics
- Protein Subunits/metabolism
- Proto-Oncogene Proteins c-bcl-6/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Wistar
- Ribonucleotides/pharmacology
- Signal Transduction/drug effects
- Transcriptional Activation/drug effects
Collapse
Affiliation(s)
- Khadija El Hadri
- Sorbonne Universités, Université Pierre et Marie Curie, Biological Adaptation and Ageing (B2A) CNRS UMR8256/INSERM ERL-U1064, F-75005 Paris, France
- * E-mail:
| | - Chantal Denoyelle
- Université Paris Diderot, Sorbonne Paris Cité, BFA CNRS UMR8251, 4 Rue MA Lagroua Weill Hallé, 75013 Paris, France
| | - Lucas Ravaux
- Sorbonne Universités, Université Pierre et Marie Curie, Biological Adaptation and Ageing (B2A) CNRS UMR8256/INSERM ERL-U1064, F-75005 Paris, France
| | - Benoit Viollet
- Institut Cochin, Inserm U1016, Paris, France
- CNRS, UMR 8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marc Foretz
- Institut Cochin, Inserm U1016, Paris, France
- CNRS, UMR 8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bertrand Friguet
- Sorbonne Universités, Université Pierre et Marie Curie, Biological Adaptation and Ageing (B2A) CNRS UMR8256/INSERM ERL-U1064, F-75005 Paris, France
| | - Mustapha Rouis
- Sorbonne Universités, Université Pierre et Marie Curie, Biological Adaptation and Ageing (B2A) CNRS UMR8256/INSERM ERL-U1064, F-75005 Paris, France
| | - Michel Raymondjean
- Sorbonne Universités, Université Pierre et Marie Curie, Biological Adaptation and Ageing (B2A) CNRS UMR8256/INSERM ERL-U1064, F-75005 Paris, France
| |
Collapse
|
17
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Vascular smooth muscle cell in atherosclerosis. Acta Physiol (Oxf) 2015; 214:33-50. [PMID: 25677529 DOI: 10.1111/apha.12466] [Citation(s) in RCA: 295] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/02/2015] [Accepted: 02/09/2015] [Indexed: 12/30/2022]
Abstract
Vascular smooth muscle cells (VSMCs) exhibit phenotypic and functional plasticity in order to respond to vascular injury. In case of the vessel damage, VSMCs are able to switch from the quiescent 'contractile' phenotype to the 'proinflammatory' phenotype. This change is accompanied by decrease in expression of smooth muscle (SM)-specific markers responsible for SM contraction and production of proinflammatory mediators that modulate induction of proliferation and chemotaxis. Indeed, activated VSMCs could efficiently proliferate and migrate contributing to the vascular wall repair. However, in chronic inflammation that occurs in atherosclerosis, arterial VSMCs become aberrantly regulated and this leads to increased VSMC dedifferentiation and extracellular matrix formation in plaque areas. Proatherosclerotic switch in VSMC phenotype is a complex and multistep mechanism that may be induced by a variety of proinflammatory stimuli and hemodynamic alterations. Disturbances in hemodynamic forces could initiate the proinflammatory switch in VSMC phenotype even in pre-clinical stages of atherosclerosis. Proinflammatory signals play a crucial role in further dedifferentiation of VSMCs in affected vessels and propagation of pathological vascular remodelling.
Collapse
Affiliation(s)
- D. A. Chistiakov
- Research Center for Children's Health; Moscow Russia
- The Mount Sinai Community Clinical Oncology Program; Mount Sinai Comprehensive Cancer Center; Mount Sinai Medical Center; Miami Beach FL USA
| | - A. N. Orekhov
- Institute for Atherosclerosis; Skolkovo Innovative Center; Moscow Russia
- Laboratory of Angiopathology; Institute of General Pathology and Pathophysiology; Russian Academy of Sciences; Moscow Russia
- Department of Biophysics; Biological Faculty; Moscow State University; Moscow Russia
| | - Y. V. Bobryshev
- Institute for Atherosclerosis; Skolkovo Innovative Center; Moscow Russia
- Faculty of Medicine; School of Medical Sciences; University of New South Wales; Kensington Sydney NSW Australia
- School of Medicine; University of Western Sydney; Campbelltown NSW Australia
| |
Collapse
|
18
|
Mokgokong R, Wang S, Taylor CJ, Barrand MA, Hladky SB. Ion transporters in brain endothelial cells that contribute to formation of brain interstitial fluid. Pflugers Arch 2014; 466:887-901. [PMID: 24022703 PMCID: PMC4006130 DOI: 10.1007/s00424-013-1342-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 08/24/2013] [Accepted: 08/24/2013] [Indexed: 02/07/2023]
Abstract
Ions and water transported across the endothelium lining the blood–brain barrier contribute to the fluid secreted into the brain and are important in maintaining appropriate volume and ionic composition of brain interstitial fluid. Changes in this secretion process may occur after stroke. The present study identifies at transcript and protein level ion transporters involved in the movement of key ions and examines how levels of certain of these alter following oxidative stress. Immunohistochemistry provides evidence for Cl−/HCO3− exchanger, AE2, and Na+, HCO3− cotransporters, NBCe1 and NBCn1, on brain microvessels. mRNA analysis by RT-PCR reveals expression of these transporters in cultured rat brain microvascular endothelial cells (both primary and immortalized GPNT cells) and also Na+/H+ exchangers, NHE1 (primary and immortalized) and NHE2 (primary cells only). Knock-down using siRNA in immortalized GPNT cells identifies AE2 as responsible for much of the Cl−/HCO3− exchange following extracellular chloride removal and NHE1 as the transporter that accounts for most of the Na+/H+ exchange following intracellular acidification. Transcript levels of both AE2 and NHE1 are increased following hypoxia/reoxygenation. Further work is now required to determine the localization of the bicarbonate transporters to luminal or abluminal membranes of the endothelial cells as well as to identify and localize additional transport mechanisms that must exist for K+ and Cl−.
Collapse
Affiliation(s)
- Ruth Mokgokong
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| | - Shanshan Wang
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| | - Caroline J. Taylor
- O’Brien Institute and Department of Surgery, University of Melbourne, St. Vincent’s Hospital, 42 Fitzroy Street, Fitzroy, Melbourne, VIC 3065 Australia
- Faculty of Health Sciences, Australian Catholic University, Melbourne, VIC 3065 Australia
| | - Margery A. Barrand
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| | - Stephen B. Hladky
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| |
Collapse
|
19
|
Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, Huang J, Yu Y, Fan XG, Yan Z, Sun X, Wang H, Wang Q, Tsung A, Billiar TR, Zeh HJ, Lotze MT, Tang D. HMGB1 in health and disease. Mol Aspects Med 2014; 40:1-116. [PMID: 25010388 PMCID: PMC4254084 DOI: 10.1016/j.mam.2014.05.001] [Citation(s) in RCA: 740] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/05/2014] [Indexed: 12/22/2022]
Abstract
Complex genetic and physiological variations as well as environmental factors that drive emergence of chromosomal instability, development of unscheduled cell death, skewed differentiation, and altered metabolism are central to the pathogenesis of human diseases and disorders. Understanding the molecular bases for these processes is important for the development of new diagnostic biomarkers, and for identifying new therapeutic targets. In 1973, a group of non-histone nuclear proteins with high electrophoretic mobility was discovered and termed high-mobility group (HMG) proteins. The HMG proteins include three superfamilies termed HMGB, HMGN, and HMGA. High-mobility group box 1 (HMGB1), the most abundant and well-studied HMG protein, senses and coordinates the cellular stress response and plays a critical role not only inside of the cell as a DNA chaperone, chromosome guardian, autophagy sustainer, and protector from apoptotic cell death, but also outside the cell as the prototypic damage associated molecular pattern molecule (DAMP). This DAMP, in conjunction with other factors, thus has cytokine, chemokine, and growth factor activity, orchestrating the inflammatory and immune response. All of these characteristics make HMGB1 a critical molecular target in multiple human diseases including infectious diseases, ischemia, immune disorders, neurodegenerative diseases, metabolic disorders, and cancer. Indeed, a number of emergent strategies have been used to inhibit HMGB1 expression, release, and activity in vitro and in vivo. These include antibodies, peptide inhibitors, RNAi, anti-coagulants, endogenous hormones, various chemical compounds, HMGB1-receptor and signaling pathway inhibition, artificial DNAs, physical strategies including vagus nerve stimulation and other surgical approaches. Future work further investigating the details of HMGB1 localization, structure, post-translational modification, and identification of additional partners will undoubtedly uncover additional secrets regarding HMGB1's multiple functions.
Collapse
Affiliation(s)
- Rui Kang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| | - Ruochan Chen
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Qiuhong Zhang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Wen Hou
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Sha Wu
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Lizhi Cao
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jin Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yan Yu
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xue-Gong Fan
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhengwen Yan
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA; Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Xiaofang Sun
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Experimental Department of Institute of Gynecology and Obstetrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510510, China
| | - Haichao Wang
- Laboratory of Emergency Medicine, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - Qingde Wang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Allan Tsung
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Herbert J Zeh
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Daolin Tang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| |
Collapse
|
20
|
Lim S, Park S. Role of vascular smooth muscle cell in the inflammation of atherosclerosis. BMB Rep 2014; 47:1-7. [PMID: 24388105 PMCID: PMC4163848 DOI: 10.5483/bmbrep.2014.47.1.285] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Indexed: 01/13/2023] Open
Abstract
Atherosclerosis is a pathologic process occurring within the artery, in which many cell types, including T cell, macrophages, endothelial cells, and smooth muscle cells, interact, and cause chronic inflammation, in response to various inner- or outer-cellular stimuli. Atherosclerosis is characterized by a complex interaction of inflammation, lipid deposition, vascular smooth muscle cell proliferation, endothelial dysfunction, and extracellular matrix remodeling, which will result in the formation of an intimal plaque. Although the regulation and function of vascular smooth muscle cells are important in the progression of atherosclerosis, the roles of smooth muscle cells in regulating vascular inflammation are rarely focused upon, compared to those of endothelial cells or inflammatory cells. Therefore, in this review, we will discuss here how smooth muscle cells contribute or regulate the inflammatory reaction in the progression of atherosclerosis, especially in the context of the activation of various membrane receptors, and how they may regulate vascular inflammation.
Collapse
Affiliation(s)
| | - Sungha Park
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases, Yonsei University College of Medicine; Cardiovascular Research Institute, Yonsei University College of Medicine; Division of Cardiology, Yonsei University College of Medicine, Seoul 120-752, Korea
| |
Collapse
|
21
|
Sadamura-Takenaka Y, Ito T, Noma S, Oyama Y, Yamada S, Kawahara KI, Inoue H, Maruyama I. HMGB1 promotes the development of pulmonary arterial hypertension in rats. PLoS One 2014; 9:e102482. [PMID: 25032709 PMCID: PMC4102514 DOI: 10.1371/journal.pone.0102482] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 06/19/2014] [Indexed: 11/18/2022] Open
Abstract
Rationale Pulmonary arterial hypertension (PAH) is characterized by increased pulmonary vascular resistance leading to right ventricular failure and death. Recent studies have suggested that chronic inflammatory processes are involved in the pathogenesis of PAH. However, the molecular and cellular mechanisms driving inflammation have not been fully elucidated. Objectives To elucidate the roles of high mobility group box 1 protein (HMGB1), a ubiquitous DNA-binding protein with extracellular pro-inflammatory activity, in a rat model of PAH. Methods Male Sprague-Dawley rats were administered monocrotaline (MCT). Concentrations of HMGB1 in bronchoalveolar lavage fluid (BALF) and serum, and localization of HMGB1 in the lung were examined over time. The protective effects of anti-HMGB1 neutralizing antibody against MCT-induced PAH were tested. Results HMGB1 levels in BALF were elevated 1 week after MCT injection, and this elevation preceded increases of other pro-inflammatory cytokines, such as TNF-α, and the development of PAH. In contrast, serum HMGB1 levels were elevated 4 weeks after MCT injection, at which time the rats began to die. Immunohistochemical analyses indicated that HMGB1 was translocated to the extranuclear space in periarterial infiltrating cells, alveolar macrophages, and bronchial epithelial cells of MCT-injected rats. Anti-HMGB1 neutralizing antibody protected rats against MCT-induced lung inflammation, thickening of the pulmonary artery wall, and elevation of right ventricular systolic pressure, and significantly improved the survival of the MCT-induced PAH rats. Conclusions Our results identify extracellular HMGB1 as a promoting factor for MCT-induced PAH. The blockade of HMGB1 activity improved survival of MCT-induced PAH rats, and thus might be a promising therapy for the treatment of PAH.
Collapse
Affiliation(s)
- Yukari Sadamura-Takenaka
- Department of Pulmonary Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takashi Ito
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Satoshi Noma
- Department of Pulmonary Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yoko Oyama
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | | | - Ko-ichi Kawahara
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka, Japan
| | - Hiromasa Inoue
- Department of Pulmonary Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Ikuro Maruyama
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
- * E-mail:
| |
Collapse
|
22
|
Ahn KB, Jeon JH, Baik JE, Park OJ, Kang SS, Yun CH, Park JH, Han SH. Muramyl dipeptide potentiates staphylococcal lipoteichoic acid induction of cyclooxygenase-2 expression in macrophages. Microbes Infect 2014; 16:153-60. [DOI: 10.1016/j.micinf.2013.10.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 10/01/2013] [Accepted: 10/25/2013] [Indexed: 01/01/2023]
|
23
|
Exogenous rhTRX reduces lipid accumulation under LPS-induced inflammation. Exp Mol Med 2014; 46:e71. [PMID: 24406320 PMCID: PMC3909889 DOI: 10.1038/emm.2013.136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 09/03/2013] [Accepted: 09/09/2013] [Indexed: 01/01/2023] Open
Abstract
Redox-regulating molecule, recombinant human thioredoxin (rhTRX) which shows anti-inflammatory, and anti-oxidative effects against lipopolysaccharide (LPS)-stimulated inflammation and regulate protein expression levels. LPS-induced reactive oxygen intermediates (ROI) and NO production were inhibited by exogenous rhTRX. We identified up/downregulated intracellular proteins under the LPS-treated condition in exogenous rhTRX-treated A375 cells compared with non-LPS-treated cells via 2-DE proteomic analysis. Also, we quantitatively measured cytokines of in vivo mouse inflammation models using cytometry bead array. Exogenous rhTRX inhibited LPS-stimulated production of ROI and NO levels. TIP47 and ATP synthase may influence the inflammation-related lipid accumulation by affecting lipid metabolism. The modulation of skin redox environments during inflammation is most likely to prevent alterations in lipid metabolism through upregulation of TIP47 and ATP synthase and downregulation of inflammatory cytokines. Our results demonstrate that exogenous rhTRX has anti-inflammatory properties and intracellular regulatory activity in vivo and in vitro. Monitoring of LPS-stimulated pro-inflammatory conditions treated with rhTRX in A375 cells could be useful for diagnosis and follow-up of inflammation reduction related with candidate proteins. These results have a therapeutic role in skin inflammation therapy.
Collapse
|
24
|
Gomez I, Foudi N, Longrois D, Norel X. The role of prostaglandin E2 in human vascular inflammation. Prostaglandins Leukot Essent Fatty Acids 2013; 89:55-63. [PMID: 23756023 DOI: 10.1016/j.plefa.2013.04.004] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 04/04/2013] [Accepted: 04/05/2013] [Indexed: 10/26/2022]
Abstract
Prostaglandins (PG) are the product of a cascade of enzymes such as cyclooxygenases and PG synthases. Among PG, PGE2 is produced by 3 isoforms of PGE synthase (PGES) and through activation of its cognate receptors (EP1-4), this PG is involved in the pathophysiology of vascular diseases. Some anti-inflammatory drugs (e.g. glucocorticoids, nonsteroidal anti-inflammatory drugs) interfere with its metabolism or effects. Vascular cells can initiate many of the responses associated with inflammation. In human vascular tissue, PGE2 is involved in many physiological processes, such as increasing vascular permeability, cell proliferation, cell migration and control of vascular smooth muscle tone. PGE2 has been shown to contribute to the pathogenesis of atherosclerosis, abdominal aortic aneurysm but also in physiologic/adaptive processes such as angiogenesis. Understanding the roles of PGE2 and its cognate receptors in vascular diseases could help to identify diagnostic and prognostic biomarkers. In addition, from these recent studies new promising therapeutic approaches like mPGES-1 inhibition and/or EP4-antagonism should be investigated.
Collapse
Affiliation(s)
- I Gomez
- INSERM, U698, Paris F-75018, France; University Paris Nord, UMR-S698, Paris F-75018, France
| | | | | | | |
Collapse
|
25
|
Leclerc P, Wähämaa H, Idborg H, Jakobsson PJ, Harris HE, Korotkova M. IL-1β/HMGB1 complexes promote The PGE2 biosynthesis pathway in synovial fibroblasts. Scand J Immunol 2013; 77:350-60. [PMID: 23488692 PMCID: PMC3670302 DOI: 10.1111/sji.12041] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 02/15/2013] [Indexed: 01/09/2023]
Abstract
PGE2 is a potent lipid mediator of pain and oedema found elevated in RA. Microsomal prostaglandin E synthase-1 (mPGES-1) is a terminal enzyme of the PGE2 pathway inducible by proinflammatory cytokines. mPGES-1 is markedly upregulated in RA synovial tissue despite antirheumatic treatments, suggesting that multiple inflammatory stimuli contribute to its induction. High-mobility group box chromosomal protein 1 (HMGB1) is known to induce inflammation both by direct interaction with TLR4 and by enhancement of other proinflammatory molecules signalling, through complex formation. The high expression of extracellular HMGB1 within the inflamed synovium, implies its pro-arthritogenic role in RA. We aimed to investigate the effects of IL-1β/HMGB1 complexes on mPGES-1 and other enzymes of the PGE2 pathway in synovial fibroblasts (SFs) from patients with arthritis. Furthermore, we studied the effect of COX-2 inhibition and IL-1RI antagonism on prostanoid and cytokine production by SFs. Stimulation of SFs with HMGB1 in complex with suboptimal amounts of IL-1β significantly increased mPGES-1 and COX-2 expressions as well as PGE2 production, as compared to treatment with HMGB1 or IL-1β alone. Furthermore, NS-398 reduced the production of IL-6 and IL-8, thus indicating that IL-1β/HMGB1 complexes modulate cytokine production in part through prostanoid synthesis. Treatment with IL-1RA completely abolished the induced PGE2 and cytokine production, suggesting an effect mediated through IL-1RI. IL-1β/HMGB1 complexes promote the induction of mPGES-1, COX-2 and PGE2 in SF. The amplification of the PGE2 biosynthesis pathway by HMGB1 might constitute an important pathogenic mechanism perpetuating inflammatory and destructive activities in rheumatoid arthritis.
Collapse
Affiliation(s)
- P Leclerc
- Rheumatology Research Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
26
|
Iori V, Maroso M, Rizzi M, Iyer AM, Vertemara R, Carli M, Agresti A, Antonelli A, Bianchi ME, Aronica E, Ravizza T, Vezzani A. Receptor for Advanced Glycation Endproducts is upregulated in temporal lobe epilepsy and contributes to experimental seizures. Neurobiol Dis 2013; 58:102-14. [PMID: 23523633 DOI: 10.1016/j.nbd.2013.03.006] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 03/05/2013] [Accepted: 03/13/2013] [Indexed: 01/07/2023] Open
Abstract
Toll-like receptor 4 (TLR4) activation in neuron and astrocytes by High Mobility Group Box 1 (HMGB1) protein is a key mechanism of seizure generation. HMGB1 also activates the Receptor for Advanced Glycation Endproducts (RAGE), but it was unknown whether RAGE activation contributes to seizures or to HMGB1 proictogenic effects. We found that acute EEG seizures induced by 7ng intrahippocampal kainic acid (KA) were significantly reduced in Rage-/- mice relative to wild type (Wt) mice. The proictogenic effect of HMGB1 was decreased in Rage-/- mice, but less so, than in Tlr4-/- mice. In a mouse mesial temporal lobe epilepsy (mTLE) model, status epilepticus induced by 200ng intrahippocampal KA and the onset of the spontaneous epileptic activity were similar in Rage-/-, Tlr4-/- and Wt mice. However, the number of hippocampal paroxysmal episodes and their duration were both decreased in epileptic Rage-/- and Tlr4-/- mice vs Wt mice. All strains of epileptic mice displayed similar cognitive deficits in the novel object recognition test vs the corresponding control mice. CA1 neuronal cell loss was increased in epileptic Rage-/- vs epileptic Wt mice, while granule cell dispersion and doublecortin (DCX)-positive neurons were similarly affected. Notably, DCX neurons were preserved in epileptic Tlr4-/- mice. We did not find compensatory changes in HMGB1-related inflammatory signaling nor in glutamate receptor subunits in Rage-/- and Tlr4-/- naïve mice, except for ~20% NR2B subunit reduction in Rage-/- mice. RAGE was induced in neurons, astrocytes and microvessels in human and experimental mTLE hippocampi. We conclude that RAGE contributes to hyperexcitability underlying acute and chronic seizures, as well as to the proictogenic effects of HMGB1. RAGE and TLR4 play different roles in the neuropathologic sequelae developing after status epilepticus. These findings reveal new molecular mechanisms underlying seizures, cell loss and neurogenesis which involve inflammatory pathways upregulated in human epilepsy.
Collapse
Affiliation(s)
- Valentina Iori
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Lercanidipine and labedipinedilol-A attenuate lipopolysaccharide/interferon-γ-induced inflammation in rat vascular smooth muscle cells through inhibition of HMGB1 release and MMP-2, 9 activities. Atherosclerosis 2013; 226:364-72. [DOI: 10.1016/j.atherosclerosis.2012.12.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 11/23/2012] [Accepted: 12/10/2012] [Indexed: 01/30/2023]
|
28
|
Pessolano LG, Sullivan CP, Seidl SE, Rich CB, Liscum L, Stone PJ, Sipe JD, Schreiber BM. Trafficking of endogenous smooth muscle cell cholesterol: a role for serum amyloid A and interleukin-1β. Arterioscler Thromb Vasc Biol 2012; 32:2741-50. [PMID: 22995521 DOI: 10.1161/atvbaha.112.300243] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Intracellular cholesterol distribution impacts cell function; however, processes influencing endogenous cholesterol trafficking remain largely unknown. Atherosclerosis is associated with vascular inflammation and these studies address the role of inflammatory mediators on smooth muscle cell cholesterol trafficking. METHODS AND RESULTS Interestingly, in the absence of an exogenous cholesterol source, serum amyloid A increased [(14)C] oleic acid incorporation into cholesteryl ester in rat smooth muscle cells, suggesting endogenous cholesterol trafficking to the endoplasmic reticulum. [(3)H] cholesteryl ester accumulated in cells prelabeled with [(3)H] cholesterol, confirming that serum amyloid A mediated the movement of endogenous cholesterol. Cholesterol movement was dependent upon functional endolysosomes. The cholesterol oxidase-sensitive pool of cholesterol decreased in serum amyloid A-treated cells. Furthermore, the mechanism whereby serum amyloid A induced cholesterol trafficking was determined to be via activation of expression of secretory phospholipase A(2), group IIA (sPLA(2)) and sPLA(2)-dependent activation of sphingomyelinase. Interestingly, although neither tumor necrosis factor-α nor interferon-γ induced cholesterol trafficking, interleukin-1β induced [(14)C] cholesteryl ester accumulation that was also dependent upon sPLA(2) and sphingomyelinase activities. Serum amyloid A activates smooth muscle cell interleukin-1β expression, and although the interleukin-1-receptor antagonist inhibited the interleukin-1β-induced cholesterol trafficking, it had no effect on the movement of cholesterol mediated by serum amyloid A. CONCLUSIONS These data support a role for inflammation in endogenous smooth muscle cell cholesterol trafficking from the plasma membrane to the endoplasmic reticulum.
Collapse
Affiliation(s)
- Lawrence G Pessolano
- Department of Biochemistry, Boston University School of Medicine, K207, 72 East Concord Street, Boston, MA 02118, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Dohi T, Miyauchi K, Ohkawa R, Nakamura K, Thuboi S, Ogita M, Miyazaki T, Nishino A, Yokoyama K, Kurata T, Yatomi Y, Daida H. Higher lipoprotein-associated phospholipase A2 levels are associated with coronary atherosclerosis documented by coronary angiography. Ann Clin Biochem 2012; 49:527-33. [PMID: 22933444 DOI: 10.1258/acb.2012.011252] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Lipoprotein-associated phospholipase A2 (Lp-PLA2) has been proposed as an inflammatory marker of cardiovascular disease. The present study investigates associations between Lp-PLA2 and other important biomarkers in Japanese patients with coronary artery disease. METHODS We measured Lp-PLA2 levels in 141 consecutive patients (age 62.6 ± 3.8 years; men 69.2%) with angiographic evidence of coronary artery disease (acute coronary syndrome [ACS]; n = 38), stable angina pectoris (SAP; n = 72) or with angiographically normal coronary arteries (NCA; n = 31). RESULTS Levels of Lp-PLA2 significantly correlated with low-density lipoprotein-cholesterol (r = 0.302), homocysteine (r = 0.528) and paraoxonase (r = 0.401) in all patients (all P < 0.01). Levels of Lp-PLA2 were significantly higher in patients with coronary atherosclerosis (ACS and SAP) than with NCA (P < 0.05). Levels of highly sensitive C-reactive protein were significantly higher in patients with ACS than with SAP and NCA (both P < 0.05). Multivariate logistic regression analyses revealed that higher Lp-PLA2 levels were independently associated with coronary atherosclerosis (odds ratio: 1.058; 95% confidence interval: 1.012-1.121; P = 0.001). CONCLUSIONS Higher Lp-PLA2 levels are associated with coronary atherosclerosis independently of traditional coronary risk factors. Thus, Lp-PLA2 is a novel biomarker of coronary atherosclerosis in Japanese patients.
Collapse
Affiliation(s)
- Tomotaka Dohi
- Department of Cardiovascular Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Tokyo 113-0033, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW The phospholipase A2 (PLA2) family of proteins includes lipolytic enzymes that liberate the sn-2 fatty acyl chains from phospholipids to yield nonesterified fatty acids and lysophospholipids. The purpose of this review is to discuss recent findings showing distinct roles of several of these PLA2 enzymes in inflammatory metabolic diseases such as diabetes and atherosclerosis. RECENT FINDINGS The group 1B PLA2 digestion of phospholipids in the intestinal lumen facilitates postprandial lysophospholipid absorption, which suppresses hepatic fatty acid oxidation leading to increased VLDL synthesis, decreased glucose tolerance, and promotion of tissue lipid deposition to accentuate diet-induced hyperlipidemia, diabetes, and obesity. Other secretory PLA2s promote inflammatory metabolic diseases by generating bioactive lipid metabolites to induce inflammatory cytokine production, whereas the major intracellular PLA2s, cPLA2α, and iPLA2, generate arachidonic acid and lysophosphatic acid in response to extracellular stimuli to activate leukocyte chemotactic response. SUMMARY Each member of the PLA2 family of enzymes serves a distinct role in generating active lipid metabolites that promote inflammatory metabolic diseases including atherosclerosis, hyperlipidemia, obesity, and diabetes. The development of specific drugs that target one or more of these PLA2 enzymes may be novel strategies for treatment of these chronic inflammatory metabolic disorders.
Collapse
Affiliation(s)
- David Y Hui
- Department of Pathology, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
31
|
Al-Zoairy R, Melmer A, Ress C, Laimer M, Kaser S, Ebenbichler C. Lipid profile changes after pronounced weight loss induced by bariatric surgery. ACTA ACUST UNITED AC 2012. [DOI: 10.2217/clp.12.9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
32
|
Lee W, Ku SK, Bae JW, Bae JS. Inhibitory effects of lycopene on HMGB1-mediated pro-inflammatory responses in both cellular and animal models. Food Chem Toxicol 2012; 50:1826-33. [PMID: 22429818 DOI: 10.1016/j.fct.2012.03.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Revised: 02/27/2012] [Accepted: 03/02/2012] [Indexed: 01/01/2023]
Abstract
High mobility group box 1 (HMGB1) mediates proinflammatory responses in inflammatory diseases. Lycopene found in tomatoes and tomato products has anti-oxidant, anti-cancer and antiinflammatory effects. The potential anti-inflammatory roles of lycopene in HMGB1-mediated proinflammatory responses in both primary human umbilical vein endothelial cells (HUVECs) and animal were investigated. The anti-inflammatory effects of lycopene were determined including permeability, monocyte adhesion and migration, and activation of proinflammatory proteins and HMGB1 receptors on HMGB1 activated HUVECs. In the in vivo model, the anti-inflammatory effect of lycopene was assessed by monitoring vascular permeability and migration of leukocytes to the peritoneal cavity of mice injected with lycopene. Lycopene inhibited lipopolysaccharide (LPS)-mediated release of HMGB1, expression of HMGB1-mediated tumor necrosis factor (TNF)-secretory phospholipase A2 (sPLA2)-IIA, and HMGB1-mediated pro-inflammatory signaling responses in endothelial cells. It did this through down-regulation of cell surface expression of cell adhesion molecules (CAMs), HMGB1 receptors, toll-like receptor (TLR)-2, and -4, and receptors for advanced glycation end products (RAGE). These findings suggest that lycopene promotes barrier integrity, inhibits monocyte adhesion and migration to HMGB1 activating HUVECs by blocking activation of proinflammatory cytokines and expression of CAMs and HMGB1 receptors, thereby showing its usefulness as a therapy for vascular inflammatory diseases.
Collapse
Affiliation(s)
- Wonhwa Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | | | | | | |
Collapse
|
33
|
Xiang L, Lu S, Fuller W, Aneja A, Russell GV, Jones LB, Hester R. Impaired blood pressure recovery to hemorrhage in obese Zucker rats with orthopedic trauma. Am J Physiol Heart Circ Physiol 2011; 302:H340-8. [PMID: 22003055 DOI: 10.1152/ajpheart.00439.2011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have shown that obese Zucker rats with orthopedic trauma (OZT) exhibit a loss of arteriolar tone in skeletal muscle. We hypothesize that the loss of arteriolar tone in OZT blunts vasoconstrictor responses to hemorrhage, resulting in an impaired blood pressure recovery. Orthopedic trauma was induced with soft tissue injury and local injection of bone components in both hindlimbs in lean (LZT) and OZT (11-13 wk). One day after the orthopedic trauma, blood pressure responses following hemorrhage were measured in conscious control lean, control obese, LZT, and OZT. In another set of experiments, the spinotrapezius muscle of control and trauma animals was prepared for microcirculatory observation. Arteriolar responses to phenylephrine (PE) or hemorrhage were determined. Hemorrhage resulted in similar blood pressure responses in control animals and LZT, but the blood pressure recovery following hemorrhage was blunted in the OZT. In the spinotrapezius, OZT exhibited decreased arteriolar tone and blunted vasoconstrictor responses to PE and hemorrhage. Treatment with glibenclamide improved the blood pressure recovery in the conscious OZT and improved the arteriolar tone, and PE induced vasoconstriction in the spinotrapezius of the OZT. Thus, ATP-dependent K(+) channel-mediated loss of arteriolar tone in OZT blunts the arteriolar constriction to hemorrhage, resulting in impaired blood pressure recovery.
Collapse
Affiliation(s)
- Lusha Xiang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, 39216-4505, USA.
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Oxidative stress and associated reactive oxygen species can modify lipids, proteins, carbohydrates, and nucleic acids, and induce the mitochondrial permeability transition, providing a signal leading to the induction of autophagy, apoptosis, and necrosis. High-mobility group box 1 (HMGB1) protein, a chromatin-binding nuclear protein and damage-associated molecular pattern molecule, is integral to oxidative stress and downstream apoptosis or survival. Accumulation of HMGB1 at sites of oxidative DNA damage can lead to repair of the DNA. As a redox-sensitive protein, HMGB1 contains three cysteines (Cys23, 45, and 106). In the setting of oxidative stress, it can form a Cys23-Cys45 disulfide bond; a role for oxidative homo- or heterodimerization through the Cys106 has been suggested for some of its biologic activities. HMGB1 causes activation of nicotinamide adenine dinucleotide phosphate oxidase and increased reactive oxygen species production in neutrophils. Reduced and oxidized HMGB1 have different roles in extracellular signaling and regulation of immune responses, mediated by signaling through the receptor for advanced glycation end products and/or Toll-like receptors. Antioxidants such as ethyl pyruvate, quercetin, green tea, N-acetylcysteine, and curcumin are protective in the setting of experimental infection/sepsis and injury including ischemia-reperfusion, partly through attenuating HMGB1 release and systemic accumulation.
Collapse
Affiliation(s)
- Daolin Tang
- The DAMP Laboratory, Department of Surgery, G.27 Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA.
| | | | | | | |
Collapse
|
35
|
Nouvelles cibles thérapeutiques du sepsis — HMGB1: bon acteur mais mauvais marqueur ? MEDECINE INTENSIVE REANIMATION 2011. [DOI: 10.1007/s13546-010-0134-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
36
|
Mallat Z, Lambeau G, Tedgui A. Lipoprotein-associated and secreted phospholipases A₂ in cardiovascular disease: roles as biological effectors and biomarkers. Circulation 2010; 122:2183-200. [PMID: 21098459 DOI: 10.1161/circulationaha.110.936393] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Ziad Mallat
- INSERM, Paris-Cardiovascular Research Center, Université Paris Descartes, UMR, Paris, France
| | | | | |
Collapse
|
37
|
Xiang L, Hester RL, Fuller WL, Sebai ME, Mittwede PN, Jones EK, Aneja A, Russell GV. Orthopedic trauma-induced pulmonary injury in the obese Zucker rat. Microcirculation 2010; 17:650-9. [PMID: 21044219 PMCID: PMC3270564 DOI: 10.1111/j.1549-8719.2010.00061.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Obese subjects with orthopedic trauma exhibit increased inflammation and an increased risk of pulmonary edema. Prostaglandin E(2) (PGE(2) ) production is elevated during inflammation and associated with increased vascular permeability. We hypothesize that pulmonary edema in obesity following orthopedic trauma is due to elevated PGE(2) and resultant increases in pulmonary permeability. METHODS Orthopedic trauma was induced in both hindlimbs in lean (LZ) and obese Zucker rats (OZ). On the following day, plasma interleukin-6 (IL-6) and PGE(2) levels, pulmonary edema, and pulmonary gas exchange capability were compared between groups: LZ, OZ, LZ with trauma (LZT), and OZ with trauma (OZT). Vascular permeability in isolated lungs was measured in LZ and OZ before and after application of PGE(2) . RESULTS As compared with the other groups, the OZT exhibited elevated plasma IL-6 and PGE(2) levels, increased lung wet/dry weight ratio and bronchoalveolar protein concentration, and an impaired pulmonary gas exchange. Indomethacin treatment normalized plasma PGE(2) levels and pulmonary edema. Basal pulmonary permeability in isolated lungs was higher in OZ than LZ, with a further increase in permeability following treatment with PGE(2) . CONCLUSIONS These results suggest that pulmonary edema in OZ following orthopedic trauma is due to an elevated PGE(2) and resultant increases in pulmonary permeability.
Collapse
Affiliation(s)
- Lusha Xiang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi 39216-4505, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Zhu S, Li W, Ward MF, Sama AE, Wang H. High mobility group box 1 protein as a potential drug target for infection- and injury-elicited inflammation. ACTA ACUST UNITED AC 2010; 9:60-72. [PMID: 19906009 DOI: 10.2174/187152810791292872] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Accepted: 11/03/2009] [Indexed: 12/11/2022]
Abstract
In response to infection or injury, a ubiquitous nucleosomal protein, HMGB1 is secreted actively by innate immune cells, and / or released passively by injured/damaged cells. Subsequently, extracellular HMGB1 alerts, recruits, and activates various innate immune cells to sustain a rigorous inflammatory response. A growing number of HMGB1 inhibitors ranging from neutralizing antibodies, endogenous hormones, to medicinal herb-derived small molecule HMGB1 inhibitors (such as nicotine, glycyrrhizin, tanshinones, and EGCG) are proven protective against lethal infection and ischemic injury. Here we review emerging evidence that support extracellular HMGB1 as a proinflammatory alarmin(g) danger signal, and discuss a wide array of HMGB1 inhibitors as potential therapeutic agents for sepsis and ischemic injury.
Collapse
Affiliation(s)
- Shu Zhu
- The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, NY 11030, USA
| | | | | | | | | |
Collapse
|
39
|
Association of carotid plaque Lp-PLA(2) with macrophages and Chlamydia pneumoniae infection among patients at risk for stroke. PLoS One 2010; 5:e11026. [PMID: 20543948 PMCID: PMC2882946 DOI: 10.1371/journal.pone.0011026] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Accepted: 05/11/2010] [Indexed: 11/20/2022] Open
Abstract
Background We previously showed that the burden of Chlamydia pneumoniae in carotid plaques was significantly associated with plaque interleukin (IL)-6, and serum IL-6 and C-reactive protein (CRP), suggesting that infected plaques contribute to systemic inflammatory markers in patients with stroke risk. Since lipoprotein-associated phospholipase A2 (Lp-PLA2) mediates inflammation in atherosclerosis, we hypothesized that serum Lp-PLA2 mass and activity levels and plaque Lp-PLA2 may be influenced by plaque C. pneumoniae infection. Methodology/Principal Findings Forty-two patients underwent elective carotid endarterectomy. Tissue obtained at surgery was stained by immunohistochemistry for Lp-PLA2 grade, macrophages, IL-6, C. pneumoniae and CD4+ and CD8+ cells. Serum Lp-PLA2 activity and mass were measured using the colorimetric activity method (CAM™) and ELISA, respectively. Serum homocysteine levels were measured by HPLC. Eleven (26.2%) patients were symptomatic with transient ischemic attacks. There was no correlation between patient risk factors (smoking, coronary artery disease, elevated cholesterol, diabetes, obesity, hypertension and family history of genetic disorders) for atherosclerosis and serum levels or plaque grade for Lp-PLA2. Plaque Lp-PLA2 correlated with serum homocysteine levels (p = 0.013), plaque macrophages (p<0.01), and plaque C. pneumoniae (p<0.001), which predominantly infected macrophages, co-localizing with Lp-PLA2. Conclusions The significant association of plaque Lp-PLA2 with plaque macrophages and C. pneumoniae suggests an interactive role in accelerating inflammation in atherosclerosis. A possible mechanism for C. pneumoniae in the atherogenic process may involve infection of macrophages that induce Lp-PLA2 production leading to upregulation of inflammatory mediators in plaque tissue. Additional in vitro and in vivo research will be needed to advance our understanding of specific C. pneumoniae and Lp-PLA2 interactions in atherosclerosis.
Collapse
|
40
|
Kwiatkowski S, Torbé A, Dołegowska B, Błogowski W, Czajka R, Chlubek D, Rzepka R. Isoprostanes 8-iPF2alpha-III: risk markers of premature rupture of fetal membranes? Biomarkers 2009; 14:406-13. [PMID: 19548773 DOI: 10.1080/13547500903045583] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
AIMS Isoprostanes may serve as sensitive and specific markers of in vivo oxidative stress intensity. We wanted to determine, whether or not isoprostane concentration may be considered as a risk marker of premature rupture of fetal membranes (PROM). METHODS On the basis of the presence of PROM and gestational maturity, a total of 128 patients were divided into: (1) preterm PROM (pPROM) group; (2) PROM at term group; (3) control preterm (C1) group and (4) control at term (C2) group. The concentrations of 8-iPF(2alpha)-III were determined using the enzyme-linked immunosorbent assay method. RESULTS The mean free isoprostane concentrations, examined in amniotic fluid and maternal plasma in the PROM at term patients were significantly higher than in C2 individuals (p < 0.01). The mean concentrations of free 8-iPF(2alpha)-III measured in blood plasma from women in the C1 group were significantly lower than in patients from the pPROM, PROM at term and C2 groups (p < 0.001, p < 0.00001 and p < 0.00001, respectively). CONCLUSION The measurement of free isoprostane concentration in maternal plasma and amniotic fluid may be considered as a laboratory marker of a PROM-risk pregnancy.
Collapse
Affiliation(s)
- Sebastian Kwiatkowski
- Department of Obstetrics and Gynecology, Pomeranian Medical University, Szczecin, Poland
| | | | | | | | | | | | | |
Collapse
|
41
|
Farmer DG, Kennedy S. RAGE, vascular tone and vascular disease. Pharmacol Ther 2009; 124:185-94. [DOI: 10.1016/j.pharmthera.2009.06.013] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 06/29/2009] [Indexed: 12/13/2022]
|
42
|
Sullivan CP, Seidl SE, Rich CB, Raymondjean M, Schreiber BM. Secretory phospholipase A2, group IIA is a novel serum amyloid A target gene: activation of smooth muscle cell expression by an interleukin-1 receptor-independent mechanism. J Biol Chem 2009; 285:565-75. [PMID: 19850938 DOI: 10.1074/jbc.m109.070565] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Atherosclerosis is a multifactorial vascular disease characterized by formation of inflammatory lesions. Elevated circulating acute phase proteins indicate disease risk. Serum amyloid A (SAA) is one such marker but its function remains unclear. To determine the role of SAA on aortic smooth muscle cell gene expression, a preliminary screen of a number of genes was performed and a strong up-regulation of expression of secretory phospholipase A(2), group IIA (sPLA(2)) was identified. The SAA-induced increase in sPLA(2) was validated by real time PCR, Western blot analysis, and enzyme activity assays. Demonstrating that SAA increased expression of sPLA(2) heteronuclear RNA and that inhibiting transcription eliminated the effect of SAA on sPLA(2) mRNA suggested that the increase was transcriptional. Transient transfections and electrophoretic mobility shift assays identified CAAT enhancer-binding protein (C/EBP) and nuclear factor kappaB (NFkappaB) as key regulatory sites mediating the induction of sPLA(2). Moreover, SAA activated the inhibitor of NF-kappaB kinase (IKK) in cultured smooth muscle cells. Previous reports showed that interleukin (IL)-1beta up-regulates Pla2g2a gene transcription via C/EBPbeta and NFkappaB. Interestingly, SAA activated smooth muscle cell IL-1beta mRNA expression, however, blocking IL-1 receptors had no effect on SAA-mediated activation of sPLA(2) expression. Thus, the observed changes in sPLA(2) expression were not secondary to SAA-induced IL-1 receptor activation. The association of SAA with high density lipoprotein abrogated the SAA-induced increase in sPLA(2) expression. These data suggest that during atherogenesis, SAA can amplify the involvement of smooth muscle cells in vascular inflammation and that this can lead to deposition of sPLA(2) and subsequent local changes in lipid homeostasis.
Collapse
Affiliation(s)
- Christopher P Sullivan
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | |
Collapse
|
43
|
Iyer JP, Srivastava PK, Dev R, Dastidar SG, Ray A. Prostaglandin E(2) synthase inhibition as a therapeutic target. Expert Opin Ther Targets 2009; 13:849-65. [PMID: 19530988 DOI: 10.1517/14728220903018932] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Most NSAIDs function by inhibiting biosynthesis of PGE(2) by inhibition of COX-1 and/or COX-2. Since COX-1 has a protective function in the gastro-intestinal tract (GIT), non-selective inhibition of both cycloxy genases leads to moderate to severe gastro-intestinal intolerance. Attempts to identify selective inhibitors of COX-2, led to the identification of celecoxib and rofecoxib. However, long-term use of these drugs has serious adverse effects of sudden myocardial infarction and thrombosis. Drug-mediated imbalance in the levels of prostaglandin I(2) (PGI(2)) and thromboxane A(2) (TXA(2)) with a bias towards TXA(2) may be the primary reason for these events. This resulted in the drugs being withdrawn from the market, leaving a need for an effective and safe anti-inflammatory drug. METHODS Recently, the focus of research has shifted to enzymes downstream of COX in the prosta glandin biosynthetic pathway such as prostaglandin E(2) synthases. Microsomal prostaglandin E(2) synthase-1 (mPGES-1) specifically isomerizes PGH(2) to PGE(2), under inflammatory conditions. In this review, we examine the biology of mPGES-1 and its role in disease. Progress in designing molecules that can selectively inhibit mPGES-1 is reviewed. CONCLUSION mPGES-1 has the potential to be a target for anti-inflammatory therapy, devoid of adverse GIT and cardiac effects and warrants further investigation.
Collapse
Affiliation(s)
- Jitesh P Iyer
- Department of Pharmacology, New Drug Discovery Research, Ranbaxy Research Laboratories, Plot No-20, Sector-18, Udyog Vihar, Gurgaon, Haryana, India-122015
| | | | | | | | | |
Collapse
|
44
|
Kitsiouli E, Nakos G, Lekka ME. Phospholipase A2 subclasses in acute respiratory distress syndrome. Biochim Biophys Acta Mol Basis Dis 2009; 1792:941-53. [PMID: 19577642 DOI: 10.1016/j.bbadis.2009.06.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 06/25/2009] [Accepted: 06/25/2009] [Indexed: 01/12/2023]
Abstract
Phospholipases A2 (PLA2) catalyse the cleavage of fatty acids esterified at the sn-2 position of glycerophospholipids. In acute lung injury-acute respiratory distress syndrome (ALI-ARDS) several distinct isoenzymes appear in lung cells and fluid. Some are capable to trigger molecular events leading to enhanced inflammation and lung damage and others have a role in lung surfactant recycling preserving lung function: Secreted forms (groups sPLA2-IIA, -V, -X) can directly hydrolyze surfactant phospholipids. Cytosolic PLA2 (cPLA2-IVA) requiring Ca2+ has a preference for arachidonate, the precursor of eicosanoids which participate in the inflammatory response in the lung. Ca(2+)-independent intracellular PLA2s (iPLA2) take part in surfactant phospholipids turnover within alveolar cells. Acidic Ca(2+)-independent PLA2 (aiPLA2), of lysosomal origin, has additionally antioxidant properties, (peroxiredoxin VI activity), and participates in the formation of dipalmitoyl-phosphatidylcholine in lung surfactant. PAF-AH degrades PAF, a potent mediator of inflammation, and oxidatively fragmented phospholipids but also leads to toxic metabolites. Therefore, the regulation of PLA2 isoforms could be a valuable approach for ARDS treatment.
Collapse
Affiliation(s)
- Eirini Kitsiouli
- Department of Biological Applications and Technologies, School of Sciences and Technologies, University of Ioannina, Greece
| | | | | |
Collapse
|
45
|
Chen D, Wei Y, Li X, Epstein S, Wolosin JM, Asbell P. sPLA2-IIa is an inflammatory mediator when the ocular surface is compromised. Exp Eye Res 2009; 88:880-8. [PMID: 19116146 PMCID: PMC4001256 DOI: 10.1016/j.exer.2008.11.035] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 11/19/2008] [Accepted: 11/21/2008] [Indexed: 10/21/2022]
Abstract
sPLA2-IIa is an enzyme at high concentration in tears that has been known as an innate barrier of the ocular surface against microbial infection. sPLA2-IIa and other enzymes in the same protein family are known to hydrolyze fatty acids resulting in the generation of free arachidonic acid (AA) and lysophospholipids, which are the precursors of pro-inflammatory lipid mediators, such as PGE(2). sPLA2-IIa has been shown to be an inflammatory mediator in non-ocular inflammatory diseases such as rheumatoid arthritis (RA). It was also found to be increased in the tears of the patients with dry eye disease, chronic blepharitis and contact lens intolerance. However, the role of sPLA2-IIa in chronic ocular surface inflammation has yet to be determined. In the current study, we examined the potential role of sPLA2-IIa in inflammation of ocular surface diseases. Our results show that the activity of sPLA2-IIa was significantly increased in tears from dry eye disease patients compared with that from normal subjects. Also, sPLA2-IIa stimulated the production of PGE(2) in ocular surface epithelial cell cultures. The stimulating effect was markedly enhanced when the cells or tissues were pre-compromised with TNF-alpha, IL-1beta or desiccation. Furthermore, sPLA2-IIa stimulated inflammatory cytokine production in the ocular surface epithelial cell cultures in vitro. To our knowledge, this is the first report regarding the role of sPLA2-IIa as an inflammatory mediator in ocular surface inflammation. These findings indicate that sPLA2-IIa may play an important role in chronic ocular surface inflammation, especially when the ocular surface is compromised.
Collapse
Affiliation(s)
- Dongmei Chen
- Department of Ophthalmology, The Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Yi Wei
- Department of Ophthalmology, The Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Xiaohong Li
- Department of Ophthalmology, The Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Seth Epstein
- Department of Ophthalmology, The Mount Sinai School of Medicine, New York, NY 10029, USA
| | - J. Mario Wolosin
- Department of Ophthalmology, The Mount Sinai School of Medicine, New York, NY 10029, USA
- Black Family Stem Cell Institute, The Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Penny Asbell
- Department of Ophthalmology, The Mount Sinai School of Medicine, New York, NY 10029, USA
| |
Collapse
|
46
|
Yuk JM, Yang CS, Shin DM, Kim KK, Lee SK, Song YJ, Lee HM, Cho CH, Jeon BH, Jo EK. A dual regulatory role of apurinic/apyrimidinic endonuclease 1/redox factor-1 in HMGB1-induced inflammatory responses. Antioxid Redox Signal 2009; 11:575-88. [PMID: 18715145 DOI: 10.1089/ars.2008.2196] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Apurinic/apyrimidinic endonuclease 1/Redox factor-1 (APE1) is a multifunctional protein involved in reduction-oxidation regulation. High-mobility group box 1 (HMGB1) is released by necrotic cells and various inflammatory stimuli, acting as an inflammatory marker in sepsis and autoimmune diseases. Here, we report the dual regulatory role of APE1 in inflammatory signaling to extracellular HMGB1 or in the release of endogenous HMGB1 in human monocytes/macrophages. Forced cytoplasmic overexpression of APE1 profoundly attenuated the upregulation of HMGB1-mediated reactive oxygen species generation, cytokine secretion, and cyclooxygenase-2 expression by primary monocytes and macrophage-like THP-1 cell lines. In addition, HMGB1-induced activation of p38 and c-Jun N-terminal kinase (JNK), but not extracellular signal-regulated kinase 1/2, was strongly abrogated by the overexpression of APE1. The activation of apoptosis signal-regulating kinase 1 was required for both the p38 and JNK activation challenge with HMGB1. The extracellular release of HMGB1 by activated macrophages was inhibited by APE1 transfection. Small interfering RNA (siRNA) knockdown of endogenous APE1 impaired HMGB1-mediated cytokine expression and MAPK activation in THP-1 cells. HMGB1 stimulation induced the translocation of APE1 to the nucleus of the cell. In addition, APE1 silencing via siRNA transfection inhibited both the nuclear and cytoplasmic expression of APE1. These data identify APE1 as a novel dual regulator of inflammatory signaling to HMGB1 by human monocytes/macrophages. The modulation of cytosolic APE1 expression might be useful as a potential therapeutic modality for the treatment of inflammatory or autoimmune diseases.
Collapse
Affiliation(s)
- Jae-Min Yuk
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lin CC, Lin WN, Wang WJ, Sun CC, Tung WH, Wang HH, Yang CM. Functional coupling expression of COX-2 and cPLA2 induced by ATP in rat vascular smooth muscle cells: role of ERK1/2, p38 MAPK, and NF-kappaB. Cardiovasc Res 2009; 82:522-31. [PMID: 19233864 DOI: 10.1093/cvr/cvp069] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Vascular smooth muscle cells (VSMCs) that function as synthetic units play important roles in inflammatory diseases such as atherosclerosis and angiogenesis. As extracellular nucleotides such as ATP have been shown to act via activation of P(2) purinoceptors implicated in various inflammatory diseases, we hypothesized that extracellular nucleotides contribute to vascular diseases via upregulated expression of inflammatory proteins, such as cyclooxygenase (COX-2) and cytosolic phospholipase A2 (cPLA2) in VSMCs. METHODS AND RESULTS Western blotting, promoter assay, RT-PCR, and PGE2 immunoassay revealed that ATPgammaS induced expression of COX-2 and prostaglandin (PGE2) synthesis through the activation of p42/p44 MAPK (mitogen-activated protein kinase), p38 MAPK, and nuclear factor-kappaB (NF-kappaB). These responses were attenuated by inhibitors of MAPK/ERK kinase (MEK1/2; U0126), p38 MAPK (SB202190), and NF-kappaB (helenalin), or by tranfection with dominant negative mutants of p42, p38, IkappaB kinase (IKK)alpha, and IKKbeta. Furthermore, the ATPgammaS-stimulated translocation of NF-kappaB into the nucleus and degradation of IkappaBalpha was blocked by U0126 and helenalin. In addition, the ATPgammaS-stimulated cPLA2 expression was inhibited by U0126, SB202190, helenalin, celecoxib (a selective COX-2 inhibitor), and PGE2 receptor antagonists (AH6809, GW627368X, and SC-19220). However, the inhibitory effect of celecoxib on cPLA2 expression was reversed by addition of exogenous PGE2. CONCLUSION Our results suggest that in VSMCs, activation of p42/p44 MAPK, p38 MAPK, and NF-kappaB is essential for ATPgammaS-induced COX-2 expression and PGE2 synthesis. Newly synthesized PGE2 was observed to act as an autocrine signal contributing to cPLA2 expression, which may be implicated in inflammatory responses. Collectively, our findings provide insights into the correlation between COX-2 and cPLA2 expression in ATPgammaS-stimulated VSMCs with similar molecular mechanisms and functional coupling to amplify the occurrence of vessel disease-related vascular inflammation.
Collapse
Affiliation(s)
- Chih-Chung Lin
- Department of Anesthetics, Chang Gung University, Tao-Yuan, Taiwan, Republic of China
| | | | | | | | | | | | | |
Collapse
|
48
|
Qing X, Pitashny M, Thomas DB, Barrat F, Hogarth MP, Putterman C. Pathogenic anti-DNA antibodies modulate gene expression in mesangial cells: involvement of HMGB1 in anti-DNA antibody-induced renal injury. Immunol Lett 2008; 121:61-73. [PMID: 18822317 PMCID: PMC2677706 DOI: 10.1016/j.imlet.2008.08.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Revised: 08/24/2008] [Accepted: 08/25/2008] [Indexed: 11/23/2022]
Abstract
Although anti-DNA antibodies have been decisively linked to the pathogenesis of lupus nephritis, the mechanisms have not been conclusively determined. Recently, we reported that anti-DNA antibodies may contribute to kidney damage by upregulation of proinflammatory genes in mesangial cells (MC), a process involving both Fc receptor-dependent and independent pathways. In investigating the mechanism by which pathogenic anti-DNA antibodies modulate gene expression in MC, we found that the pathogenic anti-DNA antibody 1A3F bound to high mobility group binding protein 1 (HMGB1), an endogenous ligand for TLR2/4 and RAGE (receptor for advanced glycation end products). Interestingly, HMGB1 treatment of MC induced a similar pattern of genes as stimulation with 1A3F. Furthermore, HMGB1 and 1A3F exhibited a synergistic proinflammatory effect in the kidney, where increased expression of HMGB1 was found in lupus patients but not in patients with other types of renal disease. TLR2/Fc and RAGE/Fc inhibited the proinflammatory effects of 1A3F on MC. Finally, we found enhanced susceptibility of lupus prone MRL-lpr/lpr (MRL/lpr) as compared to normal BALB/c derived MC to pathogenic anti-DNA antibody and LPS stimulation (in particular enhanced chemokine synthesis), in addition to significantly increased expression of TLR4. Our results suggest that gene upregulation in MC induced by nephritogenic anti-DNA antibodies is TLR2/4 and RAGE-dependent. Finally, HMGB1 may act as a proinflammatory mediator in antibody-induced kidney damage in systemic lupus erythematosus (SLE).
Collapse
Affiliation(s)
- Xiaoping Qing
- The Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Milena Pitashny
- The Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David B. Thomas
- The Department of Pathology, Montefiore Medical Center, Bronx, NY, USA
| | | | - Mark P. Hogarth
- Austin Research Institute, Austin and Repatriation Medical Center, Heidelberg, Victoria, Australia
| | - Chaim Putterman
- The Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
49
|
Bianchi R, Giambanco I, Donato R. S100B/RAGE-dependent activation of microglia via NF-kappaB and AP-1 Co-regulation of COX-2 expression by S100B, IL-1beta and TNF-alpha. Neurobiol Aging 2008; 31:665-77. [PMID: 18599158 DOI: 10.1016/j.neurobiolaging.2008.05.017] [Citation(s) in RCA: 202] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Revised: 05/09/2008] [Accepted: 05/18/2008] [Indexed: 12/16/2022]
Abstract
Extracellular S100B is known to affect astrocytic, neuronal and microglial activities, with different effects depending on its concentration. Whereas at relatively low concentrations S100B exerts trophic effects on neurons and astrocytes, at relatively high concentrations the protein causes neuronal apoptosis and activates astrocytes and microglia, thus potentially representing an endogenous factor implicated in neuroinflammation. We have reported that RAGE ligation by S100B in BV-2 microglia results in the upregulation of expression of the pro-inflammatory cyclo-oxygenase 2 (COX-2) via parallel Ras-Cdc42-Rac1-dependent activation of c-Jun NH(2) terminal protein kinase (JNK) and Ras-Rac1-dependent stimulation of NF-kappaB transcriptional activity. We show here that: (1) S100B also stimulates AP-1 transcriptional activity in microglia via RAGE-dependent activation of JNK; (2) S100B upregulates IL-1beta and TNF-alpha expression in microglia via RAGE engagement; and (3) S100B/RAGE-induced upregulation of COX-2, IL-1beta and TNF-alpha expression requires the concurrent activation of NF-kappaB and AP-1. We also show that S100B synergizes with IL-1beta and TNF-alpha to upregulate on COX-2 expression in microglia. Given the crucial roles of COX-2, IL-1beta and TNF-alpha in the inflammatory response, we propose that, by engaging RAGE, S100B might play an important role in microglia activation in the course of brain damage.
Collapse
Affiliation(s)
- Roberta Bianchi
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, C.P. 81 Succ. 3, 06122 Perugia, Italy
| | | | | |
Collapse
|
50
|
Walker DG, Dalsing-Hernandez JE, Lue LF. Human postmortem brain-derived cerebrovascular smooth muscle cells express all genes of the classical complement pathway: a potential mechanism for vascular damage in cerebral amyloid angiopathy and Alzheimer's disease. Microvasc Res 2007; 75:411-9. [PMID: 18048067 DOI: 10.1016/j.mvr.2007.10.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2006] [Revised: 09/27/2007] [Accepted: 10/12/2007] [Indexed: 10/22/2022]
Abstract
Deposition of amyloid around blood vessels, known as cerebral amyloid angiopathy (CAA), is a major pathological feature found in the majority of Alzheimer's disease (AD) cases, and activated complement fragments have been detected on CAA deposits in AD brains. In this study, we demonstrate for the first time that human cerebrovascular smooth muscle cells (HCSMC) isolated from cortical vessels derived from postmortem brains can express mRNAs for complement genes C1qB, C1r, C1s, C2, C3, C4, C5, C6, C7, C8 and C9, the components of the classical complement pathway. Secretion of the corresponding complement proteins for these genes was also demonstrated, except for C1q and C5. Of particular significance was the observation that treatment of HCSMC with aggregated amyloid beta (Abeta) 1-42 increased expression of complement C3 mRNA and increased release of C3 protein. Abeta treatment of HCSMC also increased expression of C6 mRNA. Interferon-gamma induced expression and release of complement C1r, C1s, C2 and C4. As HCSMC are closely associated with Abeta deposits in vessels in the brain, their production of complement proteins could amplify the proinflammatory effects of amyloid in the perivascular environment, further compromising brain vascular integrity.
Collapse
MESH Headings
- Aged, 80 and over
- Alzheimer Disease/metabolism
- Alzheimer Disease/pathology
- Amyloid beta-Peptides/pharmacology
- Brain/blood supply
- Cadaver
- Cells, Cultured
- Cerebral Amyloid Angiopathy/metabolism
- Cerebral Amyloid Angiopathy/pathology
- Complement System Proteins/genetics
- Complement System Proteins/metabolism
- Culture Media, Conditioned/chemistry
- Culture Media, Conditioned/metabolism
- Gene Expression
- Humans
- Interferon-gamma/pharmacology
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Postmortem Changes
- RNA, Messenger/metabolism
Collapse
Affiliation(s)
- Douglas G Walker
- Laboratory of Neuroinflammation, Sun Health Research Institute, Sun City, Arizona, USA.
| | | | | |
Collapse
|