151
|
Su HY, Waldron RT, Gong R, Ramanujan VK, Pandol SJ, Lugea A. The Unfolded Protein Response Plays a Predominant Homeostatic Role in Response to Mitochondrial Stress in Pancreatic Stellate Cells. PLoS One 2016; 11:e0148999. [PMID: 26849807 PMCID: PMC4743835 DOI: 10.1371/journal.pone.0148999] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 01/26/2016] [Indexed: 02/07/2023] Open
Abstract
Activated pancreatic stellate cells (PaSC) are key participants in the stroma of pancreatic cancer, secreting extracellular matrix proteins and inflammatory mediators. Tumors are poorly vascularized, creating metabolic stress conditions in cancer and stromal cells that necessitate adaptive homeostatic cellular programs. Activation of autophagy and the endoplasmic reticulum unfolded protein response (UPR) have been described in hepatic stellate cells, but the role of these processes in PaSC responses to metabolic stress is unknown. We reported that the PI3K/mTOR pathway, which AMPK can regulate through multiple inputs, modulates PaSC activation and fibrogenic potential. Here, using primary and immortalized mouse PaSC, we assess the relative contributions of AMPK/mTOR signaling, autophagy and the UPR to cell fate responses during metabolic stress induced by mitochondrial dysfunction. The mitochondrial uncoupler rottlerin at low doses (0.5-2.5 μM) was added to cells cultured in 10% FBS complete media. Mitochondria rapidly depolarized, followed by altered mitochondrial dynamics and decreased cellular ATP levels. This mitochondrial dysfunction elicited rapid, sustained AMPK activation, mTOR pathway inhibition, and blockade of autophagic flux. Rottlerin treatment also induced rapid, sustained PERK/CHOP UPR signaling. Subsequently, high doses (>5 μM) induced loss of cell viability and cell death. Interestingly, AMPK knock-down using siRNA did not prevent rottlerin-induced mTOR inhibition, autophagy, or CHOP upregulation, suggesting that AMPK is dispensable for these responses. Moreover, CHOP genetic deletion, but not AMPK knock-down, prevented rottlerin-induced apoptosis and supported cell survival, suggesting that UPR signaling is a major modulator of cell fate in PaSC during metabolic stress. Further, short-term rottlerin treatment reduced both PaSC fibrogenic potential and IL-6 mRNA expression. In contrast, expression levels of the angiogenic factors HGF and VEGFα were unaffected, and the immune modulator IL-4 was markedly upregulated. These data imply that metabolic stress-induced PaSC reprogramming differentially modulates neighboring cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Hsin-Yuan Su
- Pancreatic Research Group, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Richard T. Waldron
- Pancreatic Research Group, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Department of Medicine, David Geffen School of Medicine, UCLA/VA Greater Los Angeles Health Sciences Center, Los Angeles, California, United States of America
| | - Raymond Gong
- Pancreatic Research Group, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - V. Krishnan Ramanujan
- Department of Medicine, David Geffen School of Medicine, UCLA/VA Greater Los Angeles Health Sciences Center, Los Angeles, California, United States of America
- Metabolic Photonics Laboratory, Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical center, Los Angeles, California, United States of America
| | - Stephen J. Pandol
- Pancreatic Research Group, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Department of Medicine, David Geffen School of Medicine, UCLA/VA Greater Los Angeles Health Sciences Center, Los Angeles, California, United States of America
| | - Aurelia Lugea
- Pancreatic Research Group, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Department of Medicine, David Geffen School of Medicine, UCLA/VA Greater Los Angeles Health Sciences Center, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
152
|
Timberlake MA, Dwivedi Y. Altered Expression of Endoplasmic Reticulum Stress Associated Genes in Hippocampus of Learned Helpless Rats: Relevance to Depression Pathophysiology. Front Pharmacol 2016; 6:319. [PMID: 26793110 PMCID: PMC4709448 DOI: 10.3389/fphar.2015.00319] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 12/22/2015] [Indexed: 01/30/2023] Open
Abstract
The unfolded protein response (UPR) is an evolutionarily conserved defensive mechanism that is used by cells to correct misfolded proteins that accumulate in the endoplasmic reticulum. These proteins are misfolded as a result of physical stress on a cell and initiate a host of downstream effects that govern processes ranging from inflammation to apoptosis. To examine whether UPR system plays a role in depression, we examined the expression of genes that are part of the three different pathways for UPR activation, namely GRP78, GRP94, ATF6, XBP-1, ATF4, and CHOP using an animal model system that distinguishes vulnerability (learned helpless, LH) from resistance (non-learned helpless, NLH) to develop depression. Rats were exposed to inescapable shock on days 1 and 7 and were tested for escape latency on day 14. Rats not given shock but tested for escape latency were used as tested control (TC). Plasma corticosterone (CORT) levels were measured. Expression levels of various UPR associated genes were determined in hippocampus using qPCR. We found that the CORT level was higher in LH rats compared with TC and NLH rats. Expression of GRP78, GRP94, ATF6, and XBP-1 were significantly upregulated in LH rats compared with TC or NLH rats, whereas NLH rats did not show such changes. Expression levels of ATF4 and CHOP showed trends toward upregulation but were not significantly altered in LH or NLH group. Our data show strong evidence of altered UPR system in depressed rats, which could be associated with development of depressive behavior.
Collapse
Affiliation(s)
- Matthew A Timberlake
- Department of Psychiatry and Behavioral Neurobiology, The University of Alabama at Birmingham School of Medicine, Birmingham AL, USA
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, The University of Alabama at Birmingham School of Medicine, Birmingham AL, USA
| |
Collapse
|
153
|
The crossroads of autoimmunity and immunodeficiency: Lessons from polygenic traits and monogenic defects. J Allergy Clin Immunol 2016; 137:3-17. [DOI: 10.1016/j.jaci.2015.11.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/16/2015] [Accepted: 11/16/2015] [Indexed: 01/16/2023]
|
154
|
Welsby I, Goriely S. Regulation of Interleukin-23 Expression in Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 941:167-189. [DOI: 10.1007/978-94-024-0921-5_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
155
|
Abstract
Possession of the human leukocyte antigen (HLA) class I molecule B27 is strongly associated with ankylosing spondylitis (AS), but the pathogenic role of HLA-B27 is unknown. Two broad theories most likely explain the role of HLA-B27 in AS pathogenesis. The first is based on the natural immunological function of HLA-B27 of presenting antigenic peptides to cytotoxic T cells. Thus, HLA-B27-restricted immune responses to self-antigens, or arthritogenic peptides, might drive immunopathology. B27 can also "behave badly," misfolding during assembly and leading to endoplasmic reticulum stress and autophagy responses. β2m-free B27 heavy chain structures including homodimers (B272) can also be expressed at the cell surface following endosomal recycling of cell surface heterotrimers. Cell surface free heavy chains and B272 bind to innate immune receptors on T, NK, and myeloid cells with proinflammatory effects. This review describes the natural function of HLA-B27, its disease associations, and the current theories as to its pathogenic role.
Collapse
Affiliation(s)
- Paul Bowness
- Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Science (NDORMS), Botnar Research Center, University of Oxford, Headington, Oxford OX3 9DL, United Kingdom;
| |
Collapse
|
156
|
Elder MJ, Webster SJ, Williams DL, Gaston JSH, Goodall JC. TSLP production by dendritic cells is modulated by IL-1β and components of the endoplasmic reticulum stress response. Eur J Immunol 2015; 46:455-63. [PMID: 26573878 PMCID: PMC4783504 DOI: 10.1002/eji.201545537] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 10/02/2015] [Accepted: 11/09/2015] [Indexed: 12/22/2022]
Abstract
Thymic stromal lymphopoietin (TSLP) produced by epithelial cells acts on dendritic cells (DCs) to drive differentiation of TH2‐cells, and is therefore important in allergic disease pathogenesis. However, DCs themselves make significant amounts of TSLP in response to microbial products, but little is known about the key downstream signals that induce and modulate this TSLP secretion from human DCs. We show that human monocyte derived DC (mDC) secretion of TSLP in response to Candida albicans and β‐glucans requires dectin‐1, Syk, NF‐κB, and p38 MAPK signaling. In addition, TSLP production by mDCs is greatly enhanced by IL‐1β, but not TNF‐α, in contrast to epithelial cells. Furthermore, TSLP secretion is significantly increased by signals emanating from the endoplasmic reticulum (ER) stress response, specifically the unfolded protein response sensors, inositol‐requiring transmembrane kinase/endonuclease 1 and protein kinase R‐like ER kinase, which are activated by dectin‐1 stimulation. Thus, TSLP production by mDCs requires the integration of signals from dectin‐1, the IL‐1 receptor, and ER stress signaling pathways. Autocrine TSLP production is likely to play a role in mDC‐controlled immune responses at sites removed from epithelial cell production of the cytokine, such as lymphoid tissue.
Collapse
Affiliation(s)
- Matthew J Elder
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, UK
| | - Steven J Webster
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, UK
| | - David L Williams
- Department of Surgery, Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - J S Hill Gaston
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, UK
| | - Jane C Goodall
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Cambridge, UK
| |
Collapse
|
157
|
Smith JA. The role of the unfolded protein response in axial spondyloarthritis. Clin Rheumatol 2015; 35:1425-31. [DOI: 10.1007/s10067-015-3117-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 11/08/2015] [Indexed: 11/28/2022]
|
158
|
Rodvold JJ, Mahadevan NR, Zanetti M. Immune modulation by ER stress and inflammation in the tumor microenvironment. Cancer Lett 2015; 380:227-36. [PMID: 26525580 DOI: 10.1016/j.canlet.2015.09.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 09/16/2015] [Accepted: 09/19/2015] [Indexed: 12/18/2022]
Abstract
It is now increasingly evident that the immune system represents a barrier to tumor emergence, growth, and recurrence. Although this idea was originally proposed almost 50 years ago as the "immune surveillance hypothesis", it is commonly recognized that, with few rare exceptions, tumor cells always prevail. Thus, one of the central unsolved paradoxes of tumor immunology is how a tumor escapes immune control, which is reflected in the lack of effective autochthonous or vaccine-induced anti-tumor T cell responses. In this review, we discuss the role of the endoplasmic reticulum (ER) stress response/unfolded protein response (UPR) in the immunomodulation of myeloid cells and T cells. Specifically, we will discuss how the tumor cell UPR polarizes myeloid cells in a cell-extrinsic manner, and how in turn, thus polarized myeloid cells negatively affect T cell activation and clonal expansion.
Collapse
Affiliation(s)
- Jeffrey J Rodvold
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093-0815
| | - Navin R Mahadevan
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093-0815
| | - Maurizio Zanetti
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093-0815.
| |
Collapse
|
159
|
Botanical Drugs as an Emerging Strategy in Inflammatory Bowel Disease: A Review. Mediators Inflamm 2015; 2015:179616. [PMID: 26576073 PMCID: PMC4630406 DOI: 10.1155/2015/179616] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/14/2015] [Accepted: 09/21/2015] [Indexed: 01/08/2023] Open
Abstract
Crohn's disease and ulcerative colitis are the two most common categories of inflammatory bowel disease (IBD), which are characterized by chronic inflammation of the intestine that comprises the patients' life quality and requires sustained pharmacological and surgical treatments. Since their aetiology is not completely understood, nonfully efficient drugs have been developed and those that show effectiveness are not devoid of quite important adverse effects that impair their long-term use. Therefore, many patients try with some botanical drugs, which are safe and efficient after many years of use. However, it is necessary to properly evaluate these therapies to consider a new strategy for human IBD. In this report we have reviewed the main botanical drugs that have been assessed in clinical trials in human IBD and the mechanisms and the active compounds proposed for their beneficial effects.
Collapse
|
160
|
Cancer Microenvironment and Endoplasmic Reticulum Stress Response. Mediators Inflamm 2015; 2015:417281. [PMID: 26491226 PMCID: PMC4600498 DOI: 10.1155/2015/417281] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/21/2015] [Accepted: 04/24/2015] [Indexed: 12/17/2022] Open
Abstract
Different stressful conditions such as hypoxia, nutrient deprivation, pH changes, or reduced vascularization, potentially able to act as growth-limiting factors for tumor cells, activate the unfolded protein response (UPR). UPR is therefore involved in tumor growth and adaptation to severe environments and is generally cytoprotective in cancer. The present review describes the molecular mechanisms underlying UPR and able to promote survival and proliferation in cancer. The critical role of UPR activation in tumor growth promotion is discussed in detail for a few paradigmatic tumors such as prostate cancer and melanoma.
Collapse
|
161
|
Lebeaupin C, Proics E, de Bieville CHD, Rousseau D, Bonnafous S, Patouraux S, Adam G, Lavallard VJ, Rovere C, Le Thuc O, Saint-Paul MC, Anty R, Schneck AS, Iannelli A, Gugenheim J, Tran A, Gual P, Bailly-Maitre B. ER stress induces NLRP3 inflammasome activation and hepatocyte death. Cell Death Dis 2015; 6:e1879. [PMID: 26355342 PMCID: PMC4650444 DOI: 10.1038/cddis.2015.248] [Citation(s) in RCA: 316] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 07/17/2015] [Accepted: 07/21/2015] [Indexed: 01/18/2023]
Abstract
The incidence of chronic liver disease is constantly increasing, owing to the obesity epidemic. However, the causes and mechanisms of inflammation-mediated liver damage remain poorly understood. Endoplasmic reticulum (ER) stress is an initiator of cell death and inflammatory mechanisms. Although obesity induces ER stress, the interplay between hepatic ER stress, NLRP3 inflammasome activation and hepatocyte death signaling has not yet been explored during the etiology of chronic liver diseases. Steatosis is a common disorder affecting obese patients; moreover, 25% of these patients develop steatohepatitis with an inherent risk for progression to hepatocarcinoma. Increased plasma LPS levels have been detected in the serum of patients with steatohepatitis. We hypothesized that, as a consequence of increased plasma LPS, ER stress could be induced and lead to NLRP3 inflammasome activation and hepatocyte death associated with steatohepatitis progression. In livers from obese mice, administration of LPS or tunicamycin results in IRE1α and PERK activation, leading to the overexpression of CHOP. This, in turn, activates the NLRP3 inflammasome, subsequently initiating hepatocyte pyroptosis (caspase-1, -11, interleukin-1β secretion) and apoptosis (caspase-3, BH3-only proteins). In contrast, the LPS challenge is blocked by the ER stress inhibitor TUDCA, resulting in: CHOP downregulation, reduced caspase-1, caspase-11, caspase-3 activities, lowered interleukin-1β secretion and rescue from cell death. The central role of CHOP in mediating the activation of proinflammatory caspases and cell death was characterized by performing knockdown experiments in primary mouse hepatocytes. Finally, the analysis of human steatohepatitis liver biopsies showed a correlation between the upregulation of inflammasome and ER stress markers, as well as liver injury. We demonstrate here that ER stress leads to hepatic NLRP3 inflammasome pyroptotic death, thus contributing as a novel mechanism of inflammation-mediated liver injury in chronic liver diseases. Inhibition of ER-dependent inflammasome activation and cell death pathways may represent a potential therapeutic approach in chronic liver diseases.
Collapse
Affiliation(s)
- C Lebeaupin
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| | - E Proics
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| | - C H D de Bieville
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| | - D Rousseau
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| | - S Bonnafous
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Digestif, Nice, France
| | - S Patouraux
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Biologie, Nice, France
| | - G Adam
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| | - V J Lavallard
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| | - C Rovere
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, UMR7275, Valbonne, France
| | - O Le Thuc
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, UMR7275, Valbonne, France
| | - M C Saint-Paul
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Digestif, Nice, France
| | - R Anty
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Digestif, Nice, France
| | - A S Schneck
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Digestif, Nice, France
| | - A Iannelli
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Digestif, Nice, France
| | - J Gugenheim
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Digestif, Nice, France
| | - A Tran
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Digestif, Nice, France
| | - P Gual
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| | - B Bailly-Maitre
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| |
Collapse
|
162
|
Condamine T, Mastio J, Gabrilovich DI. Transcriptional regulation of myeloid-derived suppressor cells. J Leukoc Biol 2015; 98:913-22. [PMID: 26337512 DOI: 10.1189/jlb.4ri0515-204r] [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: 05/12/2015] [Accepted: 08/21/2015] [Indexed: 12/14/2022] Open
Abstract
Myeloid-derived suppressor cells are a heterogeneous group of pathologically activated immature cells that play a major role in the negative regulation of the immune response in cancer, autoimmunity, many chronic infections, and inflammatory conditions, as well as in the regulation of tumor angiogenesis, tumor cell invasion, and metastases. Accumulation of myeloid-derived suppressor cells is governed by a network of transcriptional regulators that could be combined into 2 partially overlapping groups: factors promoting myelopoiesis and preventing differentiation of mature myeloid cells and factors promoting pathologic activation of myeloid-derived suppressor cells. In this review, we discuss the specific nature of these factors and their impact on myeloid-derived suppressor cell development.
Collapse
Affiliation(s)
| | - Jérôme Mastio
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
163
|
Wright PB, McEntegart A, McCarey D, McInnes IB, Siebert S, Milling SWF. Ankylosing spondylitis patients display altered dendritic cell and T cell populations that implicate pathogenic roles for the IL-23 cytokine axis and intestinal inflammation. Rheumatology (Oxford) 2015; 55:120-32. [PMID: 26320138 PMCID: PMC4676904 DOI: 10.1093/rheumatology/kev245] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Indexed: 01/07/2023] Open
Abstract
Objective. AS is a systemic inflammatory disease of the SpA family. Polymorphisms at loci including HLA-B27, IL-23R and ERAP-1 directly implicate immune mechanisms in AS pathogenesis. Previously, in an SpA model, we identified HLA-B27–mediated effects on dendritic cells that promoted disease-associated Th17 cells. Here we extend these studies to AS patients using deep immunophenotyping of candidate pathogenic cell populations. The aim of our study was to functionally characterize the immune populations mediating AS pathology. Methods. Using 11-parameter flow cytometry, we characterized the phenotype and functions of lymphocyte and myeloid cells from peripheral blood, and the synovial phenotype of AS patients and age-matched healthy controls. Results. Significantly fewer circulating CD1c-expressing dendritic cells were observed in AS patients, offset by an increase in CD14− CD16+ mononuclear cells. Ex vivo functional analysis revealed that this latter population induced CCR6 expression and promoted secretion of IL-1β and IL-6 when co-cultured with naive CD4+ T cells. Additionally, systemic inflammation in AS patients significantly correlated with increased proportions of activated CCR9+ CD4+ T cells. Conclusion. CD14− CD16+ mononuclear cells may contribute to AS by promoting Th17 responses, and antigen-presenting cells of mucosal origin are likely to contribute to systemic inflammation in AS.
Collapse
Affiliation(s)
- Pamela B Wright
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow and
| | - Anne McEntegart
- Department of Rheumatology, Queen Elizabeth Building, Glasgow Royal Infirmary, Glasgow, UK
| | - David McCarey
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow and
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow and
| | - Stefan Siebert
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow and
| | - Simon W F Milling
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow and
| |
Collapse
|
164
|
Abstract
PURPOSE OF REVIEW Interleukin (IL)-23 and the related cytokine IL-17 play vital roles in immune-mediated inflammatory pathology. In the years since its discovery, IL-23 has been implicated as a central pathogenic factor in multiple rheumatic conditions and has been shown to act via a wide range of immune cells including type 17 T-helper (Th17) cells and innate-like immune cells. We review here the pivotal role of these cytokines and IL-23-responsive cells in both the bona fide autoimmune rheumatic diseases rheumatoid arthritis and systemic lupus erythematosus, as well as the spondyloarthropathies which more closely resemble the auto-inflammatory conditions. RECENT FINDINGS IL-23 and related cytokines have been found to be up-regulated in rheumatoid arthritis, systemic lupus erythematosus and spondyloarthropathy, and preclinical models suggest that they play important pathological roles in these conditions. SUMMARY It is anticipated that agents which target the IL-23 pathway will have profound roles in modifying the natural history of these diseases and in preventing the structural damage which occurs secondary to such chronic inflammation. This is especially relevant in the case of spondyloarthropathy in which case prevention of the novel bone formation is a particular challenge. It is also potentially pertinent for patients with rheumatoid arthritis, particularly those who do not respond to other biological therapies.
Collapse
|
165
|
Abstract
A recently paper published in Cell reports that dendritic cells (DCs) are dysfunctional in the tumor environment. Tumor impairs DC function through induction of endoplasmic reticulum stress response and subsequent disruption of lipid metabolic homeostasis.
Collapse
|
166
|
Zinc regulates expression of IL-23 p19 mRNA via activation of eIF2α/ATF4 axis in HAPI cells. Biometals 2015; 28:891-902. [PMID: 26174742 DOI: 10.1007/s10534-015-9874-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 07/03/2015] [Indexed: 12/22/2022]
Abstract
Zinc (Zn(2+)) is considered to be one of the factors aggravating brain damage after cerebral ischemia. Since Zn(2+) activates microglia, immune cells in the brain, this metal is proposed to modulate neuroinflammatory responses in the post-ischemic brain. Interleukin (IL)-23 is a heterodimeric cytokine composed of the p19 subunit unique to IL-23 and the p40 subunit common to IL-12. IL-23 has been shown to play a critical role in the progression of ischemic brain injury. However, whether Zn(2+) participates in the expression of IL-23 in microglia remains unknown. In this study, we examined the effect of Zn(2+) on IL-23 p19 mRNA expression using rat immortalized microglia HAPI cells. Exposure to Zn(2+) dose- and time-dependently induced the expression of IL-23 p19 mRNA in HAPI cells. Inhibitors of MAPK and NF-κB pathways failed to suppress this induction. Interestingly, we found that Zn(2+) stimulated the phosphorylation of eIF2α and promoted the nuclear accumulation of activating transcription factor 4 (ATF4). Treatment with salubrinal, an eIF2α dephosphorylation inhibitor, enhanced Zn(2+)-induced ATF4 accumulation and IL-23 p19 mRNA expression. In addition, reporter assay using the IL-23 p19 promoter region revealed that ATF4 directly transactivated IL-23 p19 promoter and that dominant-negative ATF4 suppressed Zn(2+)-induced activation of IL-23 p19 promoter. Taken together, these findings suggest that Zn(2+) up-regulates expression of the IL-23 p19 gene via the eIF2α/ATF4 axis in HAPI cells.
Collapse
|
167
|
Abstract
The term axial spondyloarthritis covers both non-radiographic disease and radiographic disease (also known as ankylosing spondylitis). Some studies have been performed to investigate the prevalence of axial spondyloarthritis, although most are limited to patients with radiographic disease. A strong genetic association has been shown between axial spondyloarthritis and human leukocyte antigen-B27 (HLA-B27), but the pathogenetic role of HLA-B27 has not yet been clarified. Tumour necrosis factor (TNF), IL-17, IL-23 and downstream pathways also seem to be important - based on the good results of therapies directed against these molecules - but their exact role in the inflammatory process is also not yet clear. Elucidating the interaction between osteoproliferation and inflammation will be crucial for the prevention of long-term structural damage of the bone. The development of new criteria for classification, diagnosis and screening of patients with axial spondyloarthritis will enable earlier intervention for this chronic inflammatory disease. MRI has become an important tool for the early detection of axial spondyloarthritis. NSAIDs and TNF blockers are effective therapies, including in the early non-radiographic stage. Therapeutic blockade of IL-17 or IL-23 seems to be a promising new treatment option. Tools for measuring quality of life in axial spondyloarthritis have become relevant to assess the impact that the disease has on patients. These diagnostic and therapeutic advances will continue to change the management of axial spondyloarthritis, and new insights into the disease pathogenesis will hopefully accelerate this process. For an illustrated summary of this Primer, visit: http://go.nature.com/51b1af.
Collapse
Affiliation(s)
- Joachim Sieper
- Rheumatology, Charité, Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany
| | | | - Maxime Dougados
- Faculty of Medicine, Paris Descartes University, Department of Rheumatology, Hôpital Cochin, Assistance Publique, Hôpitaux de Paris, INSERM (U1153), Clinical Epidemiology and Biostatistics, PRES Sorbonne Paris-Cité, Paris, France
| | - Dominique Baeten
- Clinical Immunology and Rheumatology and Amsterdam Rheumatology and Immunology Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
168
|
Datta S, Barrera N, Pavicic PG, Zhao C, Freeman M, Min B, Hamilton T. cEBP Homologous Protein Expression in Macrophages Regulates the Magnitude and Duration of IL-6 Expression and Dextran Sodium Sulfate Colitis. J Interferon Cytokine Res 2015; 35:785-94. [PMID: 26134251 DOI: 10.1089/jir.2014.0204] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cellular stress enhances inflammatory cytokine gene expression by inducing cEBP homologous protein (CHOP). Engaging cell stress via thapsigargin induced CHOP and selectively prolonged lipopolysaccharide-stimulated interleukin-6 (IL-6) expression in bone marrow-derived macrophages from wild-type (WT) but not CHOP knockout (KO) mice. To determine the impact of this mechanism in vivo we employed dextran sodium sulfate (DSS)-induced colitis in irradiated mice reconstituted with bone marrow from WT or CHOP KO mice. WT recipients of CHOP KO bone marrow exhibited more rapid recovery from disease than did mice reconstituted with WT bone marrow as reflected in increased survival, reduced clinical scores, and colonic histopathology. No differences in mesenteric lymph node cell populations were observed between mice with WT or CHOP KO bone marrow during colitis. CD11b(+) macrophages infiltrating the lamina propria were, however, reduced in DSS-treated mice reconstituted with CHOP KO bone marrow. CHOP expression was observed within the infiltrating inflammatory CD11b(+) macrophages. Furthermore, IL-6 expression within the inflamed colon was significantly lower in mice with CHOP-deficient bone marrow. Our findings indicate that CHOP expression in myeloid cells plays an important role in determining the magnitude and duration of inflammatory response in vivo by modulating expression of proinflammatory cytokines such as IL-6 in infiltrating macrophages.
Collapse
Affiliation(s)
- Shyamasree Datta
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation , Cleveland, Ohio
| | - Natilibeth Barrera
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation , Cleveland, Ohio
| | - Paul G Pavicic
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation , Cleveland, Ohio
| | - Chenyang Zhao
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation , Cleveland, Ohio
| | - Michael Freeman
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation , Cleveland, Ohio
| | - Booki Min
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation , Cleveland, Ohio
| | - Thomas Hamilton
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation , Cleveland, Ohio
| |
Collapse
|
169
|
Lugea A, Waldron RT, Pandol SJ. Pancreatic adaptive responses in alcohol abuse: Role of the unfolded protein response. Pancreatology 2015; 15:S1-5. [PMID: 25736240 PMCID: PMC4515411 DOI: 10.1016/j.pan.2015.01.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 01/22/2015] [Accepted: 01/27/2015] [Indexed: 12/11/2022]
Abstract
The majority of those who drink excessive amounts of alcohol do not develop pancreatic disease. One overarching hypothesis is that alcohol abuse requires additional risk factors, either environmental or genetic, for disease to occur. However, another reason be a result of alcohol-induced activation of adaptive systems that protect the pancreas from the toxic effects of alcohol. We show that mechanisms within the unfolded protein response (UPR) of the endoplasmic reticulum (ER) that can lead to protection of the pancreas from pancreatic diseases with alcohol abuse. The remarkable ability of the pancreas to adapt its machinery to alcohol abuse using UPR systems and continue functioning is the likely reason that pancreatitis from alcohol abuse does not occur in the majority of heavy drinkers. These findings indicate that methods to enhance the protective responses of the UPR can provide opportunities for prevention and treatment of pancreatic diseases.
Collapse
Affiliation(s)
- Aurelia Lugea
- Cedars-Sinai Medical Center and VA Greater Los Angeles Health Care System, Los Angeles, CA, USA
| | - Richard T Waldron
- Cedars-Sinai Medical Center and VA Greater Los Angeles Health Care System, Los Angeles, CA, USA
| | - Stephen J Pandol
- Cedars-Sinai Medical Center and VA Greater Los Angeles Health Care System, Los Angeles, CA, USA.
| |
Collapse
|
170
|
Watkin LB, Jessen B, Wiszniewski W, Vece T, Jan M, Sha Y, Thamsen M, Santos-Cortez RLP, Lee K, Gambin T, Forbes L, Law CS, Stray-Petersen A, Cheng MH, Mace EM, Anderson MS, Liu D, Tang LF, Nicholas SK, Nahmod K, Makedonas G, Canter D, Kwok PY, Hicks J, Jones KD, Penney S, Jhangiani SN, Rosenblum MD, Dell SD, Waterfield MR, Papa FR, Muzny DM, Zaitlen N, Leal SM, Gonzaga-Jauregui C, Baylor-Hopkins Center for Mendelian Genomics, Boerwinkle E, Eissa NT, Gibbs RA, Lupski JR, Orange JS, Shum AK. COPA mutations impair ER-Golgi transport and cause hereditary autoimmune-mediated lung disease and arthritis. Nat Genet 2015; 47:654-60. [PMID: 25894502 PMCID: PMC4513663 DOI: 10.1038/ng.3279] [Citation(s) in RCA: 272] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 03/19/2015] [Indexed: 12/12/2022]
Abstract
Unbiased genetic studies have uncovered surprising molecular mechanisms in human cellular immunity and autoimmunity. We performed whole-exome sequencing and targeted sequencing in five families with an apparent mendelian syndrome of autoimmunity characterized by high-titer autoantibodies, inflammatory arthritis and interstitial lung disease. We identified four unique deleterious variants in the COPA gene (encoding coatomer subunit α) affecting the same functional domain. Hypothesizing that mutant COPA leads to defective intracellular transport via coat protein complex I (COPI), we show that COPA variants impair binding to proteins targeted for retrograde Golgi-to-ER transport. Additionally, expression of mutant COPA results in ER stress and the upregulation of cytokines priming for a T helper type 17 (TH17) response. Patient-derived CD4(+) T cells also demonstrate significant skewing toward a TH17 phenotype that is implicated in autoimmunity. Our findings uncover an unexpected molecular link between a vesicular transport protein and a syndrome of autoimmunity manifested by lung and joint disease.
Collapse
Affiliation(s)
- Levi B. Watkin
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
- Texas Children’s Hospital Center for Human Immuno-Biology, Houston, TX
| | - Birthe Jessen
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Wojciech Wiszniewski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Timothy Vece
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Max Jan
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Youbao Sha
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Maike Thamsen
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | | | - Kwanghyuk Lee
- Center for Statistical Genetics, Baylor College of Medicine, Houston, TX
| | - Tomasz Gambin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Lisa Forbes
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
- Texas Children’s Hospital Center for Human Immuno-Biology, Houston, TX
| | - Christopher S. Law
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Asbjørg Stray-Petersen
- Texas Children’s Hospital Center for Human Immuno-Biology, Houston, TX
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Mickie H. Cheng
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Emily M. Mace
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
- Texas Children’s Hospital Center for Human Immuno-Biology, Houston, TX
| | - Mark S. Anderson
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Dongfang Liu
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
- Texas Children’s Hospital Center for Human Immuno-Biology, Houston, TX
| | - Ling Fung Tang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA
| | - Sarah K. Nicholas
- Texas Children’s Hospital Center for Human Immuno-Biology, Houston, TX
| | - Karen Nahmod
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
- Texas Children’s Hospital Center for Human Immuno-Biology, Houston, TX
| | - George Makedonas
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
- Texas Children’s Hospital Center for Human Immuno-Biology, Houston, TX
| | - Debra Canter
- Texas Children’s Hospital Center for Human Immuno-Biology, Houston, TX
| | - Pui-Yan Kwok
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA
- Department of Dermatology, University of California San Francisco, San Francisco, CA
| | - John Hicks
- Department of Pathology, Texas Children’s Hospital, Houston, TX
| | - Kirk D. Jones
- Department of Pathology, University of California San Francisco, San Francisco, CA
| | - Samantha Penney
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | | | - Michael D. Rosenblum
- Department of Dermatology, University of California San Francisco, San Francisco, CA
| | - Sharon D. Dell
- Division of Respiratory Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Feroz R. Papa
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Donna M. Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Noah Zaitlen
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Suzanne M. Leal
- Center for Statistical Genetics, Baylor College of Medicine, Houston, TX
| | | | | | - Eric Boerwinkle
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
- Human Genetics Center and Institute of Molecular Medicine, University of Texas-Houston Health Science Center, Houston, TX
| | - N. Tony Eissa
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Richard A. Gibbs
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
- Human Genetics Center and Institute of Molecular Medicine, University of Texas-Houston Health Science Center, Houston, TX
| | - James R. Lupski
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
- Texas Children’s Hospital Center for Human Immuno-Biology, Houston, TX
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Jordan S. Orange
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
- Texas Children’s Hospital Center for Human Immuno-Biology, Houston, TX
| | - Anthony K. Shum
- Department of Medicine, University of California San Francisco, San Francisco, CA
| |
Collapse
|
171
|
The evolving paradigm of cell-nonautonomous UPR-based regulation of immunity by cancer cells. Oncogene 2015; 35:269-78. [PMID: 25893303 DOI: 10.1038/onc.2015.108] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 03/02/2015] [Accepted: 03/04/2015] [Indexed: 12/19/2022]
Abstract
The endoplasmic reticulum (ER) stress response/unfolded protein response (UPR) has been thought to influence tumorigenesis mainly through cell-intrinsic, pro-survival effects. In recent years, however, new evidence has emerged showing that the UPR is also the source of cell-extrinsic effects, particularly directed at those immune cells within the tumor microenvironment. Here we will review and discuss this new body of information with focus on the role of cell-extrinsic effects on innate and adaptive immunity, suggesting that the transmission of ER stress from cancer cells to myeloid cells in particular is an expedient used by cancer cells to control the immune microenvironment, which acquires pro-inflammatory as well as immune-suppressive characteristics. These new findings can now be seen in the broader context of similar phenomena described in Caenorhabditis elegans, and an analogy with quorum sensing and 'community effects' in prokaryotes and eukaryotes can be drawn, arguing that a cell-nonautonomous UPR-based regulation of heterologous cells may be phylogenetically conserved. Finally, we will discuss the role of aneuploidy as an inducer of proteotoxic stress and potential initiator of cell-nonautonomous UPR-based regulation. In presenting these new views, we wish to bring attention to the cell-extrinsic regulation of tumor growth, including tumor UPR-based cell-nonautonomous signaling as a mechanism of maintaining tumor heterogeneity and resistance to therapy, and suggest therapeutically targeting such mechanisms within the tumor microenvironment.
Collapse
|
172
|
IL-23 Responsive Innate-Like T Cells in Spondyloarthritis: the Less Frequent They Are, the More Vital They Appear. Curr Rheumatol Rep 2015; 17:30. [DOI: 10.1007/s11926-015-0507-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
173
|
Benham H, Robinson PC, Baillet AC, Rehaume LM, Thomas R. Role of genetics in infection-associated arthritis. Best Pract Res Clin Rheumatol 2015; 29:213-25. [PMID: 26362740 DOI: 10.1016/j.berh.2015.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 02/12/2015] [Accepted: 02/16/2015] [Indexed: 01/17/2023]
Abstract
Genetic discoveries in arthritis and their associated biological pathways spanning the innate and adaptive immune system demonstrate the strong association between susceptibility to arthritis and control of exogenous organisms. The canonical theory of the aetiology of immune-mediated arthritis and other immune-mediated diseases is that the introduction of exogenous antigenic stimuli to a genetically susceptible host sets up the environment for an abnormal immune response manifesting as disease. A disruption in host-microbe homeostasis driven by disease-associated genetic variants could ultimately provide the source of exogenous antigen triggering disease development. We discuss genetic variants impacting the innate and adaptive arms of the immune system and their relationship to microbial control and arthritic disease. We go on to consider the evidence for a relationship between HLA-B27, infection and arthritis, and then emerging evidence for an interaction between microbiota and rheumatoid arthritis.
Collapse
Affiliation(s)
- Helen Benham
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia; The University of Queensland School of Medicine, Translational Research Institute, Brisbane, QLD, Australia
| | - Philip C Robinson
- Centre for Neurogenetics and Statistical Genomics, Queensland Brain Institute, University of Queensland, QLD, Australia
| | - Athan C Baillet
- GREPI AGIM FRE3405 CNRS-EPHE, University Joseph Fourier, Grenoble, France
| | - Linda M Rehaume
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Ranjeny Thomas
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia.
| |
Collapse
|
174
|
Abstract
In eukaryotic cells, protein folding and modification in the endoplasmic reticulum (ER) is highly sensitive to disturbances of homeostasis. The accumulation of unfolded and misfolded proteins in the ER lumen, termed ER stress, activates intracellular signaling pathways to resolve the protein-folding defect. This unfolded protein response (UPR) increases the capacity of ER protein folding, reduces global protein synthesis, and activates ER-associated protein degradation. If ER stress is too severe or chronic, or the UPR is compromised and not able to restore ER protein-folding homeostasis, numerous apoptotic signaling pathways are activated. Preclinical and clinical studies in the past decade indicate that ER stress and the UPR have a significant impact on the pathogenesis of inflammatory bowel disease. Paneth and goblet cells, 2 epithelial cell populations in the gut, rely on a robust ER function for protein folding and secretion. Several immune cells are orchestrated by ER stress and the UPR for differentiation, activation, migration, and survival. In addition, a variety of exogenous and endogenous molecules in the intestinal lumen affect ER function, making ER stress and the UPR relevant cellular signals in intestinal homeostasis. Recent studies demonstrated that unresolved ER stress and/or dysregulated UPR may cause inflammatory bowel disease by inducing epithelial cell death, impairing mucosal barrier function, and activating proinflammatory response in the gut. With our increased understanding of ER stress in inflammatory bowel disease pathogenesis, it is now possible to develop novel therapies to improve ER protein-folding homeostasis and target-specific UPR pathways in cells residing in the intestinal mucosa.
Collapse
|
175
|
Shima K, Klinger M, Schütze S, Kaufhold I, Solbach W, Reiling N, Rupp J. The role of endoplasmic reticulum-related BiP/GRP78 in interferon gamma-induced persistent Chlamydia pneumoniae infection. Cell Microbiol 2015; 17:923-34. [PMID: 25588955 DOI: 10.1111/cmi.12416] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 12/12/2014] [Accepted: 01/01/2015] [Indexed: 12/26/2022]
Abstract
Direct interaction of Chlamydiae with the endoplasmic reticulum (ER) is essential in intracellular productive infection. However, little is known about the interplay between Chlamydiae and the ER under cellular stress conditions that are observed in interferon gamma (IFN-γ) induced chlamydial persistent infection. ER stress responses are centrally regulated by the unfolded protein response (UPR) under the control of the ER chaperone BiP/GRP78 to maintain cellular homeostasis. In this study, we could show that the ER directly contacted with productive and IFN-γ-induced persistent inclusions of Chlamydia pneumoniae (Cpn). BiP/GRP78 induction was observed in the early phase but not in the late phase of IFN-γ-induced persistent infection. Enhanced BiP/GRP78 expression in the early phase of IFN-γ-induced persistent Cpn infection was accompanied by phosphorylation of the eukaryotic initiation factor-2α (eIF2α) and down-regulation of the vesicle-associated membrane protein-associated protein B. Loss of BiP/GRP78 function resulted in enhanced phosphorylation of eIF2α and increased host cell apoptosis. In contrast, enhanced BiP/GRP78 expression in IFN-γ-induced persistent Cpn infection attenuated phosphorylation of eIF2α upon an exogenous ER stress inducer. In conclusion, ER-related BiP/GRP78 plays a key role to restore cells from stress conditions that are observed in the early phase of IFN-γ-induced persistent infection.
Collapse
Affiliation(s)
- Kensuke Shima
- Department of Molecular and Clinical Infectious Diseases, University of Lübeck, Ratzeburger Allee 160, Lübeck, 23538, Germany
| | | | - Stefan Schütze
- Institute of Immunology, University of Kiel, Kiel, Germany
| | - Inga Kaufhold
- Department of Molecular and Clinical Infectious Diseases, University of Lübeck, Ratzeburger Allee 160, Lübeck, 23538, Germany
| | - Werner Solbach
- Institute of Medical Microbiology and Hygiene, University of Lübeck, Lübeck, Germany
| | - Norbert Reiling
- Division of Microbial Interface Biology, Research Center Borstel, Leibniz Center for Medicine and Biosciences, Borstel, Germany
| | - Jan Rupp
- Department of Molecular and Clinical Infectious Diseases, University of Lübeck, Ratzeburger Allee 160, Lübeck, 23538, Germany
| |
Collapse
|
176
|
Kim H, Moon SY, Kim JS, Baek CH, Kim M, Min JY, Lee SK. Activation of AMP-activated protein kinase inhibits ER stress and renal fibrosis. Am J Physiol Renal Physiol 2015; 308:F226-36. [DOI: 10.1152/ajprenal.00495.2014] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
It has been suggested that endoplasmic reticulum (ER) stress facilitates fibrotic remodeling. Therefore, modulation of ER stress may serve as one of the possible therapeutic approaches to renal fibrosis. We examined whether and how activation of AMP-activated protein kinase (AMPK) suppressed ER stress induced by chemical ER stress inducers [tunicamycin (TM) and thapsigargin (TG)] and also nonchemical inducers in tubular HK-2 cells. We further investigated the in vivo effects of AMPK on ER stress and renal fibrosis. Western blot analysis, immunofluorescence, small interfering (si)RNA experiments, and immunohistochemical staining were performed. Metformin (the best known clinical activator of AMPK) suppressed TM- or TG-induced ER stress, as shown by the inhibition of TM- or TG-induced upregulation of glucose-related protein (GRP)78 and phosphorylated eukaryotic initiation factor-2α through induction of heme oxygenase-1. Metformin inhibited TM- or TG-induced epithelial-mesenchymal transitions as well. Compound C (AMPK inhibitor) blocked the effect of metformin, and 5-aminoimidazole-4-carboxamide-1β riboside (another AMPK activator) exerted the same effects as metformin. Transfection with siRNA targeting AMPK blocked the effect of metformin. Consistent with the results of cell culture experiments, metformin reduced renal cortical GRP78 expression and increased heme oxygenase-1 expression in a mouse model of ER stress-induced acute kidney injury by TM. Activation of AMPK also suppressed ER stress by transforming growth factor-β, ANG II, aldosterone, and high glucose. Furthermore, metformin reduced GRP78 expression and renal fibrosis in a mouse model of unilateral ureteral obstruction. In conclusion, AMPK may serve as a promising therapeutic target through reducing ER stress and renal fibrosis.
Collapse
Affiliation(s)
- Hyosang Kim
- Department of Internal Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan, Seoul, Korea
| | - Soo Young Moon
- Department of Internal Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan, Seoul, Korea
| | - Joon-Seok Kim
- Department of Internal Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan, Seoul, Korea
| | - Chung Hee Baek
- Department of Internal Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan, Seoul, Korea
| | - Miyeon Kim
- Department of Internal Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan, Seoul, Korea
| | - Ji Yeon Min
- Department of Internal Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan, Seoul, Korea
| | - Sang Koo Lee
- Department of Internal Medicine, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan, Seoul, Korea
| |
Collapse
|
177
|
|
178
|
Baeten D. Etiology, pathogenesis, and pathophysiology of ankylosing spondylitis. Rheumatology (Oxford) 2015. [DOI: 10.1016/b978-0-323-09138-1.00115-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
179
|
Emerging functions of the unfolded protein response in immunity. Nat Immunol 2014; 15:910-9. [PMID: 25232821 DOI: 10.1038/ni.2991] [Citation(s) in RCA: 188] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 08/18/2014] [Indexed: 12/14/2022]
Abstract
The unfolded protein response (UPR) has traditionally been viewed as an adaptive response triggered by the accumulation of unfolded proteins in the endoplasmic reticulum (ER) and aimed at restoring ER function. The UPR can also be an anticipatory response that is activated well before the disruption of protein homeostasis. UPR signaling intersects at many levels with the innate and adaptive immune responses. In some types of cells of the immune system, such as dendritic cells (DCs) and B cells, particular sensors that detect the UPR seem to be constitutively active in the absence of induction of the traditional UPR gene program and are necessary for antigen presentation and immunoglobulin synthesis. The UPR also influences signaling via Toll-like receptors (TLRs) and activation of the transcription factor NF-κB, and some pathogens subvert the UPR. This Review summarizes these emerging noncanonical functions of the UPR in immunity.
Collapse
|
180
|
Abstract
Immune responses occur in the midst of a variety of cellular stresses that can severely perturb endoplasmic reticulum (ER) function. The unfolded protein response is a three-pronged signaling axis dedicated to preserving ER homeostasis. In this review, we highlight many important and emerging functional roles for ER stress in immunity, focusing on how the bidirectional cross talk between immunological processes and basic cell biology leads to pleiotropic signaling outcomes and enhanced sensitivity to inflammatory stimuli. We also discuss how dysregulated ER stress responses can provoke many diseases, including autoimmunity, firmly positioning the unfolded protein response as a major therapeutic target in human disease.
Collapse
Affiliation(s)
- Sarah E Bettigole
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065; ,
| | | |
Collapse
|
181
|
Luís A, Martins JD, Silva A, Ferreira I, Cruz MT, Neves BM. Oxidative stress-dependent activation of the eIF2α–ATF4 unfolded protein response branch by skin sensitizer 1-fluoro-2,4-dinitrobenzene modulates dendritic-like cell maturation and inflammatory status in a biphasic manner [corrected]. Free Radic Biol Med 2014; 77:217-29. [PMID: 25236743 DOI: 10.1016/j.freeradbiomed.2014.09.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 09/02/2014] [Accepted: 09/03/2014] [Indexed: 01/10/2023]
Abstract
The pathogenesis of allergic contact dermatitis, the most common manifestation of immunotoxicity in humans, is intimately connected to hapten-induced maturation of dendritic cells (DC). The molecular mechanisms driving this maturational program are not completely known; however, initial danger signals such as the generation of reactive oxygen species (ROS) were shown to play a critical role. Recent evidence linking ROS production, endoplasmic reticulum (ER) stress, and the pathogenesis of several inflammatory diseases led us to analyze, in the present work, the ability of the skin sensitizer 1-fluoro-2,4-dinitrobenzene (DNFB) to evoke ER stress in DC-like THP-1 cells and the concomitant consequences to their immunobiology. We found that DNFB triggers a ROS-dependent activation of the PERK-eIFα-ATF4 unfolded protein response (UPR) branch conferring cytoprotection and modulating the maturation/proinflammatory cell status in a biphasic manner. Early DNFB induction of ATF4 positively modulates autophagy-related genes MAP1LC3B and ATG3 and stabilizes the transcription factor Nrf2, causing a strong induction of the HMOX1-detoxifying gene. Moreover, we observed that in a first phase, DNFB-induced ATF4 upregulates IL8 mRNA levels while blocking CD86, IL1B, IL12B, and CXL10 transcription. Later, following ATF4 decay, HMOX1 and IL8 transcription drastically decrease and CD86, IL1B, and Il12B are upregulated. Overall, our results evidence a connection between sensitizer-induced redox imbalance and the establishment of ER stress in DC-like cells and provide new insights into the role of UPR effectors such as ATF4 to the complex DC maturational program.
Collapse
Affiliation(s)
- Andreia Luís
- Department of Chemistry, Mass Spectrometry Centre, QOPNA, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - João Demétrio Martins
- Centre for Neuroscience and Cell Biology, University of Coimbra 3004-517, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra 3004-517, Coimbra, Portugal
| | - Ana Silva
- Centre for Neuroscience and Cell Biology, University of Coimbra 3004-517, Coimbra, Portugal
| | - Isabel Ferreira
- Centre for Neuroscience and Cell Biology, University of Coimbra 3004-517, Coimbra, Portugal
| | - Maria Teresa Cruz
- Centre for Neuroscience and Cell Biology, University of Coimbra 3004-517, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra 3004-517, Coimbra, Portugal
| | - Bruno Miguel Neves
- Department of Chemistry, Mass Spectrometry Centre, QOPNA, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; Centre for Neuroscience and Cell Biology, University of Coimbra 3004-517, Coimbra, Portugal.
| |
Collapse
|
182
|
Subramanian M, Thorp E, Tabas I. Identification of a non-growth factor role for GM-CSF in advanced atherosclerosis: promotion of macrophage apoptosis and plaque necrosis through IL-23 signaling. Circ Res 2014; 116:e13-24. [PMID: 25348165 DOI: 10.1161/circresaha.116.304794] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
RATIONALE Granulocyte macrophage colony-stimulating factor (GM-CSF, Csf2) is a growth factor for myeloid-lineage cells that has been implicated in the pathogenesis of atherosclerosis and other chronic inflammatory diseases. However, the role of GM-CSF in advanced atherosclerotic plaque progression, the process that gives rise to clinically dangerous plaques, is unknown. OBJECTIVE To understand the role of GM-CSF in advanced atherosclerotic plaque progression. METHODS AND RESULTS Ldlr(-/-) mice and Csf2(-/-)Ldlr(-/-) mice were fed a Western-type diet for 12 weeks, and then parameters of advanced plaque progression in the aortic root were quantified. Lesions from the GM-CSF-deficient mice showed a substantial decrease in 2 key hallmarks of advanced atherosclerosis, lesional macrophage apoptosis and plaque necrosis, which indicates that GM-CSF promotes plaque progression. Based on a combination of in vitro and in vivo studies, we show that the mechanism involves GM-CSF-mediated production of interleukin-23, which increases apoptosis susceptibility in macrophages by promoting proteasomal degradation of the cell survival protein Bcl-2 (B-cell lymphoma 2) and by increasing oxidative stress. CONCLUSIONS In low-density lipoprotein-driven atherosclerosis in mice, GM-CSF promotes advanced plaque progression by increasing macrophage apoptosis susceptibility. This action of GM-CSF is mediated by its interleukin-23-inducing activity rather than its role as a growth factor.
Collapse
Affiliation(s)
- Manikandan Subramanian
- From the Departments of Medicine (M.S., I.T.), Pathology and Cell Biology (I.T.), and Physiology and Cellular Biophysics (I.T.), Columbia University, New York, NY; and Department of Pathology, Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (E.T.)
| | - Edward Thorp
- From the Departments of Medicine (M.S., I.T.), Pathology and Cell Biology (I.T.), and Physiology and Cellular Biophysics (I.T.), Columbia University, New York, NY; and Department of Pathology, Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (E.T.)
| | - Ira Tabas
- From the Departments of Medicine (M.S., I.T.), Pathology and Cell Biology (I.T.), and Physiology and Cellular Biophysics (I.T.), Columbia University, New York, NY; and Department of Pathology, Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (E.T.).
| |
Collapse
|
183
|
Kroese FGM, Baeten D, Huizinga TWJ. Autoimmunity: break-through in the diagnosis and treatment of immune-mediated inflammatory diseases. Immunol Lett 2014; 162:150-62. [PMID: 25455603 DOI: 10.1016/j.imlet.2014.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The study of fundamental mechanisms of autoimmunity has been instrumental to clinical progress in the diagnosis and treatment of a range of immune-mediated inflammatory disorders. Dutch immunology has made major contributions to these developments, ranging from fundamental studies on immune cells, antibodies and cytokines to translational and clinical studies with targeted therapies in patients. In this paper we illustrate the progress made in our understanding of autoimmunity and the translational implications for human disease management by focusing on three areas: the autoantibody response in rheumatoid arthritis (RA), T-B cell interactions in Sjögren's syndrome (SS), and cytokine targeting in spondylarthritis (SpA).
Collapse
Affiliation(s)
- Frans G M Kroese
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| | - Dominique Baeten
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Tom W J Huizinga
- Department of Rheumatology, C1-41 Leiden University Medical Center, Albinusdreef 2, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
184
|
Abstract
Systemic autoinflammatory diseases are caused by abnormal activation of the cells that mediate innate immunity. In the past two decades, single-gene defects in different pathways, driving clinically distinct autoinflammatory syndromes, have been identified. Studies of these aberrant pathways have substantially advanced understanding of the cellular mechanisms that contribute to mounting effective and balanced innate immune responses. For example, mutations affecting the function of cytosolic immune sensors known as inflammasomes and the IL-1 signalling pathway can trigger excessive inflammation. A surge in discovery of new genes associated with autoinflammation has pointed to other mechanisms of disease linking innate immune responses to a number of basic cellular pathways, such as maintenance of protein homeostasis (proteostasis), protein misfolding and clearance, endoplasmic reticulum stress and mitochondrial stress, metabolic stress, autophagy and abnormalities in differentiation and development of myeloid cells. Although the spectrum of autoinflammatory diseases has been steadily expanding, a substantial number of patients remain undiagnosed. Next-generation sequencing technologies will be instrumental in finding disease-causing mutations in as yet uncharacterized diseases. As more patients are reported to have clinical features of autoinflammation and immunodeficiency or autoimmunity, the complex interactions between the innate and adaptive immune systems are unveiled.
Collapse
|
185
|
Loose M, Hudel M, Zimmer KP, Garcia E, Hammerschmidt S, Lucas R, Chakraborty T, Pillich H. Pneumococcal hydrogen peroxide-induced stress signaling regulates inflammatory genes. J Infect Dis 2014; 211:306-16. [PMID: 25183769 DOI: 10.1093/infdis/jiu428] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Microbial infections can induce aberrant responses in cellular stress pathways, leading to translational attenuation, metabolic restriction, and activation of oxidative stress, with detrimental effects on cell survival. Here we show that infection of human airway epithelial cells with Streptococcus pneumoniae leads to induction of endoplasmic reticulum (ER) and oxidative stress, activation of mitogen-associated protein kinase (MAPK) signaling pathways, and regulation of their respective target genes. We identify pneumococcal H2O2 as the causative agent for these responses, as both catalase-treated and pyruvate oxidase-deficient bacteria lacked these activities. Pneumococcal H2O2 induced nuclear NF-κB translocation and transcription of proinflammatory cytokines. Inhibition of translational arrest and ER stress by salubrinal or of MAPK signaling pathways attenuate cytokine transcription. These results provide strong evidence for the notion that inhibition of translation is an important host pathway in monitoring harmful pathogen-associated activities, thereby enabling differentiation between pathogenic and nonpathogenic bacteria.
Collapse
Affiliation(s)
- Maria Loose
- Institute for Medical Microbiology, German Center for Infection Giessen-Marburg-Langen Site
| | - Martina Hudel
- Institute for Medical Microbiology, German Center for Infection Giessen-Marburg-Langen Site
| | | | - Ernesto Garcia
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, and Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Madrid, Spain
| | - Sven Hammerschmidt
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald, Germany
| | - Rudolf Lucas
- Vascular Biology Center, Department of Pharmacology and Toxicology, Georgia Health Sciences University, Augusta
| | - Trinad Chakraborty
- Institute for Medical Microbiology, German Center for Infection Giessen-Marburg-Langen Site
| | - Helena Pillich
- Institute for Medical Microbiology, German Center for Infection Giessen-Marburg-Langen Site
| |
Collapse
|
186
|
Wang M, Kaufman RJ. The impact of the endoplasmic reticulum protein-folding environment on cancer development. Nat Rev Cancer 2014; 14:581-97. [PMID: 25145482 DOI: 10.1038/nrc3800] [Citation(s) in RCA: 830] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The endoplasmic reticulum (ER) is an essential organelle in eukaryotic cells for the storage and regulated release of calcium and as the entrance to the secretory pathway. Protein misfolding in the ER causes accumulation of misfolded proteins (ER stress) and activation of the unfolded protein response (UPR), which has evolved to maintain a productive ER protein-folding environment. Both ER stress and UPR activation are documented in many different human cancers. In this Review, we summarize the impact of ER stress and UPR activation on every aspect of cancer and discuss outstanding questions for which answers will pave the way for therapeutics.
Collapse
Affiliation(s)
- Miao Wang
- Degenerative Diseases Program, Center for Cancer Research, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd, La Jolla, California 92037, USA
| | - Randal J Kaufman
- Degenerative Diseases Program, Center for Cancer Research, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd, La Jolla, California 92037, USA
| |
Collapse
|
187
|
PERK-dependent activation of JAK1 and STAT3 contributes to endoplasmic reticulum stress-induced inflammation. Mol Cell Biol 2014; 34:3911-25. [PMID: 25113558 DOI: 10.1128/mcb.00980-14] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Neuroinflammation and endoplasmic reticulum (ER) stress are associated with many neurological diseases. Here, we have examined the interaction between ER stress and JAK/STAT-dependent inflammation in glial cells. We show that ER stress is present in the central nervous system (CNS) concomitant with inflammation and astrogliosis in the multiple sclerosis (MS) mouse model of experimental autoimmune encephalomyelitis (EAE). Astrocytes do not easily succumb to ER stress but rather activate an inflammatory program involving activation of STAT3 in a JAK1-dependent fashion. ER stress-induced activation of the JAK1/STAT3 axis leads to expression of interleukin 6 (IL-6) and several chemokines. Moreover, the activation of STAT3 signaling is dependent on PERK, a central component of the ER stress response, which we show is phosphorylated by JAK1. Disruption of PERK abrogates ER stress-induced activation of STAT3 and subsequent gene expression. Additionally, ER-stressed astrocytes, via paracrine signaling, can stimulate activation of microglia, leading to production of IL-6 and oncostatin M (OSM). These IL-6 cytokines can then synergize with ER stress in astrocytes to drive inflammation. Together, this work describes a new PERK/JAK1/STAT3 signaling pathway that elicits a feed-forward inflammatory loop involving astrocytes and microglia to drive neuroinflammation, which may be relevant in diseases such as MS.
Collapse
|
188
|
van 't Wout EFA, Hiemstra PS, Marciniak SJ. The integrated stress response in lung disease. Am J Respir Cell Mol Biol 2014; 50:1005-9. [PMID: 24605820 DOI: 10.1165/rcmb.2014-0019tr] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Lungs are repeatedly exposed to inhaled toxic insults, such as smoke, diesel exhaust, and microbes, which elicit cellular stress responses. The phosphorylation of eukaryotic translation initiation factor 2α by one of four stress-sensing kinases triggers a pathway called the integrated stress response that helps protect cellular reserves of nutrients and prevents the accumulation of toxic proteins. In this review, we discuss how activation of the integrated stress response has been shown to play an important role in pulmonary pathology, and how its study may help in the development of novel therapies for diverse conditions, from hypoxia to cancer.
Collapse
Affiliation(s)
- Emily F A van 't Wout
- 1 Department of Pulmonology, Leiden University Medical Centre, Leiden, the Netherlands; and
| | | | | |
Collapse
|
189
|
Ciccia F, Accardo-Palumbo A, Rizzo A, Guggino G, Raimondo S, Giardina A, Cannizzaro A, Colbert RA, Alessandro R, Triolo G. Evidence that autophagy, but not the unfolded protein response, regulates the expression of IL-23 in the gut of patients with ankylosing spondylitis and subclinical gut inflammation. Ann Rheum Dis 2014; 73:1566-74. [PMID: 23740229 PMCID: PMC3883901 DOI: 10.1136/annrheumdis-2012-202925] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Interleukin (IL)-23 has been implicated in the pathogenesis of ankylosing spondylitis (AS). The aim of the study was to clarify the mechanisms underlying the increased IL-23 expression in the gut of AS patients. METHODS Consecutive gut biopsies from 30 HLA-B27(+) AS patients, 15 Crohn's disease (CD) patients and 10 normal subjects were obtained. Evidence for HLA-B27 misfolding was studied. Unfolded protein response (UPR) and autophagy were assessed by RT-PCR and immunohistochemistry. The contribution of UPR and autophagy in the regulation of IL-23 expression was evaluated in in vitro experiments on isolated lamina propria mononuclear cells (LPMCs). RESULTS Intracellular colocalisation of SYVN1 and FHCs but not a significant overexpression of UPR genes was observed in the gut of AS patients. Conversely, upregulation of the genes involved in the autophagy pathway was observed in the gut of AS and CD patients. Immunohistochemistry showed an increased expression of LC3II, ATG5 and ATG12 but not of SQSTM1 in the ileum of AS and CD patients. LC3II was expressed among infiltrating mononuclear cells and epithelial cells resembling Paneth cells (PC) and colocalised with ATG5 in AS and CD. Autophagy but not UPR was required to modulate the expression of IL-23 in isolated LPMCs of AS patients with chronic gut inflammation, CD patients and controls. CONCLUSIONS Our data suggest that HLA-B27 misfolding occurs in the gut of AS patients and is accompanied by activation of autophagy rather than a UPR. Autophagy appears to be associated with intestinal modulation of IL-23 in AS.
Collapse
Affiliation(s)
- Francesco Ciccia
- Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università di Palermo, Italy
| | | | - Aroldo Rizzo
- Anatomia Patologica, Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy
| | - Giuliana Guggino
- Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università di Palermo, Italy
| | - Stefania Raimondo
- Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di Palermo, Italy
| | - AnnaRita Giardina
- Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università di Palermo, Italy
| | | | - Robert A. Colbert
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| | - Riccardo Alessandro
- Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di Palermo, Italy
| | - Giovanni Triolo
- Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università di Palermo, Italy
| |
Collapse
|
190
|
Banerjee C, Singh A, Das TK, Raman R, Shrivastava A, Mazumder S. Ameliorating ER-stress attenuates Aeromonas hydrophila-induced mitochondrial dysfunctioning and caspase mediated HKM apoptosis in Clarias batrachus. Sci Rep 2014; 4:5820. [PMID: 25059203 PMCID: PMC5376045 DOI: 10.1038/srep05820] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 06/16/2014] [Indexed: 02/07/2023] Open
Abstract
Endoplasmic reticulum (ER)-stress and unfolding protein response (UPR) has not been implied in Aeromonas hydrophila-pathogenicity. We report increased expression of the ER-stress markers: CHOP, BiP and phospho-eIF2α in A. hydrophila-infected headkidney macrophages (HKM) in Clarias batrachus. Pre-treatment with ER-stress inhibitor, 4-PBA alleviated ER-stress and HKM apoptosis suggesting ER-UPR critical for the process. The ER-Ca(2+) released via inositol-triphosphate and ryanodine receptors induced calpain-2 mediated superoxide ion generation and consequent NF-κB activation. Inhibiting NF-κB activation attenuated NO production suggesting the pro-apoptotic role of NF-κB on HKM pathology. Calpain-2 activated caspase-12 to intensify the apoptotic cascade through mitochondrial-membrane potential (ψm) dissipation and caspase-9 activation. Altered mitochondrial ultra-structure consequent to ER-Ca(2+) uptake via uniporters reduced ψm and released cytochrome C. Nitric oxide induced the cGMP/PKG-dependent activation of caspase-8 and truncated-Bid formation. Both the caspases converge onto caspase-3 to execute HKM apoptosis. These findings offer a possible molecular explanation for A. hydrophila pathogenicity.
Collapse
Affiliation(s)
- Chaitali Banerjee
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| | - Ambika Singh
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| | - Taposh Kumar Das
- Department of Anatomy, All India Institute of Medical Sciences, Delhi 110 029, India
| | - Rajagopal Raman
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| | - Anju Shrivastava
- Cell Signalling and Molecular Immunology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| | - Shibnath Mazumder
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi 110 007, India
| |
Collapse
|
191
|
Cao SS, Kaufman RJ. Endoplasmic reticulum stress and oxidative stress in cell fate decision and human disease. Antioxid Redox Signal 2014; 21:396-413. [PMID: 24702237 PMCID: PMC4076992 DOI: 10.1089/ars.2014.5851] [Citation(s) in RCA: 984] [Impact Index Per Article: 89.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SIGNIFICANCE The endoplasmic reticulum (ER) is a specialized organelle for the folding and trafficking of proteins, which is highly sensitive to changes in intracellular homeostasis and extracellular stimuli. Alterations in the protein-folding environment cause accumulation of misfolded proteins in the ER that profoundly affect a variety of cellular signaling processes, including reduction-oxidation (redox) homeostasis, energy production, inflammation, differentiation, and apoptosis. The unfolded protein response (UPR) is a collection of adaptive signaling pathways that evolved to resolve protein misfolding and restore an efficient protein-folding environment. RECENT ADVANCES Production of reactive oxygen species (ROS) has been linked to ER stress and the UPR. ROS play a critical role in many cellular processes and can be produced in the cytosol and several organelles, including the ER and mitochondria. Studies suggest that altered redox homeostasis in the ER is sufficient to cause ER stress, which could, in turn, induce the production of ROS in the ER and mitochondria. CRITICAL ISSUES Although ER stress and oxidative stress coexist in many pathologic states, whether and how these stresses interact is unknown. It is also unclear how changes in the protein-folding environment in the ER cause oxidative stress. In addition, how ROS production and protein misfolding commit the cell to an apoptotic death and contribute to various degenerative diseases is unknown. FUTURE DIRECTIONS A greater fundamental understanding of the mechanisms that preserve protein folding homeostasis and redox status will provide new information toward the development of novel therapeutics for many human diseases.
Collapse
Affiliation(s)
- Stewart Siyan Cao
- 1 Degenerative Diseases Program, Sanford Burnham Medical Research Institute , La Jolla, California
| | | |
Collapse
|
192
|
Rao J, Yue S, Fu Y, Zhu J, Wang X, Busuttil RW, Kupiec-Weglinski JW, Lu L, Zhai Y. ATF6 mediates a pro-inflammatory synergy between ER stress and TLR activation in the pathogenesis of liver ischemia-reperfusion injury. Am J Transplant 2014; 14:1552-61. [PMID: 24903305 PMCID: PMC4074706 DOI: 10.1111/ajt.12711] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 02/07/2014] [Accepted: 02/15/2014] [Indexed: 01/25/2023]
Abstract
Although the roles of the metabolic stress in organ ischemia-reperfusion injury (IRI) have been well recognized, the question of whether and how these stress responses regulate innate immune activation against IR remains unclear. In a murine liver partial warm ischemia mode, we showed that prolonged ischemia triggered endoplasmic reticulum (ER) stress response, particularly, the activating transcription factor 6 (ATF6) branch, in liver Kupffer cells (KCs) and altered their responsiveness against Toll-like receptor (TLR) stimulation. Ischemia-primed cells increased pro-, but decreased anti-, inflammatory cytokine productions. Alleviation of ER stress in vivo by small chemical chaperon 4-phenylbutyrate or ATF6 small interfering RNA (siRNA) diminished the pro-inflammatory priming effect of ischemia in KCs, leading to the inhibition of liver immune response against IR and protection of livers from IRI. In vitro, ATF6 siRNA abrogated the ER stress-mediated pro-inflammatory enhancement of macrophage TLR4 response, by restricting NF-κB and restoring Akt activations. Thus, ischemia primes liver innate immune cells by ATF6-mediated ER stress response. The IR-induced metabolic stress and TLR activation function in synergy to activate tissue inflammatory immune response.
Collapse
Affiliation(s)
- Jianhua Rao
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA, USA,Department of Liver Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiansu Province, China
| | - Shi Yue
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA, USA
| | - Yuanfang Fu
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA, USA
| | - Jianjun Zhu
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA, USA,Department of Liver Surgery, Renji Hospital, Shanghai Jiaotong University Medical School, Shanghai, China
| | - Xuehao Wang
- Department of Liver Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiansu Province, China
| | - Ronald W. Busuttil
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA, USA
| | - Jerzy W. Kupiec-Weglinski
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA, USA
| | - Ling Lu
- Department of Liver Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiansu Province, China,Address correspondence: Yuan Zhai, MD, PhD. Dumont-UCLA Transplant Center 77-120 CHS, 10833 Le Conte Ave, Los Angeles, CA 90095. Phone: (310) 825-9426; Fax: (310) 267-2367, ; Ling Lu, MD, Department of Liver Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guanzhou Road, Nanjing, P.R.China, Phone: 86-25-68136053; Fax:86-25-84630769;
| | - Yuan Zhai
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA, USA,Address correspondence: Yuan Zhai, MD, PhD. Dumont-UCLA Transplant Center 77-120 CHS, 10833 Le Conte Ave, Los Angeles, CA 90095. Phone: (310) 825-9426; Fax: (310) 267-2367, ; Ling Lu, MD, Department of Liver Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guanzhou Road, Nanjing, P.R.China, Phone: 86-25-68136053; Fax:86-25-84630769;
| |
Collapse
|
193
|
Benham H, Rehaume LM, Hasnain SZ, Velasco J, Baillet AC, Ruutu M, Kikly K, Wang R, Tseng HW, Thomas GP, Brown MA, Strutton G, McGuckin MA, Thomas R. Interleukin-23 mediates the intestinal response to microbial β-1,3-glucan and the development of spondyloarthritis pathology in SKG mice. Arthritis Rheumatol 2014; 66:1755-67. [PMID: 24664521 DOI: 10.1002/art.38638] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 03/18/2014] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Spondyloarthritides (SpA) occur in 1% of the population and include ankylosing spondylitis (AS) and arthropathy of inflammatory bowel disease (IBD), with characteristic spondylitis, arthritis, enthesitis, and IBD. Genetic studies implicate interleukin-23 (IL-23) receptor signaling in the development of SpA and IBD, and IL-23 overexpression in mice is sufficient for enthesitis, driven by entheseal-resident T cells. However, in genetically prone individuals, it is not clear where IL-23 is produced and how it drives the SpA syndrome, including IBD or subclinical gut inflammation of AS. Moreover, it is unclear why specific tissue involvement varies between patients with SpA. We undertook this study to determine the location of IL-23 production and its role in SpA pathogenesis in BALB/c ZAP-70(W163C)-mutant (SKG) mice injected intraperitoneally with β-1,3-glucan (curdlan). METHODS Eight weeks after curdlan injection in wild-type or IL-17A(-/-) SKG or BALB/c mice, pathology was scored in tissue sections. Mice were treated with anti-IL-23 or anti-IL-22. Cytokine production and endoplasmic reticulum (ER) stress were determined in affected organs. RESULTS In curdlan-treated SKG mice, arthritis, enthesitis, and ileitis were IL-23 dependent. Enthesitis was specifically dependent on IL-17A and IL-22. IL-23 was induced in the ileum, where it amplified ER stress, goblet cell dysfunction, and proinflammatory cytokine production. IL-17A was pathogenic, while IL-22 was protective against ileitis. IL-22+CD3- innate-like cells were increased in lamina propria mononuclear cells of ileitis-resistant BALB/c mice, which developed ileitis after curdlan injection and anti-IL-22. CONCLUSION In response to systemic β-1,3-glucan, intestinal IL-23 provokes local mucosal dysregulation and cytokines driving the SpA syndrome, including IL-17/IL-22-dependent enthesitis. Innate IL-22 production promotes ileal tolerance.
Collapse
Affiliation(s)
- Helen Benham
- University of Queensland and Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Rodríguez M, Domingo E, Alonso S, Frade JG, Eiros J, Crespo MS, Fernández N. The unfolded protein response and the phosphorylations of activating transcription factor 2 in the trans-activation of il23a promoter produced by β-glucans. J Biol Chem 2014; 289:22942-22957. [PMID: 24982422 DOI: 10.1074/jbc.m113.522656] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Current views on the control of IL-23 production focus on the regulation of il23a, the gene encoding IL-23 p19, by NF-κB in combination with other transcription factors. C/EBP homologous protein (CHOP), X2-Box-binding protein 1 (XBP1), activator protein 1 (AP1), SMAD, CCAAT/enhancer-binding protein (C/EBPβ), and cAMP-response element-binding protein (CREB) have been involved in response to LPS, but no data are available regarding the mechanism triggered by the fungal mimic and β-glucan-containing stimulus zymosan, which produces IL-23 and to a low extent the related cytokine IL-12 p70. Zymosan induced the mobilization of CHOP from the nuclear fractions to phagocytic vesicles. Hypha-forming Candida also induced the nuclear disappearance of CHOP. Assay of transcription factor binding to the il23a promoter showed an increase of Thr(P)-71-Thr(P)-69-activating transcription factor 2 (ATF2) binding in response to zymosan. PKC and PKA/mitogen- and stress-activated kinase inhibitors down-regulated Thr(P)-71-ATF2 binding to the il23a promoter and il23a mRNA expression. Consistent with the current concept of complementary phosphorylations on N-terminal Thr-71 and Thr-69 of ATF2 by ERK and p38 MAPK, MEK, and p38 MAPK inhibitors blunted Thr(P)-69-ATF2 binding. Knockdown of atf2 mRNA with siRNA correlated with inhibition of il23a mRNA, but it did not affect the expression of il12/23b and il10 mRNA. These data indicate the following: (i) zymosan decreases nuclear proapoptotic CHOP, most likely by promoting its accumulation in phagocytic vesicles; (ii) zymosan-induced il23a mRNA expression is best explained through coordinated κB- and ATF2-dependent transcription; and (iii) il23a expression relies on complementary phosphorylation of ATF2 on Thr-69 and Thr-71 dependent on PKC and MAPK activities.
Collapse
Affiliation(s)
- Mario Rodríguez
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valladolid, 47005-Valladolid
| | - Esther Domingo
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, 47003-Valladolid
| | - Sara Alonso
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, 47003-Valladolid
| | - Javier García Frade
- Division of Hematology, Hospital Universitario Rio Hortega, 47012-Valladolid, and
| | - José Eiros
- Division of Microbiology, Hospital Universitario Rio Hortega, 47012-Valladolid, Spain
| | - Mariano Sánchez Crespo
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, 47003-Valladolid,.
| | - Nieves Fernández
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valladolid, 47005-Valladolid
| |
Collapse
|
195
|
Zhao C, Pavicic PG, Datta S, Sun D, Novotny M, Hamilton TA. Cellular stress amplifies TLR3/4-induced CXCL1/2 gene transcription in mononuclear phagocytes via RIPK1. THE JOURNAL OF IMMUNOLOGY 2014; 193:879-88. [PMID: 24920846 DOI: 10.4049/jimmunol.1303396] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The impact of environmental stressors on the magnitude of specific chemokine gene expression was examined in mouse bone marrow-derived macrophages stimulated through various TLRs. Levels of TLR-stimulated CXCL1 and CXCL2 but not CXCL10 or CCL5 mRNAs were selectively enhanced (>10-fold) in stressed macrophages. The amplification was also manifested for other proinflammatory cytokines, including TNF-α, IL-1α, and IL-6. Responses through TLR3 and TLR4 exhibited the greatest sensitivity, reflecting a requirement for Toll/IL-IR domain-containing adaptor-inducing IFN-β (TRIF), the adaptor protein selectively associated with these TLRs. IFN regulatory factor 3, a transcription factor that is downstream of TLR4/TRIF signaling, was not required for sensitivity to stress-induced chemokine amplification. c/EBP homologous protein and X box binding protein 1 have been reported to enhance inflammatory cytokine responses but are not required for amplification of TLR3/4-induced CXCL1 expression. Rather, receptor-interacting protein kinase 1, a kinase also linked with TLR3/4/TRIF signaling, is required and involves a stress-dependent increase in its abundance and ubiquitination. Whereas NF-κB activation is necessary for TLR-induced chemokine gene transcription, this factor does not appear to be the primary mechanistic target of environmental stress. The application of stress also enhanced chemokine expression in macrophages infiltrating the peritoneal cavity but was not observed in the resident peritoneal cells or in the liver. These findings identify novel mechanisms for modulating the magnitude and duration of selective TLR-induced chemokine and cytokine gene expression and further establish the importance of cell stress pathways in coordinating the outcomes of cellular and tissue injury.
Collapse
Affiliation(s)
- Chenyang Zhao
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Paul G Pavicic
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Shyamasree Datta
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Dongxu Sun
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Michael Novotny
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Thomas A Hamilton
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| |
Collapse
|
196
|
Smith JA, Colbert RA. Review: The interleukin-23/interleukin-17 axis in spondyloarthritis pathogenesis: Th17 and beyond. Arthritis Rheumatol 2014; 66:231-41. [PMID: 24504793 DOI: 10.1002/art.38291] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 11/19/2013] [Indexed: 12/13/2022]
|
197
|
Smith JA. A new paradigm: innate immune sensing of viruses via the unfolded protein response. Front Microbiol 2014; 5:222. [PMID: 24904537 PMCID: PMC4032990 DOI: 10.3389/fmicb.2014.00222] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 04/27/2014] [Indexed: 12/17/2022] Open
Abstract
The immune system depends upon combinations of signals to mount appropriate responses: pathogen specific signals in the context of co-stimulatory “danger” signals drive immune strength and accuracy. Viral infections trigger anti-viral type I interferon (IFN) responses by stimulating endosomal and cytosolic pattern recognition receptors (PRRs). However, viruses have also evolved many strategies to counteract IFN responses. Are there intracellular danger signals that enhance immune responses to viruses? During infection, viruses place a heavy demand on the protein folding machinery of the host endoplasmic reticulum (ER). To survive ER stress, host cells mount an unfolded protein response (UPR) to decrease ER protein load and enhance protein-folding capacity. Viruses also directly elicit the UPR to enhance their replication. Increasing evidence supports an intersection between the host UPR and inflammation, in particular the production of pro-inflammatory cytokines and type I IFN. The UPR directly activates pro-inflammatory cytokine transcription factors and dramatically enhances cytokine production in response to viral PRR engagement. Additionally, viral PRR engagement may stimulate specific pathways within the UPR to enhance cytokine production. Through these mechanisms, viral detection via the UPR and inflammatory cytokine production are intertwined. Consequently, the UPR response is perfectly poised to act as an infection-triggered “danger” signal. The UPR may serve as an internal “co-stimulatory” signal that (1) provides specificity and (2) critically augments responses to overcome viral subterfuge. Further work is needed to test this hypothesis during viral infections.
Collapse
Affiliation(s)
- Judith A Smith
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health Madison, WI, USA
| |
Collapse
|
198
|
Mehlitz A, Karunakaran K, Herweg JA, Krohne G, van de Linde S, Rieck E, Sauer M, Rudel T. The chlamydial organism Simkania negevensis forms ER vacuole contact sites and inhibits ER-stress. Cell Microbiol 2014; 16:1224-43. [PMID: 24528559 DOI: 10.1111/cmi.12278] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 01/28/2014] [Accepted: 02/05/2014] [Indexed: 01/01/2023]
Abstract
Most intracellular bacterial pathogens reside within membrane-surrounded host-derived vacuoles. Few of these bacteria exploit membranes from the host's endoplasmic reticulum (ER) to form a replicative vacuole. Here, we describe the formation of ER-vacuole contact sites as part of the replicative niche of the chlamydial organism Simkania negevensis. Formation of ER-vacuole contact sites is evolutionary conserved in the distantly related protozoan host Acanthamoeba castellanii. Simkania growth is accompanied by mitochondria associating with the Simkania-containing vacuole (SCV). Super-resolution microscopy as well as 3D reconstruction from electron micrographs of serial ultra-thin sections revealed a single vacuolar system forming extensive ER-SCV contact sites on the Simkania vacuolar surface. Simkania infection induced an ER-stress response, which was later downregulated. Induction of ER-stress with Thapsigargin or Tunicamycin was strongly inhibited in cells infected with Simkania. Inhibition of ER-stress was required for inclusion formation and efficient growth, demonstrating a role of ER-stress in the control of Simkania infection. Thus, Simkania forms extensive ER-SCV contact sites in host species evolutionary as diverse as human and amoeba. Moreover, Simkania is the first bacterial pathogen described to interfere with ER-stress induced signalling to promote infection.
Collapse
Affiliation(s)
- Adrian Mehlitz
- University of Wuerzburg, Biocenter, Department of Microbiology, Am Hubland, D-97074, Wuerzburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
199
|
Bates J, Diehl L. Dendritic cells in IBD pathogenesis: an area of therapeutic opportunity? J Pathol 2014; 232:112-20. [PMID: 24122796 PMCID: PMC4285849 DOI: 10.1002/path.4277] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 09/03/2013] [Accepted: 09/22/2013] [Indexed: 12/15/2022]
Abstract
Dysfunction of the mucosal immune system plays an important role in inflammatory bowel disease (IBD) pathogenesis. Dendritic cells are emerging as central players based on both our increasing understanding of how genetic susceptibility impacts the mucosal immune system and the key role of dendritic cells in regulating response to gut microflora. We discuss areas of therapeutic opportunity in this evolving landscape. © 2013 The Authors. Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
|
200
|
Shenderov K, Riteau N, Yip R, Mayer-Barber KD, Oland S, Hieny S, Fitzgerald P, Oberst A, Dillon CP, Green DR, Cerundolo V, Sher A. Cutting edge: Endoplasmic reticulum stress licenses macrophages to produce mature IL-1β in response to TLR4 stimulation through a caspase-8- and TRIF-dependent pathway. THE JOURNAL OF IMMUNOLOGY 2014; 192:2029-2033. [PMID: 24489101 DOI: 10.4049/jimmunol.1302549] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The accumulation of improperly folded proteins within the endoplasmic reticulum (ER) generates perturbations known as ER stress that engage the unfolded protein response. ER stress is involved in many inflammatory pathologies that are also associated with the production of the proinflammatory cytokine IL-1β. In this study, we demonstrate that macrophages undergoing ER stress are able to drive the production and processing of pro-IL-1β in response to LPS stimulation in vitro. Interestingly, the classical NLRP3 inflammasome is dispensable, because maturation of pro-IL-1β occurs normally in the absence of the adaptor protein ASC. In contrast, processing of pro-IL-1β is fully dependent on caspase-8. Intriguingly, we found that neither the unfolded protein response transcription factors XBP1 and CHOP nor the TLR4 adaptor molecule MyD88 is necessary for caspase-8 activation. Instead, both caspase activation and IL-1β production require the alternative TLR4 adaptor TRIF. This pathway may contribute to IL-1-driven tissue pathology in certain disease settings.
Collapse
Affiliation(s)
- Kevin Shenderov
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK, OX3 9DS.,Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD. 20892
| | - Nicolas Riteau
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD. 20892
| | - Ronald Yip
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK, OX3 9DS
| | - Katrin D Mayer-Barber
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD. 20892
| | - Sandy Oland
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD. 20892
| | - Sara Hieny
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD. 20892
| | - Pat Fitzgerald
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Andrew Oberst
- Department of Immunology, University of Washington, Seattle, WA 98109
| | - Christopher P Dillon
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Vincenzo Cerundolo
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK, OX3 9DS
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD. 20892
| |
Collapse
|