1
|
McFarlin BK, Deemer SE, Bridgeman EA. Oral Spore-Based Probiotic Supplementation Alters Post-Prandial Expression of mRNA Associated with Gastrointestinal Health. Biomedicines 2024; 12:2386. [PMID: 39457699 PMCID: PMC11504401 DOI: 10.3390/biomedicines12102386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Unregulated post-prandial dietary endotoxemia may accumulate over time and underlie the development of chronic disease (e.g., leaky gut, inflammatory bowel disease, etc.), for which oral probiotic supplementation may be a prophylactic. The purpose of this study was to determine if 45 d of oral spore-based probiotic supplementation altered gastrointestinal-associated mRNA expression following a high-fat meal. Methods: A subset of apparently healthy individuals from a larger study who had dietary endotoxemia at baseline completed 45 d of supplementation with either a placebo (rice flour; n = 10) or spore-based probiotic (Megasporebiotic™; Novonesis, Kongens Lyngby, Denmark; Bacillus indicus (HU36™), Bacillus subtilis (HU58™), Bacillus coagulans (SC208™), and Bacillus licheniformis (SL-307), and Bacillus clausii (SC109™); n = 10). Venous blood was collected in Paxgene RNA tubes prior to (PRE), 3 h, and 5 h after consumption of a high-fat meal (85% of the daily fat RDA and 65% of the daily calorie needs). Total RNA was analyzed for 579 mRNAs of interest (Nanostring nCounter Sprint; Seattle, WA, USA). After normalization to housekeeping controls and calculation of differential expression relative to PRE and controlled for FDR, 15 mRNAs were determined to be significantly changed at either 3 h and/or 5 h post-prandial in the probiotic group but not in the placebo group. Results: Significant mRNA expressions were associated with gastrointestinal tract barrier function (four mRNAs: BATF3, CCR6, CXCR6, and PDCD2), gastrointestinal immunity (four mRNAs: CLEC5A, IL7, CARD9, and FCER1G), or future IBD risk (seven mRNAs: PD-L1, CSF1R, FAS, BID, FADD, GATA3, and KIR3DL). Conclusions: Collectively, the present findings may support the notion that post-prandial immune response to eating is enhanced following 45 d of probiotic supplementation.
Collapse
Affiliation(s)
- Brian K. McFarlin
- Applied Physiology Laboratory, University of North Texas, Denton, TX 76205, USA; (S.E.D.); (E.A.B.)
- Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA
| | - Sarah E. Deemer
- Applied Physiology Laboratory, University of North Texas, Denton, TX 76205, USA; (S.E.D.); (E.A.B.)
| | - Elizabeth A. Bridgeman
- Applied Physiology Laboratory, University of North Texas, Denton, TX 76205, USA; (S.E.D.); (E.A.B.)
- Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA
| |
Collapse
|
2
|
Lombardi F, Augello FR, Palumbo P, Bonfili L, Artone S, Altamura S, Sheldon JM, Latella G, Cifone MG, Eleuteri AM, Cinque B. Bacterial Lysate from the Multi-Strain Probiotic SLAB51 Triggers Adaptative Responses to Hypoxia in Human Caco-2 Intestinal Epithelial Cells under Normoxic Conditions and Attenuates LPS-Induced Inflammatory Response. Int J Mol Sci 2023; 24:ijms24098134. [PMID: 37175841 PMCID: PMC10179068 DOI: 10.3390/ijms24098134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/28/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α), a central player in maintaining gut-microbiota homeostasis, plays a pivotal role in inducing adaptive mechanisms to hypoxia and is negatively regulated by prolyl hydroxylase 2 (PHD2). HIF-1α is stabilized through PI3K/AKT signaling regardless of oxygen levels. Considering the crucial role of the HIF pathway in intestinal mucosal physiology and its relationships with gut microbiota, this study aimed to evaluate the ability of the lysate from the multi-strain probiotic formulation SLAB51 to affect the HIF pathway in a model of in vitro human intestinal epithelium (intestinal epithelial cells, IECs) and to protect from lipopolysaccharide (LPS) challenge. The exposure of IECs to SLAB51 lysate under normoxic conditions led to a dose-dependent increase in HIF-1α protein levels, which was associated with higher glycolytic metabolism and L-lactate production. Probiotic lysate significantly reduced PHD2 levels and HIF-1α hydroxylation, thus leading to HIF-1α stabilization. The ability of SLAB51 lysate to increase HIF-1α levels was also associated with the activation of the PI3K/AKT pathway and with the inhibition of NF-κB, nitric oxide synthase 2 (NOS2), and IL-1β increase elicited by LPS treatment. Our results suggest that the probiotic treatment, by stabilizing HIF-1α, can protect from an LPS-induced inflammatory response through a mechanism involving PI3K/AKT signaling.
Collapse
Affiliation(s)
- Francesca Lombardi
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | | | - Paola Palumbo
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Serena Artone
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Serena Altamura
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Jenna Marie Sheldon
- Dr. Kiran C Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314-7796, USA
| | - Giovanni Latella
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Maria Grazia Cifone
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| |
Collapse
|
3
|
Tang YY, Wang DC, Wang YQ, Huang AF, Xu WD. Emerging role of hypoxia-inducible factor-1α in inflammatory autoimmune diseases: A comprehensive review. Front Immunol 2023; 13:1073971. [PMID: 36761171 PMCID: PMC9905447 DOI: 10.3389/fimmu.2022.1073971] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/13/2022] [Indexed: 01/26/2023] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is a primary metabolic sensor, and is expressed in different immune cells, such as macrophage, dendritic cell, neutrophil, T cell, and non-immune cells, for instance, synovial fibroblast, and islet β cell. HIF-1α signaling regulates cellular metabolism, triggering the release of inflammatory cytokines and inflammatory cells proliferation. It is known that microenvironment hypoxia, vascular proliferation, and impaired immunological balance are present in autoimmune diseases. To date, HIF-1α is recognized to be overexpressed in several inflammatory autoimmune diseases, such as systemic lupus erythematosus (SLE), rheumatoid arthritis, and function of HIF-1α is dysregulated in these diseases. In this review, we narrate the signaling pathway of HIF-1α and the possible immunopathological roles of HIF-1α in autoimmune diseases. The collected information will provide a theoretical basis for the familiarization and development of new clinical trials and treatment based on HIF-1α and inflammatory autoimmune disorders in the future.
Collapse
Affiliation(s)
- Yang-Yang Tang
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Da-Cheng Wang
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - You-Qiang Wang
- Department of Laboratory Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - An-Fang Huang
- Department of Rheumatology and Immunology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wang-Dong Xu
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China,*Correspondence: Wang-Dong Xu,
| |
Collapse
|
4
|
Lun J, Zhang H, Guo J, Yu M, Fang J. Hypoxia inducible factor prolyl hydroxylases in inflammatory bowel disease. Front Pharmacol 2023; 14:1045997. [PMID: 37201028 PMCID: PMC10187758 DOI: 10.3389/fphar.2023.1045997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 04/18/2023] [Indexed: 05/20/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic disease that is characterized by intestinal inflammation. Epithelial damage and loss of intestinal barrier function are believed to be the hallmark pathologies of the disease. In IBD, the resident and infiltrating immune cells consume much oxygen, rendering the inflamed intestinal mucosa hypoxic. In hypoxia, the hypoxia-inducible factor (HIF) is induced to cope with the lack of oxygen and protect intestinal barrier. Protein stability of HIF is tightly controlled by prolyl hydroxylases (PHDs). Stabilization of HIF through inhibition of PHDs is appearing as a new strategy of IBD treatment. Studies have shown that PHD-targeting is beneficial to the treatment of IBD. In this Review, we summarize the current understanding of the role of HIF and PHDs in IBD and discuss the therapeutic potential of targeting PHD-HIF pathway for IBD treatment.
Collapse
Affiliation(s)
- Jie Lun
- Department of Oncology, Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongwei Zhang
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Jing Guo
- Department of Oncology, Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mengchao Yu
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, China
| | - Jing Fang
- Department of Oncology, Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Jing Fang,
| |
Collapse
|
5
|
Liu Y, Li X, Zhou X, Wang J, Ao X. FADD as a key molecular player in cancer progression. Mol Med 2022; 28:132. [DOI: 10.1186/s10020-022-00560-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/05/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022] Open
Abstract
AbstractCancer is a leading disease-related cause of death worldwide. Despite advances in therapeutic interventions, cancer remains a major global public health problem. Cancer pathogenesis is extremely intricate and largely unknown. Fas-associated protein with death domain (FADD) was initially identified as an adaptor protein for death receptor-mediated extrinsic apoptosis. Recent evidence suggests that FADD plays a vital role in non-apoptotic cellular processes, such as proliferation, autophagy, and necroptosis. FADD expression and activity of are modulated by a complicated network of processes, such as DNA methylation, non-coding RNA, and post-translational modification. FADD dysregulation has been shown to be closely associated with the pathogenesis of numerous types of cancer. However, the detailed mechanisms of FADD dysregulation involved in cancer progression are still not fully understood. This review mainly summarizes recent findings on the structure, functions, and regulatory mechanisms of FADD and focuses on its role in cancer progression. The clinical implications of FADD as a biomarker and therapeutic target for cancer patients are also discussed. The information reviewed herein may expand researchers’ understanding of FADD and contribute to the development of FADD-based therapeutic strategies for cancer patients.
Collapse
|
6
|
Steiner CA, Cartwright IM, Taylor CT, Colgan SP. Hypoxia-inducible factor as a bridge between healthy barrier function, wound healing, and fibrosis. Am J Physiol Cell Physiol 2022; 323:C866-C878. [PMID: 35912990 PMCID: PMC9467472 DOI: 10.1152/ajpcell.00227.2022] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/07/2022] [Accepted: 07/23/2022] [Indexed: 11/22/2022]
Abstract
The healthy mammalian intestine is lined by a single layer of epithelial cells. These cells provide a selectively permeable barrier to luminal contents and normally do so in an efficient and effective manner. Barrier function in the healthy mucosa is provided via several mechanisms including epithelial junctional complexes, mucus production, as well as mucosal-derived antimicrobial proteins. As tissue metabolism is central to the maintenance of homeostasis in the mucosa, intestinal [Formula: see text] levels are uniquely low due to counter-current blood flow and the presence of the microbiota, resulting in the stabilization of the transcription factor hypoxia-inducible factor (HIF). Ongoing studies have revealed that HIF molds normal intestinal metabolism and is central to the coordination of barrier regulation during both homeostasis and active disease. During acute inflammation, HIF is central to controlling the rapid restitution of the epithelium consistent with normal wound healing responses. In contrast, HIF may also contribute to the fibrostenotic response associated with chronic, nonresolving inflammation. As such, HIF may function as a double-edged sword in the overall course of the inflammatory response. Here, we review recent literature on the contribution of HIF to mucosal barrier function, wound healing, and fibrosis.
Collapse
Affiliation(s)
- Calen A Steiner
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado
| | - Ian M Cartwright
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado
| | - Cormac T Taylor
- School of Medicine, Conway Institute and Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Sean P Colgan
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado
| |
Collapse
|
7
|
Liu S, Yu H, Li P, Wang C, Liu G, Zhang X, Zhang C, Qi M, Ji H. Dietary nano-selenium alleviated intestinal damage of juvenile grass carp ( Ctenopharyngodon idella) induced by high-fat diet: Insight from intestinal morphology, tight junction, inflammation, anti-oxidization and intestinal microbiota. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2022; 8:235-248. [PMID: 34988305 PMCID: PMC8688880 DOI: 10.1016/j.aninu.2021.07.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/24/2021] [Accepted: 07/11/2021] [Indexed: 01/30/2023]
Abstract
In recent years, high-fat diet (HFD) has been widely applied in aquaculture, which reduces the intestinal health of cultured fish. The current study evaluated the protective effects of nano-selenium (nano-Se) on intestinal health of juvenile grass carp (Ctenopharyngodon idella) fed with HFD. A total of 135 experimental fish were fed with a regular diet (Con), a HFD (HFD) and a HFD containing nano-Se at 0.6 mg/kg (HSe) for 10 weeks. The results showed that dietary nano-Se significantly improved the survival rate and feed efficiency which were reduced by HFD in juvenile grass carp (P < 0.05). Also, nano-Se (0.6 mg/kg) supplement alleviated intestinal damage caused by the HFD, thus maintaining the integrity of the intestine. Moreover, it significantly up-regulated the expression of genes related to tight junction (ZO-1, c laudin-3 and o ccludin), anti-oxidization (GPx4a andGPx4b), and the protein of ZO-1 in the intestine of juvenile grass carp, which were depressed by the HFD (P < 0.05). Furthermore, nano-Se supplementation significantly suppressed the expressions of genes related to the inflammation, including inflammatory cytokines (IL-8, IL-1β, IFN-γ, TNF-α and IL-6), signaling molecules (TLR4, p38 MAPK and NF-κB p65), and protein expression of NF-κB p65 and TNF-α in the intestine of juvenile grass carp which were induced by the HFD (P < 0.05). Besides, dietary nano-Se normalized the intestinal microbiota imbalance of juvenile grass carp caused by the HFD through increasing the abundance of the beneficial bacteria, e.g., Fusobacteria. Finally, dietary nano-Se increased the production of short chain fatty acids (SCFA) in the intestine, especially for butyric acid and caproic acid, which were negatively related to the increase of intestinal permeability and inflammation. In summary, supply of nano-Se (0.6 mg/kg) in HFD could effectively alleviate intestinal injury of juvenile grass carp by improving intestinal barrier function and reducing intestinal inflammation and oxidative stress. These positive effects may be due to the regulation of nano-Se on intestinal microbiota and the subsequently increased beneficial SCFA levels.
Collapse
Affiliation(s)
- Sha Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Haibo Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Pengju Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Chi Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Guohao Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xiaotian Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Cheng Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Meng Qi
- Key Laboratory of Se-enriched Products Development and Quality Control, Ministry of Agriculture and Rural Affairs, Ankang, 725000, China
| | - Hong Ji
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| |
Collapse
|
8
|
De Galan C, De Vos M, Hindryckx P, Laukens D, Van Welden S. Long-Term Environmental Hypoxia Exposure and Haematopoietic Prolyl Hydroxylase-1 Deletion Do Not Impact Experimental Crohn's Like Ileitis. BIOLOGY 2021; 10:biology10090887. [PMID: 34571764 PMCID: PMC8464968 DOI: 10.3390/biology10090887] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/23/2021] [Accepted: 09/03/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary Hypoxia-induced signalling represents an important contributor to inflammatory bowel disease (IBD) pathophysiology. However, available data solely focus on colonic inflammation while the primary disease location in Crohn’s disease patients is the terminal ileum. Therefore, we explored the effects of environmental hypoxia and immune cell-specific deletion of oxygen sensor prolyl hydroxylase (PHD) 1 in a Crohn’s like ileitis mouse model. Five-week-old TNF∆ARE/+ mice and wildtype (WT) littermates were housed in normoxia (21% O2) or hypoxia (8% O2) for 10 weeks. Although environmental hypoxia increased both systemic as ileal markers of hypoxia, the body weight evolution in both WT and TNF∆ARE/+ mice was not affected. Interestingly, hypoxia did increase circulatory monocytes, ileal mononuclear phagocytes and proinflammatory cytokine expression in WT mice. However, no histological or inflammatory gene expression differences in the ileum could be identified between TNF∆ARE/+ mice housed in hypoxia versus normoxia nor between TNF∆ARE/+ and WT mice with additional loss of immune cell-specific Phd1 expression. This is the first study showing that long-term environmental hypoxia or haematopoietic Phd1-deletion does not impact experimental ileitis. Therefore, it strongly questions whether targeting hypoxia-induced signalling via currently available PHD inhibitors would exert an immune suppressive effect in IBD patients with ileal inflammation. Abstract Environmental hypoxia and hypoxia-induced signalling in the gut influence inflammatory bowel disease pathogenesis, however data is limited to colitis. Hence, we investigated the effect of environmental hypoxia and immune cell-specific deletion of oxygen sensor prolyl hydroxylase (PHD) 1 in a Crohn’s like ileitis mouse model. Therefore, 5-week-old C57/BL6 TNF∆ARE/+ mice and wildtype (WT) littermates were housed in normoxia (21% O2) or hypoxia (8% O2) for 10 weeks. Systemic inflammation was assessed by haematology. Distal ileal hypoxia was evaluated by pimonidazole staining. The ileitis degree was scored on histology, characterized via qPCR and validated in haematopoietic Phd1-deficient TNF∆ARE/+ mice. Our results demonstrated that hypoxia did not impact body weight evolution in WT and TNF∆ARE/+ mice. Hypoxia increased red blood cell count, haemoglobin, haematocrit and increased pimonidazole intensity in the ileum. Interestingly, hypoxia evoked an increase in circulatory monocytes, ileal mononuclear phagocytes and proinflammatory cytokine expression in WT mice. Despite these alterations, no histological or ileal gene expression differences could be identified between TNF∆ARE/+ mice housed in hypoxia versus normoxia nor between haematopoietic Phd1-deficient TNF∆ARE/+ and their WT counterparts. Therefore, we demonstrated for the first time that long-term environmental hypoxia or haematopoietic Phd1-deletion does not impact experimental ileitis development.
Collapse
Affiliation(s)
- Cara De Galan
- Department of Internal Medicine and Paediatrics, Ghent University, 9000 Ghent, Belgium; (C.D.G.); (M.D.V.); (P.H.); (D.L.)
- Ghent Gut Inflammation Group (GGIG), Ghent University, 9000 Ghent, Belgium
- VIB Centre for Inflammation Research, 9000 Ghent, Belgium
| | - Martine De Vos
- Department of Internal Medicine and Paediatrics, Ghent University, 9000 Ghent, Belgium; (C.D.G.); (M.D.V.); (P.H.); (D.L.)
- Ghent Gut Inflammation Group (GGIG), Ghent University, 9000 Ghent, Belgium
| | - Pieter Hindryckx
- Department of Internal Medicine and Paediatrics, Ghent University, 9000 Ghent, Belgium; (C.D.G.); (M.D.V.); (P.H.); (D.L.)
- Department of Gastroenterology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Debby Laukens
- Department of Internal Medicine and Paediatrics, Ghent University, 9000 Ghent, Belgium; (C.D.G.); (M.D.V.); (P.H.); (D.L.)
- Ghent Gut Inflammation Group (GGIG), Ghent University, 9000 Ghent, Belgium
- VIB Centre for Inflammation Research, 9000 Ghent, Belgium
| | - Sophie Van Welden
- Department of Internal Medicine and Paediatrics, Ghent University, 9000 Ghent, Belgium; (C.D.G.); (M.D.V.); (P.H.); (D.L.)
- Ghent Gut Inflammation Group (GGIG), Ghent University, 9000 Ghent, Belgium
- VIB Centre for Inflammation Research, 9000 Ghent, Belgium
- Correspondence: ; Tel.: +32-9-332-58-30
| |
Collapse
|
9
|
Malkov MI, Lee CT, Taylor CT. Regulation of the Hypoxia-Inducible Factor (HIF) by Pro-Inflammatory Cytokines. Cells 2021; 10:cells10092340. [PMID: 34571989 PMCID: PMC8466990 DOI: 10.3390/cells10092340] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/27/2021] [Accepted: 09/02/2021] [Indexed: 12/28/2022] Open
Abstract
Hypoxia and inflammation are frequently co-incidental features of the tissue microenvironment in a wide range of inflammatory diseases. While the impact of hypoxia on inflammatory pathways in immune cells has been well characterized, less is known about how inflammatory stimuli such as cytokines impact upon the canonical hypoxia-inducible factor (HIF) pathway, the master regulator of the cellular response to hypoxia. In this review, we discuss what is known about the impact of two major pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), on the regulation of HIF-dependent signaling at sites of inflammation. We report extensive evidence for these cytokines directly impacting upon HIF signaling through the regulation of HIF at transcriptional and post-translational levels. We conclude that multi-level crosstalk between inflammatory and hypoxic signaling pathways plays an important role in shaping the nature and degree of inflammation occurring at hypoxic sites.
Collapse
Affiliation(s)
- Mykyta I. Malkov
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland; (M.I.M.); (C.T.L.)
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Chee Teik Lee
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland; (M.I.M.); (C.T.L.)
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Cormac T. Taylor
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland; (M.I.M.); (C.T.L.)
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Correspondence:
| |
Collapse
|
10
|
Gojkovic M, Cunha PP, Darmasaputra GS, Barbieri L, Rundqvist H, Veliça P, Johnson RS. Oxygen-Mediated Suppression of CD8+ T Cell Proliferation by Macrophages: Role of Pharmacological Inhibitors of HIF Degradation. Front Immunol 2021; 12:633586. [PMID: 34054802 PMCID: PMC8153186 DOI: 10.3389/fimmu.2021.633586] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/22/2021] [Indexed: 12/28/2022] Open
Abstract
Myeloid cell interactions with cells of the adaptive immune system are an essential aspect of immunity. A key aspect of that interrelationship is its modulation by the microenvironment. Oxygen is known to influence myelosuppression of T cell activation in part via the Hypoxia inducible (HIF) transcription factors. A number of drugs that act on the HIF pathway are currently in clinical use and it is important to evaluate how they act on immune cell function as part of a better understanding of how they will influence patient outcomes. We show here that increased activation of the HIF pathway, either through deletion of the negative regulator of HIF, the von Hippel-Lindau (VHL) gene, in myeloid cells, or through pharmacological inhibitors of VHL-mediated degradation of HIF, potently suppresses T cell proliferation in myeloid cell/T cell culture. These data demonstrate that both pharmacological and genetic activation of HIF in myeloid cells can suppress adaptive cell immune response.
Collapse
Affiliation(s)
- Milos Gojkovic
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Pedro P. Cunha
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | | | - Laura Barbieri
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Helene Rundqvist
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Pedro Veliça
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Randall S. Johnson
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
11
|
Ghezzi P. Redox regulation of immunity and the role of small molecular weight thiols. Redox Biol 2021; 44:102001. [PMID: 33994345 PMCID: PMC8212150 DOI: 10.1016/j.redox.2021.102001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/31/2021] [Accepted: 05/02/2021] [Indexed: 01/07/2023] Open
Abstract
It is thought that excessive production of reactive oxygen species (ROS) can be a causal component in many diseases, some of which have an inflammatory component. This led to an oversimplification whereby ROS are seen as inflammatory and antioxidants anti-inflammatory. This paper aims at reviewing some of the literature on thiols in host defense. The review will first summarize the mechanisms by which we survive infections by pathogens. Then we will consider how the redox field evolved from the concept of oxidative stress to that of redox regulation and how it intersects the field of innate immunity. A third section will analyze how an oversimplified oxidative stress theory of disease led to a hypothesis on the role of ROS and glutathione (GSH) in immunity, respectively as pro- and anti-inflammatory mediators. Finally, we will discuss some recent research and how to think out of the box of that oversimplification and link the role of thiols in redox regulation to the mechanisms by which we survive an infection outlined in the first section.
Collapse
Affiliation(s)
- Pietro Ghezzi
- Brighton and Sussex Medical School, Brighton, Great Britain, BN1 9RY, UK.
| |
Collapse
|
12
|
The Prolyl Hydroxylase Inhibitor Dimethyl Oxalyl Glycine Decreases Early Gastrointestinal GVHD in Experimental Allogeneic Hematopoietic Cell Transplantation. Transplantation 2021; 104:2507-2515. [PMID: 32639407 DOI: 10.1097/tp.0000000000003383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Prolyl hydroxylase inhibitors (PHI) promote stabilization of hypoxia-inducible factor-1 alpha and affect signaling cascades of inflammation and cell death. Their beneficial use in experimental models of ulcerative colitis and lung allograft rejection led us to test the effect of the PHI dimethyl oxalyl glycine (DMOG) in the pathophysiology of graft versus host disease (GVHD). METHODS Acute GVHD was induced in lethally irradiated BALB/c mice. DMOG was administered intraperitoneally on alternate days for the first 2-weeks posttransplant, and then twice a week till day +50, while controls received vehicle only. Animals were monitored for clinical GVHD and analyzed at day +7 and at day +50. RESULTS DMOG treatment of allogeneic recipients improved survival by day +50, which was associated with decreased early gut injury and serum tumor necrosis factor-α compared with allogeneic controls. DMOG treatment of allogeneic recipients resulted in increased hypoxia-inducible factor-1 alpha expression and reduced apoptosis in the terminal ileum via Fas-associated protein with death domain protein repression along with decreased T-cell infiltration. Reduced pathology in colon after DMOG treatment associates with intestinal epithelium integrity and reduced damage caused by diminished recruitment of neutrophils. CONCLUSIONS Taken together, we show protective effects of DMOG on early gut GVHD and improved survival in a model of allogeneic hematopoietic cell transplantation, providing the rationale for further evaluation of PHIs, in the prevention and treatment of acute GVHD.
Collapse
|
13
|
Abstract
Recent years have witnessed an emergence of interest in understanding metabolic changes associated with immune responses, termed immunometabolism. As oxygen is central to all aerobic metabolism, hypoxia is now recognized to contribute fundamentally to inflammatory and immune responses. Studies from a number of groups have implicated a prominent role for oxygen metabolism and hypoxia in innate immunity of healthy tissue (physiologic hypoxia) and during active inflammation (inflammatory hypoxia). This inflammatory hypoxia emanates from a combination of recruited inflammatory cells (e.g., neutrophils, eosinophils, and monocytes), high rates of oxidative metabolism, and the activation of multiple oxygen-consuming enzymes during inflammation. These localized shifts toward hypoxia have identified a prominent role for the transcription factor hypoxia-inducible factor (HIF) in the regulation of innate immunity. Such studies have provided new and enlightening insight into our basic understanding of immune mechanisms, and extensions of these findings have identified potential therapeutic targets. In this review, we summarize recent literature around the topic of innate immunity and mucosal hypoxia with a focus on transcriptional responses mediated by HIF.
Collapse
Affiliation(s)
- Sean P Colgan
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, USA;
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Glenn T Furuta
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Cormac T Taylor
- UCD Conway Institute, Systems Biology Ireland and School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
14
|
Harnoss JM, Gebhardt JM, Radhakrishnan P, Leowardi C, Burmeister J, Halligan DN, Yuan S, Kennel KB, Strowitzki MJ, Schaible A, Lasitschka F, Taylor CT, Schneider M. Prolyl Hydroxylase Inhibition Mitigates Pouchitis. Inflamm Bowel Dis 2020; 26:192-205. [PMID: 31618435 DOI: 10.1093/ibd/izz218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Pouchitis is the most common long-term complication after restorative proctocolectomy with ileal pouch-anal anastomosis (IPAA) for ulcerative colitis (UC) or familial adenomatous polyposis (FAP), which can eventually progress to pouch failure, necessitating permanent stoma construction. Hypoxia-inducible transcription factor prolyl hydroxylase-containing enzymes (PHD1, PHD2, and PHD3) are molecular oxygen sensors that control adaptive gene expression through hypoxia-inducible factor (HIF). Emerging evidence supports PHDs as being therapeutic targets in intestinal inflammation. However, pharmacological inhibition of PHDs has not been validated as a treatment strategy in pouchitis. METHODS PHD1-3 mRNA and protein expression were analyzed in mucosal pouch and prepouch ileal patient biopsies. After establishment of a preclinical IPAA model in rats, the impact of the pan-PHD small-molecule inhibitor dimethyloxalylglycine (DMOG) on dextran sulfate sodium (DSS)-induced pouchitis was studied. Clinical and molecular parameters were investigated. RESULTS PHD1, but not PHD2 or PHD3, was overexpressed in pouchitis in biopsies of patients with IPAA for UC but not FAP. In addition, PHD1 expression correlated with disease activity. DMOG treatment profoundly mitigated DSS-induced pouchitis in a rodent IPAA model. Mechanistically, DMOG restored intestinal epithelial barrier function by induction of tight junction proteins zona occludens-1 and claudin-1 and alleviation of intestinal epithelial cell apoptosis, thus attenuating pouch inflammation. CONCLUSIONS Together, these results establish a strong therapeutic rationale for targeting PHD1 with small-molecule inhibitors in pouchitis after IPAA for UC.
Collapse
Affiliation(s)
- Jonathan M Harnoss
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Jasper M Gebhardt
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Praveen Radhakrishnan
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Christine Leowardi
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Julius Burmeister
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Doug N Halligan
- School of Medicine, Systems Biology Ireland and the Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Shuai Yuan
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Kilian B Kennel
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Moritz J Strowitzki
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany.,School of Medicine, Systems Biology Ireland and the Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Anja Schaible
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Felix Lasitschka
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Cormac T Taylor
- School of Medicine, Systems Biology Ireland and the Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
15
|
Hypoxia inhibits TNF-α-induced TSLP expression in keratinocytes. PLoS One 2019; 14:e0224705. [PMID: 31682627 PMCID: PMC6827910 DOI: 10.1371/journal.pone.0224705] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/18/2019] [Indexed: 12/20/2022] Open
Abstract
The expression of thymic stromal lymphopoietin (TSLP), a cytokine which greatly contributes to the induction of type I allergy, is upregulated in chronic inflammation such as atopic dermatitis and psoriasis. As hypoxia in the epidermis is important for maintaining skin homeostasis, we examined the regulation of TSLP expression by hypoxic conditions in normal skin epithelial tissues. TNF-α-induced expression of TSLP in human keratinocyte HaCaT and in mouse keratinocyte PAM212 cell lines were inhibited under hypoxic condition (1% O2), although the mRNA expressions of TNF-α, IL-6, IL-8, MCP-1, and VEGF-A were not inhibited. Hypoxia-mimicking conditions, which include NiCl2, CoCl2, and DMOG, an inhibitor of 2-oxoglutarate-dependent enzymes, also selectively inhibited TNF-α-induced TSLP expression. These results suggested that inactivation of prolyl hydroxylase by hypoxia and hypoxia-mimicking conditions is involved in the repression of TNF-α-induced TSLP expression. Interestingly, the inhibition of TSLP production by hypoxic treatment was significantly reversed by treatment with the HIF-2α antagonist but not with the HIF-1α inhibitor. DMOG-induced inhibition of TSLP promoter activity was dependent on the -71 to +185 bp promoter region, suggesting that the binding of HIF-2 to hypoxia response element (HRE) in this region repressed the TSLP expression. These results indicated that hypoxia and hypoxia-mimicking conditions inhibited TSLP expression via HIF-2 and HRE-dependent mechanisms. Therefore, PHD and HIF-2α could be a new strategy for treatment of atopic dermatitis and psoriasis.
Collapse
|
16
|
FADD in Cancer: Mechanisms of Altered Expression and Function, and Clinical Implications. Cancers (Basel) 2019; 11:cancers11101462. [PMID: 31569512 PMCID: PMC6826683 DOI: 10.3390/cancers11101462] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/25/2019] [Accepted: 09/27/2019] [Indexed: 12/15/2022] Open
Abstract
FADD was initially described as an adaptor molecule for death receptor-mediated apoptosis, but subsequently it has been implicated in nonapoptotic cellular processes such as proliferation and cell cycle control. During the last decade, FADD has been shown to play a pivotal role in most of the signalosome complexes, such as the necroptosome and the inflammasome. Interestingly, various mechanisms involved in regulating FADD functions have been identified, essentially posttranslational modifications and secretion. All these aspects have been thoroughly addressed in previous reviews. However, FADD implication in cancer is complex, due to pleiotropic effects. It has been reported either as anti- or protumorigenic, depending on the cell type. Regulation of FADD expression in cancer is a complex issue since both overexpression and downregulation have been reported, but the mechanisms underlying such alterations have not been fully unveiled. Posttranslational modifications also constitute a relevant mechanism controlling FADD levels and functions in tumor cells. In this review, we aim to provide detailed, updated information on alterations leading to changes in FADD expression and function in cancer. The participation of FADD in various biological processes is recapitulated, with a mention of interesting novel functions recently proposed for FADD, such as regulation of gene expression and control of metabolic pathways. Finally, we gather all the available evidence regarding the clinical implications of FADD alterations in cancer, especially as it has been proposed as a potential biomarker with prognostic value.
Collapse
|
17
|
Halligan DN, Khan MN, Brown E, Rowan CR, Coulter IS, Doherty GA, Tambuwala MM, Taylor CT. Hypoxia-inducible factor hydroxylase inhibition enhances the protective effects of cyclosporine in colitis. Am J Physiol Gastrointest Liver Physiol 2019; 317:G90-G97. [PMID: 31070931 DOI: 10.1152/ajpgi.00049.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Inflammatory bowel disease (IBD) is characterized by epithelial barrier dysfunction with resultant inflammation as the mucosal immune system becomes exposed to luminal antigens. The hydroxylase inhibitor dimethyloxalylglycine (DMOG) reduces symptoms in experimental colitis through the upregulation of genes promoting barrier function and inhibition of epithelial cell apoptosis. The immunosuppressive drug cyclosporine reduces inflammation associated with IBD via suppression of immune cell activation. Given the distinct barrier protective effect of DMOG and the anti-inflammatory properties of cyclosporine, we hypothesized that combining these drugs may provide an enhanced protective effect by targeting both barrier dysfunction and inflammation simultaneously. We used the dextran sulfate sodium model of colitis in C57BL/6 mice to determine the combinatorial efficacy of cyclosporine and DMOG. While cyclosporine and DMOG ameliorated disease progression, in combination they had an additive protective effect that surpassed the level of protection afforded by either drug alone. The ability of DMOG to augment the anti-inflammatory effects of cyclosporine was largely due to preservation of barrier function and at least in part due to zonula occludens-1 regulation. We propose that combining the barrier protective effects of a hydroxylase inhibitor with the anti-inflammatory effects of cyclosporine provides added therapeutic benefit in colitis.NEW & NOTEWORTHY Inflammatory bowel disease is the result of decreased intestinal epithelial barrier function leading to exposure of the mucosal immune system to luminal antigens causing inflammation, which in turn further decreases epithelial barrier function. We demonstrate for the first time that strengthening the epithelial barrier with a hydroxylase inhibitor in combination with the administration of the immunosuppressive cyclosporine provides additive therapeutic advantage in a murine model of colitis.
Collapse
Affiliation(s)
- Doug N Halligan
- The Conway Institute, University College Dublin, Belfield, Dublin, Ireland.,Sigmoid Pharma, The Invent Centre, Dublin City University, Dublin, Ireland
| | - Mohammed N Khan
- The Saad Centre for Pharmacy and Diabetes, Ulster University, Coleraine, United Kingdom
| | - Eric Brown
- The Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Catherine R Rowan
- Centre for Colorectal Disease, St Vincent's University Hospital and School of Medicine, University College Dublin, Dublin, Ireland
| | - Ivan S Coulter
- Sigmoid Pharma, The Invent Centre, Dublin City University, Dublin, Ireland
| | - Glen A Doherty
- Centre for Colorectal Disease, St Vincent's University Hospital and School of Medicine, University College Dublin, Dublin, Ireland
| | - Murtaza M Tambuwala
- The Saad Centre for Pharmacy and Diabetes, Ulster University, Coleraine, United Kingdom
| | - Cormac T Taylor
- The Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
18
|
Schley G, Klanke B, Kalucka J, Schatz V, Daniel C, Mayer M, Goppelt-Struebe M, Herrmann M, Thorsteinsdottir M, Palsson R, Beneke A, Katschinski DM, Burzlaff N, Eckardt KU, Weidemann A, Jantsch J, Willam C. Mononuclear phagocytes orchestrate prolyl hydroxylase inhibition-mediated renoprotection in chronic tubulointerstitial nephritis. Kidney Int 2019; 96:378-396. [DOI: 10.1016/j.kint.2019.02.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 01/14/2019] [Accepted: 02/14/2019] [Indexed: 12/22/2022]
|
19
|
Manresa MC, Smith L, Casals‐Diaz L, Fagundes RR, Brown E, Radhakrishnan P, Murphy SJ, Crifo B, Strowitzki MJ, Halligan DN, van den Bogaard EH, Niehues H, Schneider M, Taylor CT, Steinhoff M. Pharmacologic inhibition of hypoxia-inducible factor (HIF)-hydroxylases ameliorates allergic contact dermatitis. Allergy 2019; 74:753-766. [PMID: 30394557 DOI: 10.1111/all.13655] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 10/05/2018] [Accepted: 10/16/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND When an immune cell migrates from the bloodstream to a site of chronic inflammation, it experiences a profound decrease in microenvironmental oxygen levels leading to a state of cellular hypoxia. The hypoxia-inducible factor-1α (HIF-1α) promotes an adaptive transcriptional response to hypoxia and as such is a major regulator of immune cell survival and function. HIF hydroxylases are the family of oxygen-sensing enzymes primarily responsible for conferring oxygen dependence upon the HIF pathway. METHODS Using a mouse model of allergic contact dermatitis (ACD), we tested the effects of treatment with the pharmacologic hydroxylase inhibitor DMOG, which mimics hypoxia, on disease development. RESULTS Re-exposure of sensitized mice to 2,4-dinitrofluorobenzene (DNFB) elicited inflammation, edema, chemokine synthesis (including CXCL1 and CCL5) and the recruitment of neutrophils and eosinophils. Intraperitoneal or topical application of the pharmacologic hydroxylase inhibitors dymethyloxalylglycine (DMOG) or JNJ1935 attenuated this inflammatory response. Reduced inflammation was associated with diminished recruitment of neutrophils and eosinophils but not lymphocytes. Finally, hydroxylase inhibition reduced cytokine-induced chemokine production in cultured primary keratinocytes through attenuation of the JNK pathway. CONCLUSION These data demonstrate that hydroxylase inhibition attenuates the recruitment of neutrophils to inflamed skin through reduction of chemokine production and increased neutrophilic apoptosis. Thus, pharmacologic inhibition of HIF hydroxylases may be an effective new therapeutic approach in allergic skin inflammation.
Collapse
Affiliation(s)
- Mario C. Manresa
- UCD Charles Institute of Dermatology School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
- Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| | - Leila Smith
- UCD Charles Institute of Dermatology School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Laura Casals‐Diaz
- UCD Charles Institute of Dermatology School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Raphael R. Fagundes
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Eric Brown
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Praveen Radhakrishnan
- Department of General, Visceral and Transplantation Surgery University of Heidelberg Heidelberg Germany
| | - Stephen J. Murphy
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Bianca Crifo
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Moritz J. Strowitzki
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Doug N. Halligan
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Ellen H. van den Bogaard
- Department of Dermatology Radboud University Medical Center Radboud Institute for Molecular Life Sciences Nijmegen The Netherlands
| | - Hanna Niehues
- Department of Dermatology Radboud University Medical Center Radboud Institute for Molecular Life Sciences Nijmegen The Netherlands
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery University of Heidelberg Heidelberg Germany
| | - Cormac T. Taylor
- UCD Charles Institute of Dermatology School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
- Systems Biology Ireland School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Martin Steinhoff
- UCD Charles Institute of Dermatology School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
- Department of Dermatology & Venereology Translational Research Institute Hamad Medical Corporation Weill Cornell University‐Qatar and Qatar University Doha Qatar
| |
Collapse
|
20
|
Shen W, Wang Q, Shen Y, Gao X, Li L, Yan Y, Wang H, Cheng Y. Green Tea Catechin Dramatically Promotes RNAi Mediated by Low-Molecular-Weight Polymers. ACS CENTRAL SCIENCE 2018; 4:1326-1333. [PMID: 30410970 PMCID: PMC6202644 DOI: 10.1021/acscentsci.8b00363] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Indexed: 05/23/2023]
Abstract
Cytosolic delivery is the major challenge that limits the clinical translation of siRNA-based therapeutics. Although thousands of polymers have been developed for siRNA delivery, the efficiency-toxicity correlation is unsatisfactory. Here, we report a facile strategy to fabricate core-shell-structured nanoparticles with robust siRNA delivery efficiency. The nanoparticle is prepared by entropy-driven complexation of siRNA with a green tea catechin to yield a negatively charged core, followed by coating low-molecular-weight polymers to form the shell. This supramolecular strategy facilitates the polymers condensing siRNA into uniform nanoparticles. The nanoparticle specifically down-regulates target genes in vitro and in vivo, and efficiently attenuates chronic intestinal inflammation in an inflammatory bowel disease model. Notably, the highly efficient nanoparticles are applicable for various polymers with different topologies and chemical compositions, providing a versatile technique to break down the efficiency-toxicity correlation of cationic polymers. The proposed strategy in this study permits the development of a promising platform for polymer-mediated siRNA delivery.
Collapse
Affiliation(s)
- Wanwan Shen
- Shanghai
Key Laboratory of Regulatory Biology, East
China Normal University, Shanghai 200241, China
| | - Qingwei Wang
- Shanghai
Key Laboratory of Regulatory Biology, East
China Normal University, Shanghai 200241, China
| | - Yang Shen
- Shanghai
Key Laboratory of Regulatory Biology, East
China Normal University, Shanghai 200241, China
| | - Xiao Gao
- Shanghai
Key Laboratory of Regulatory Biology, East
China Normal University, Shanghai 200241, China
| | - Lei Li
- Shanghai
Key Laboratory of Regulatory Biology, East
China Normal University, Shanghai 200241, China
| | - Yang Yan
- Shanghai
Key Laboratory of Regulatory Biology, East
China Normal University, Shanghai 200241, China
| | - Hui Wang
- South
China Advanced Institute for Soft Matter Science and Technology, South China University of Technology, Guangzhou 510640, China
| | - Yiyun Cheng
- Shanghai
Key Laboratory of Regulatory Biology, East
China Normal University, Shanghai 200241, China
- South
China Advanced Institute for Soft Matter Science and Technology, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
21
|
Fricker M, Goggins BJ, Mateer S, Jones B, Kim RY, Gellatly SL, Jarnicki AG, Powell N, Oliver BG, Radford-Smith G, Talley NJ, Walker MM, Keely S, Hansbro PM. Chronic cigarette smoke exposure induces systemic hypoxia that drives intestinal dysfunction. JCI Insight 2018; 3:94040. [PMID: 29415878 PMCID: PMC5821186 DOI: 10.1172/jci.insight.94040] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 01/10/2018] [Indexed: 01/05/2023] Open
Abstract
Crohn's disease (CD) is a chronic inflammatory disease of the gastrointestinal tract (GIT). Cigarette smoke (CS) exposure and chronic obstructive pulmonary disease (COPD) are risk factors for CD, although the mechanisms involved are poorly understood. We employed a mouse model of CS-induced experimental COPD and clinical studies to examine these mechanisms. Concurrent with the development of pulmonary pathology and impaired gas exchange, CS-exposed mice developed CD-associated pathology in the colon and ileum, including gut mucosal tissue hypoxia, HIF-2 stabilization, inflammation, increased microvasculature, epithelial cell turnover, and decreased intestinal barrier function. Subsequent smoking cessation reduced GIT pathology, particularly in the ileum. Dimethyloxaloylglycine, a pan-prolyl hydroxylase inhibitor, ameliorated CS-induced GIT pathology independently of pulmonary pathology. Prior smoke exposure exacerbated intestinal pathology in 2,4,6-trinitrobenzenesulfonic acid-induced (TNBS-induced) colitis. Circulating vascular endothelial growth factor, a marker of systemic hypoxia, correlated with CS exposure and CD in mice and humans. Increased mucosal vascularisation was evident in ileum biopsies from CD patients who smoke compared with nonsmokers, supporting our preclinical data. We provide strong evidence that chronic CS exposure and, for the first time to our knowledge, associated impaired gas exchange cause systemic and intestinal ischemia, driving angiogenesis and GIT epithelial barrier dysfunction, resulting in increased risk and severity of CD.
Collapse
Affiliation(s)
- Michael Fricker
- Priority research Centre for Healthy Lungs, University of Newcastle and
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Bridie J. Goggins
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Sean Mateer
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Bernadette Jones
- Priority research Centre for Healthy Lungs, University of Newcastle and
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Richard Y. Kim
- Priority research Centre for Healthy Lungs, University of Newcastle and
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Shaan L. Gellatly
- Priority research Centre for Healthy Lungs, University of Newcastle and
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Andrew G. Jarnicki
- Priority research Centre for Healthy Lungs, University of Newcastle and
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Nicholas Powell
- Faculty of Translational Medicine, Guy’s and St. Thomas’ and King’s College London Comprehensive Biomedical Research Centre, Great Maze Pond, London, United Kingdom
| | - Brian G. Oliver
- Woolcock Institute of Medical Research, The University of Sydney, Sydney, New South Wales, Australia
- School of Life Sciences, The University of Technology, Sydney, New South Wales, Australia
| | - Graham Radford-Smith
- Royal Brisbane and Women’s Hospital, Brisbane, School of Medicine, University of Queensland, and
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Nicholas J. Talley
- Priority Research Centre for Digestive Health and Neurogastroenterology, University of Newcastle, New South Wales, Australia
| | - Marjorie M. Walker
- Priority Research Centre for Digestive Health and Neurogastroenterology, University of Newcastle, New South Wales, Australia
| | - Simon Keely
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, University of Newcastle, New South Wales, Australia
| | - Philip M. Hansbro
- Priority research Centre for Healthy Lungs, University of Newcastle and
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, University of Newcastle, New South Wales, Australia
| |
Collapse
|
22
|
Kim YE, Lee M, Gu H, Kim J, Jeong S, Yeo S, Lee YJ, Im SH, Sung YC, Kim HJ, Weissman IL, Ahn GO. Hypoxia-inducible factor-1 (HIF-1) activation in myeloid cells accelerates DSS-induced colitis progression in mice. Dis Model Mech 2018; 11:dmm.033241. [PMID: 29967068 PMCID: PMC6078398 DOI: 10.1242/dmm.033241] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 06/25/2018] [Indexed: 12/26/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease, in which the intestinal epithelium loses its barrier function. Given the existence of the oxygen gradient in the intestinal epithelium and that inflammation further contributes to the tissue hypoxia, we investigated the role of hypoxia-inducible factor (HIF), a transcription factor activated under hypoxic conditions in myeloid cells, in the progression of IBD. To do this, we utilized myeloid-specific knockout (KO) mice targeting HIF pathways, created by a Cre-loxP system with human MRP8 (hMRP8), an intracellular calcium-binding protein, as the myeloid promoter. By feeding 5% dextran sodium sulfate (DSS) to hMRP8 von Hippel Lindau (Vhl) KO mice, in which HIF-1α and HIF-2α are constitutively activated in myeloid cells, we found that these mice were highly susceptible to DSS-induced colitis, demonstrating greater body weight loss, increased mortality, faster onset of rectal bleeding, shortened colon length, and increased CD11b- or Gr-1-positive myeloid cells in the colon compared with wild-type (WT) mice. These parameters were restored to, if not better than, the WT levels when we examined hMRP8 Hif-1a KO mice upon 5% DSS feeding. hMRP8 Hif-2a KO mice, on the other hand, exhibited a similar degree of DSS-induced colitis to that of WT mice. Lastly, when DSS was given together with azoxymethane to induce tumorigenesis in the colon, we found that hMRP8 Hif-1a KO mice exhibited comparable levels of colorectal tumors to those of WT mice, indicating that HIF-1α in myeloid cells is dispensable for tumorigenesis. Collectively, our results suggest that HIF-1α activation in myeloid cells critically regulates IBD progression. Summary: We challenged myeloid-specific knockout mice targeting the hypoxia-inducible factor (HIF) pathway to dextran sodium sulfate-induced colitis, demonstrating that HIF-1α, but not HIF-2α, activation in myeloid cells regulates colitis severity in mice.
Collapse
Affiliation(s)
- Young-Eun Kim
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
| | - Minji Lee
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, Gyeongbuk 37673, Korea
| | - Hyejung Gu
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
| | - Jeongwoo Kim
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
| | - Seongju Jeong
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
| | - Sujin Yeo
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
| | - You Jeong Lee
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, Gyeongbuk 37673, Korea
| | - Sin-Hyeog Im
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, Gyeongbuk 37673, Korea
| | - Young-Chul Sung
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
| | - Hak Jae Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, 03080, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Irving L. Weissman
- Stem Cell Institute and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - G-One Ahn
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), 77 Cheong Am-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Korea
| |
Collapse
|
23
|
Taylor CT, Colgan SP. Regulation of immunity and inflammation by hypoxia in immunological niches. Nat Rev Immunol 2017; 17:774-785. [PMID: 28972206 PMCID: PMC5799081 DOI: 10.1038/nri.2017.103] [Citation(s) in RCA: 454] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immunological niches are focal sites of immune activity that can have varying microenvironmental features. Hypoxia is a feature of physiological and pathological immunological niches. The impact of hypoxia on immunity and inflammation can vary depending on the microenvironment and immune processes occurring in a given niche. In physiological immunological niches, such as the bone marrow, lymphoid tissue, placenta and intestinal mucosa, physiological hypoxia controls innate and adaptive immunity by modulating immune cell proliferation, development and effector function, largely via transcriptional changes driven by hypoxia-inducible factor (HIF). By contrast, in pathological immunological niches, such as tumours and chronically inflamed, infected or ischaemic tissues, pathological hypoxia can drive tissue dysfunction and disease development through immune cell dysregulation. Here, we differentiate between the effects of physiological and pathological hypoxia on immune cells and the consequences for immunity and inflammation in different immunological niches. Furthermore, we discuss the possibility of targeting hypoxia-sensitive pathways in immune cells for the treatment of inflammatory disease.
Collapse
Affiliation(s)
- Cormac T Taylor
- UCD Conway Institute, Systems Biology Ireland and the School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sean P Colgan
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, 80045 Colorado, USA
| |
Collapse
|
24
|
Brown E, Taylor CT. Hypoxia-sensitive pathways in intestinal inflammation. J Physiol 2017; 596:2985-2989. [PMID: 29114885 DOI: 10.1113/jp274350] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 10/24/2017] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a common chronic intestinal disorder characterised by a loss of epithelial barrier function leading to the unregulated movement of luminal antigenic material into mucosal tissue with resultant inflammation. In IBD, multiple components of the inflammatory response lead to tissue hypoxia. Mucosal hypoxia leads to the inactivation of prolyl hydroxylase domain-containing (PHD) enzymes, which in turn leads to the stabilisation of the hypoxia-inducible factor (HIF), which induces the expression of barrier protective genes. Furthermore, pharmacological hydroxylase inhibition has been shown to be protective in colitis, at least in part through enhancing intestinal epithelial barrier function through HIF-1-dependent barrier-protective gene expression. Therefore, targeting hypoxia-sensitive pathways represents a new and promising therapeutic approach in IBD.
Collapse
Affiliation(s)
- Eric Brown
- UCD Conway Institute & School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Cormac T Taylor
- UCD Conway Institute & School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
25
|
Van Welden S, Selfridge AC, Hindryckx P. Intestinal hypoxia and hypoxia-induced signalling as therapeutic targets for IBD. Nat Rev Gastroenterol Hepatol 2017; 14:596-611. [PMID: 28853446 DOI: 10.1038/nrgastro.2017.101] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tissue hypoxia occurs when local oxygen demand exceeds oxygen supply. In chronic inflammatory conditions such as IBD, the increased oxygen demand by resident and gut-infiltrating immune cells coupled with vascular dysfunction brings about a marked reduction in mucosal oxygen concentrations. To counter the hypoxic challenge and ensure their survival, mucosal cells induce adaptive responses, including the activation of hypoxia-inducible factors (HIFs) and modulation of nuclear factor-κB (NF-κB). Both pathways are tightly regulated by oxygen-sensitive prolyl hydroxylases (PHDs), which therefore represent promising therapeutic targets for IBD. In this Review, we discuss the involvement of mucosal hypoxia and hypoxia-induced signalling in the pathogenesis of IBD and elaborate in detail on the role of HIFs, NF-κB and PHDs in different cell types during intestinal inflammation. We also provide an update on the development of PHD inhibitors and discuss their therapeutic potential in IBD.
Collapse
Affiliation(s)
- Sophie Van Welden
- Department of Gastroenterology, Ghent University, De Pintelaan 185, 1K12-E, 9000 Ghent, Belgium
| | - Andrew C Selfridge
- Robarts Clinical Trials West, 4350 Executive Drive 210, San Diego, California 92121, USA
| | - Pieter Hindryckx
- Department of Gastroenterology, Ghent University, De Pintelaan 185, 1K12-E, 9000 Ghent, Belgium
| |
Collapse
|
26
|
Nagamine Y, Tojo K, Yazawa T, Takaki S, Baba Y, Goto T, Kurahashi K. Inhibition of Prolyl Hydroxylase Attenuates Fas Ligand-Induced Apoptosis and Lung Injury in Mice. Am J Respir Cell Mol Biol 2017; 55:878-888. [PMID: 27494234 DOI: 10.1165/rcmb.2015-0266oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Alveolar epithelial injury and increased alveolar permeability are hallmarks of acute respiratory distress syndrome. Apoptosis of lung epithelial cells via the Fas/Fas ligand (FasL) pathway plays a critical role in alveolar epithelial injury. Activation of hypoxia-inducible factor (HIF)-1 by inhibition of prolyl hydroxylase domain proteins (PHDs) is a possible therapeutic approach to attenuate apoptosis and organ injury. Here, we investigated whether treatment with dimethyloxalylglycine (DMOG), an inhibitor of PHDs, could attenuate Fas/FasL-dependent apoptosis in lung epithelial cells and lung injury. DMOG increased HIF-1α protein expression in vitro in MLE-12 cells, a murine alveolar epithelial cell line. Treatment of MLE-12 cells with DMOG significantly suppressed cell surface expression of Fas and attenuated FasL-induced caspase-3 activation and apoptotic cell death. Inhibition of the HIF-1 pathway by echinomycin or small interfering RNA transfection abolished these antiapoptotic effects of DMOG. Moreover, intraperitoneal injection of DMOG in mice increased HIF-1α expression and decreased Fas expression in lung tissues. DMOG treatment significantly attenuated caspase-3 activation, apoptotic cell death in lung tissue, and the increase in alveolar permeability in mice instilled intratracheally with FasL. In addition, inflammatory responses and histopathological changes were also significantly attenuated by DMOG treatment. In conclusion, inhibition of PHDs protects lung epithelial cells from Fas/FasL-dependent apoptosis through HIF-1 activation and attenuates lung injury in mice.
Collapse
Affiliation(s)
- Yusuke Nagamine
- 1 Department of Anesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Kentaro Tojo
- 1 Department of Anesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Takuya Yazawa
- 2 Department of Diagnostic Pathology, Chiba University Graduate School of Medicine, Chiba, Chiba, Japan; and
| | - Shunsuke Takaki
- 1 Department of Anesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Yasuko Baba
- 3 Operation Department, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Takahisa Goto
- 1 Department of Anesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Kiyoyasu Kurahashi
- 1 Department of Anesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| |
Collapse
|
27
|
Yin J, Ni B, Liao WG, Gao YQ. Hypoxia-induced apoptosis of mouse spermatocytes is mediated by HIF-1α through a death receptor pathway and a mitochondrial pathway. J Cell Physiol 2017; 233:1146-1155. [PMID: 28444885 DOI: 10.1002/jcp.25974] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 04/24/2017] [Indexed: 12/19/2022]
Abstract
Hypoxia in vivo induces oligozoospermia, azoospermia, and degeneration of the germinal epithelium, but the underlying molecular mechanism of this induction is not fully clarified. The aim of this study was to investigate the role of the death receptor pathway and the mitochondrial pathway in hypoxia-induced apoptosis of mouse GC-2spd (GC-2) cells and the relationship between HIF-1α and apoptosis of GC-2 cells induced by hypoxia. GC-2 cells were subjected to 1% oxygen for 48 hr. Apoptosis was detected by flow cytometry, TUNEL staining, LDH, caspase-3/8/9 in the absence and presence of HIF-1α siRNA. The protein levels of apoptosis-related markers were determined by Western blot in the presence and absence of HIF-1α siRNA. Mitochondrial transmembrane potential change was observed by in situ JC-1 staining. Cell viability was assessed upon treatment of caspase-8 and 9 inhibitors. The results indicated that hypoxia at 1% oxygen for 48 hr induced apoptosis of GC-2 cells. A prolonged exposure of GC-2 cells to hypoxic conditions caused downregulation of c-FLIP, Dc R2 and Bcl-2 and upregulation of DR5 , TRAIL, Fas, p53, and Bax, with an overproduction of caspase-3/8/9. Moreover, hypoxia at this level had an effect on mitochondrial depolarization. In addition, specific inhibitors of caspase-8/9 partially suppressed hypoxia-induced GC-2 cell apoptosis, and the anti-apoptotic effects of the caspase inhibitors were additive. Of note, HIF-1α knockdown attenuated hypoxia and induced apoptosis of GC-2 cells. In conclusion, our data suggest that the death receptor pathway and mitochondrial pathway, which are likely mediated by HIF-1α, contribute to hypoxia-induced GC-2 cell apoptosis.
Collapse
Affiliation(s)
- Jun Yin
- Department of Pathophysiology and High Altitude Pathology/Key Laboratory of High Altitude Environment Medicine (Third Military Medical University), Ministry of Education/Key Laboratory of High Altitude Medicine, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, PR China
| | - Bing Ni
- Department of Pathophysiology and High Altitude Pathology/Key Laboratory of High Altitude Environment Medicine (Third Military Medical University), Ministry of Education/Key Laboratory of High Altitude Medicine, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, PR China
| | - Wei-Gong Liao
- Department of Pathophysiology and High Altitude Pathology/Key Laboratory of High Altitude Environment Medicine (Third Military Medical University), Ministry of Education/Key Laboratory of High Altitude Medicine, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, PR China
| | - Yu-Qi Gao
- Institute of Medicine and Hygienic Equipment for High Altitude Region/Key Laboratory of High Altitude Environment Medicine (Third Military Medical University), Ministry of Education/Key Laboratory of High Altitude Medicine, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, PR China
| |
Collapse
|
28
|
Manresa MC, Taylor CT. Hypoxia Inducible Factor (HIF) Hydroxylases as Regulators of Intestinal Epithelial Barrier Function. Cell Mol Gastroenterol Hepatol 2017; 3:303-315. [PMID: 28462372 PMCID: PMC5404106 DOI: 10.1016/j.jcmgh.2017.02.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 02/09/2017] [Indexed: 12/12/2022]
Abstract
Human health is dependent on the ability of the body to extract nutrients, fluids, and oxygen from the external environment while at the same time maintaining a state of internal sterility. Therefore, the cell layers that cover the surface areas of the body such as the lung, skin, and gastrointestinal mucosa provide vital semipermeable barriers that allow the transport of essential nutrients, fluid, and waste products, while at the same time keeping the internal compartments free of microbial organisms. These epithelial surfaces are highly specialized and differ in their anatomic structure depending on their location to provide appropriate and effective site-specific barrier function. Given this important role, it is not surprising that significant disease often is associated with alterations in epithelial barrier function. Examples of such diseases include inflammatory bowel disease, chronic obstructive pulmonary disease, and atopic dermatitis. These chronic inflammatory disorders often are characterized by diminished tissue oxygen levels (hypoxia). Hypoxia triggers an adaptive transcriptional response governed by hypoxia-inducible factors (HIFs), which are repressed by a family of oxygen-sensing HIF hydroxylases. Here, we review recent evidence suggesting that pharmacologic hydroxylase inhibition may be of therapeutic benefit in inflammatory bowel disease through the promotion of intestinal epithelial barrier function through both HIF-dependent and HIF-independent mechanisms.
Collapse
Key Words
- CD, Crohn’s disease
- DMOG, dimethyloxalylglycine
- DSS, dextran sodium sulfate
- Epithelial Barrier
- FIH, factor inhibiting hypoxia-inducible factor
- HIF, hypoxia-inducible factor
- Hypoxia
- Hypoxia-Inducible Factor (HIF) Hydroxylases
- IBD, inflammatory bowel disease
- IL, interleukin
- Inflammatory Bowel Disease
- NF-κB, nuclear factor-κB
- PHD, hypoxia-inducible factor–prolyl hydroxylases
- TFF, trefoil factor
- TJ, tight junction
- TLR, Toll-like receptor
- TNF-α, tumor necrosis factor α
- UC, ulcerative colitis
- ZO, zonula occludens
Collapse
Affiliation(s)
- Mario C. Manresa
- Conway Institute of Biomolecular and Biomedical Research, Belfield, Dublin, Ireland
- Charles Institute of Dermatology, Belfield, Dublin, Ireland
| | - Cormac T. Taylor
- Conway Institute of Biomolecular and Biomedical Research, Belfield, Dublin, Ireland
- Charles Institute of Dermatology, Belfield, Dublin, Ireland
- Systems Biology Ireland, School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
29
|
Van Welden S, De Vos M, Wielockx B, Tavernier SJ, Dullaers M, Neyt S, Descamps B, Devisscher L, Devriese S, Van den Bossche L, Holvoet T, Baeyens A, Correale C, D'Alessio S, Vanhove C, De Vos F, Verhasselt B, Breier G, Lambrecht BN, Janssens S, Carmeliet P, Danese S, Elewaut D, Laukens D, Hindryckx P. Haematopoietic prolyl hydroxylase-1 deficiency promotes M2 macrophage polarization and is both necessary and sufficient to protect against experimental colitis. J Pathol 2017; 241:547-558. [PMID: 27981571 DOI: 10.1002/path.4861] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 11/21/2016] [Accepted: 12/06/2016] [Indexed: 12/17/2022]
Abstract
Prolyl hydroxylase domain-containing proteins (PHDs) regulate the adaptation of cells to hypoxia. Pan-hydroxylase inhibition is protective in experimental colitis, in which PHD1 plays a prominent role. However, it is currently unknown how PHD1 targeting regulates this protection and which cell type(s) are involved. Here, we demonstrated that Phd1 deletion in endothelial and haematopoietic cells (Phd1f/f Tie2:cre) protected mice from dextran sulphate sodium (DSS)-induced colitis, with reduced epithelial erosions, immune cell infiltration, and colonic microvascular dysfunction, whereas the response of Phd2f/+ Tie2:cre and Phd3f/f Tie2:cre mice to DSS was similar to that of their littermate controls. Using bone marrow chimeras and cell-specific cre mice, we demonstrated that ablation of Phd1 in haematopoietic cells but not in endothelial cells was both necessary and sufficient to inhibit experimental colitis. This effect relied, at least in part, on skewing of Phd1-deficient bone marrow-derived macrophages towards an anti-inflammatory M2 phenotype. These cells showed an attenuated nuclear factor-κB-dependent response to lipopolysaccharide (LPS), which in turn diminished endothelial chemokine expression. In addition, Phd1 deficiency in dendritic cells significantly reduced interleukin-1β production in response to LPS. Taken together, our results further support the development of selective PHD1 inhibitors for ulcerative colitis, and identify haematopoietic cells as their primary target. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
| | - Martine De Vos
- Department of Gastroenterology, Ghent University, Ghent, Belgium
| | - Ben Wielockx
- Heisenberg Research Group, Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Simon J Tavernier
- Department of Internal Medicine, Ghent University, Ghent, Belgium.,Inflammation Research Centre VIB, Zwijnaarde, Belgium
| | - Melissa Dullaers
- Inflammation Research Centre VIB, Zwijnaarde, Belgium.,Department of Pulmonary Medicine, Ghent University, Ghent, Belgium
| | - Sara Neyt
- Laboratory of Radiopharmacy, Ghent University, Ghent, Belgium
| | - Benedicte Descamps
- Infinity Imaging Laboratory (iMinds Medical IT-IBiTech-MEDISIP), Ghent University, Ghent, Belgium
| | | | - Sarah Devriese
- Department of Gastroenterology, Ghent University, Ghent, Belgium
| | | | - Tom Holvoet
- Department of Gastroenterology, Ghent University, Ghent, Belgium
| | - Ann Baeyens
- Department of Clinical Chemistry, Microbiology, and Immunology, Ghent University, Ghent, Belgium
| | - Carmen Correale
- IBD Centre, Department of Gastroenterology, Humanitas Clinical and Research Centre, Rozzano, Italy
| | - Silvia D'Alessio
- IBD Centre, Department of Gastroenterology, Humanitas Clinical and Research Centre, Rozzano, Italy
| | - Christian Vanhove
- Infinity Imaging Laboratory (iMinds Medical IT-IBiTech-MEDISIP), Ghent University, Ghent, Belgium
| | - Filip De Vos
- Laboratory of Radiopharmacy, Ghent University, Ghent, Belgium
| | - Bruno Verhasselt
- Department of Clinical Chemistry, Microbiology, and Immunology, Ghent University, Ghent, Belgium
| | - Georg Breier
- Division of Medical Biology, Department of Psychiatry, Technische Universität Dresden, Dresden, Germany
| | - Bart N Lambrecht
- Department of Internal Medicine, Ghent University, Ghent, Belgium.,Inflammation Research Centre VIB, Zwijnaarde, Belgium
| | - Sophie Janssens
- Department of Internal Medicine, Ghent University, Ghent, Belgium.,Inflammation Research Centre VIB, Zwijnaarde, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular metabolism, Vesalius Research Centre, KU Leuven, VIB, Leuven, Belgium
| | - Silvio Danese
- IBD Centre, Department of Gastroenterology, Humanitas Clinical and Research Centre, Rozzano, Italy
| | - Dirk Elewaut
- Inflammation Research Centre VIB, Zwijnaarde, Belgium.,Department of Rheumatology, Ghent University, Ghent, Belgium
| | - Debby Laukens
- Department of Gastroenterology, Ghent University, Ghent, Belgium
| | - Pieter Hindryckx
- Department of Gastroenterology, Ghent University, Ghent, Belgium
| |
Collapse
|
30
|
Sun M, He C, Wu W, Zhou G, Liu F, Cong Y, Liu Z. Hypoxia inducible factor-1α-induced interleukin-33 expression in intestinal epithelia contributes to mucosal homeostasis in inflammatory bowel disease. Clin Exp Immunol 2016; 187:428-440. [PMID: 27921309 DOI: 10.1111/cei.12896] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 10/13/2016] [Accepted: 11/02/2016] [Indexed: 01/05/2023] Open
Abstract
Intestinal epithelial cells (IECs), an important barrier to gut microbiota, are subject to low oxygen tension, particularly during intestinal inflammation. Hypoxia inducible factor-1α (HIF-1α) is expressed highly in the inflamed mucosa of inflammatory bowel disease (IBD) and functions as a key regulator in maintenance of intestinal homeostasis. However, how IEC-derived HIF-1α regulates intestinal immune responses in IBD is still not understood completely. We report here that the expression of HIF-1α and IL-33 was increased significantly in the inflamed mucosa of IBD patients as well as mice with colitis induced by dextran sulphate sodium (DSS). The levels of interleukin (IL)-33 were correlated positively with that of HIF-1α. A HIF-1α-interacting element was identified in the promoter region of IL-33, indicating that HIF-1α activity regulates IL-33 expression. Furthermore, tumour necrosis factor (TNF) facilitated the HIF-1α-dependent IL-33 expression in IEC. Our data thus demonstrate that HIF-1α-dependent IL-33 in IEC functions as a regulatory cytokine in inflamed mucosa of IBD, thereby regulating the intestinal inflammation and maintaining mucosal homeostasis.
Collapse
Affiliation(s)
- M Sun
- Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - C He
- Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - W Wu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - G Zhou
- Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - F Liu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Y Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Z Liu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| |
Collapse
|
31
|
Halligan DN, Murphy SJE, Taylor CT. The hypoxia-inducible factor (HIF) couples immunity with metabolism. Semin Immunol 2016; 28:469-477. [PMID: 27717536 DOI: 10.1016/j.smim.2016.09.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 09/23/2016] [Accepted: 09/30/2016] [Indexed: 12/16/2022]
Abstract
Crosstalk between metabolic and immune pathways has recently become appreciated to be key to the regulation of host defence. The hypoxia-inducible factor (HIF) is a transcription factor which was initially described as a ubiquitous master regulator of the transcriptional response to hypoxia. In this role, HIF regulates genes promoting adaptation to hypoxia including a number which influence the cellular metabolic strategy of a cell. It has more recently been appreciated that the regulation of HIF is not restricted to oxygen-dependent pathways, and is now known to be mediated by a number of additional metabolic and immune cues including metabolites and cytokines respectively. Furthermore, our understanding of the functional role of HIF has expanded to it now being appreciated as a major regulator of host immunity. This places HIF in an ideal position to act as a regulatory hub which links metabolic activity with immunity. In this review we synthesise recent data which identifies HIF as both a target and effector for metabolic and immune processes. Developing our understanding of the role of HIF in this context will uncover new therapeutic targets for inflammatory and infectious disease.
Collapse
Affiliation(s)
- Doug N Halligan
- Conway Institute, Charles Institute & Systems Biology Ireland, University College Dublin, Belfield Dublin 4, Ireland; Sigmoid Pharma, Invent Centre, Dublin City University, Dublin 9, Ireland
| | - Stephen J E Murphy
- Conway Institute, Charles Institute & Systems Biology Ireland, University College Dublin, Belfield Dublin 4, Ireland
| | - Cormac T Taylor
- Conway Institute, Charles Institute & Systems Biology Ireland, University College Dublin, Belfield Dublin 4, Ireland; IRCAN, Centre A. Lacassagne, University of Nice-Sophia Antipolis, 33 Avenue Valombrose, 06107 Nice, France; Centre Scientifique de Monaco (CSM), 8, Quai Antoine Premier, Monaco.
| |
Collapse
|
32
|
Kiers HD, Scheffer GJ, van der Hoeven JG, Eltzschig HK, Pickkers P, Kox M. Immunologic Consequences of Hypoxia during Critical Illness. Anesthesiology 2016; 125:237-49. [PMID: 27183167 PMCID: PMC5119461 DOI: 10.1097/aln.0000000000001163] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hypoxia and immunity are highly intertwined at clinical, cellular, and molecular levels. The prevention of tissue hypoxia and modulation of systemic inflammation are cornerstones of daily practice in the intensive care unit. Potentially, immunologic effects of hypoxia may contribute to outcome and represent possible therapeutic targets. Hypoxia and activation of downstream signaling pathways result in enhanced innate immune responses, aimed to augment pathogen clearance. On the other hand, hypoxia also exerts antiinflammatory and tissue-protective effects in lymphocytes and other tissues. Although human data on the net immunologic effects of hypoxia and pharmacologic modulation of downstream pathways are limited, preclinical data support the concept of tailoring the immune response through modulation of the oxygen status or pharmacologic modulation of hypoxia-signaling pathways in critically ill patients.
Collapse
Affiliation(s)
- Harmke D. Kiers
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Department of Anesthesiology, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Nijmegen, The Netherlands
| | - Gert-Jan Scheffer
- Department of Anesthesiology, Radboud university medical center, Nijmegen, The Netherlands
| | - Johannes G. van der Hoeven
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Nijmegen, The Netherlands
| | - Holger K. Eltzschig
- Organ Protection Program; Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Nijmegen, The Netherlands
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Department of Anesthesiology, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Nijmegen, The Netherlands
| |
Collapse
|
33
|
Fitzpatrick SF, Fábián Z, Schaible B, Lenihan CR, Schwarzl T, Rodriguez J, Zheng X, Li Z, Tambuwala MM, Higgins DG, O'Meara Y, Slattery C, Manresa MC, Fraisl P, Bruning U, Baes M, Carmeliet P, Doherty G, von Kriegsheim A, Cummins EP, Taylor CT. Prolyl hydroxylase-1 regulates hepatocyte apoptosis in an NF-κB-dependent manner. Biochem Biophys Res Commun 2016; 474:579-586. [DOI: 10.1016/j.bbrc.2016.04.085] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/18/2016] [Indexed: 01/21/2023]
|
34
|
Deng W, Feng X, Li X, Wang D, Sun L. Hypoxia-inducible factor 1 in autoimmune diseases. Cell Immunol 2016; 303:7-15. [DOI: 10.1016/j.cellimm.2016.04.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 04/06/2016] [Accepted: 04/06/2016] [Indexed: 12/29/2022]
|
35
|
The Effects of Hypoxia and Inflammation on Synaptic Signaling in the CNS. Brain Sci 2016; 6:brainsci6010006. [PMID: 26901230 PMCID: PMC4810176 DOI: 10.3390/brainsci6010006] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/21/2016] [Accepted: 02/02/2016] [Indexed: 12/16/2022] Open
Abstract
Normal brain function is highly dependent on oxygen and nutrient supply and when the demand for oxygen exceeds its supply, hypoxia is induced. Acute episodes of hypoxia may cause a depression in synaptic activity in many brain regions, whilst prolonged exposure to hypoxia leads to neuronal cell loss and death. Acute inadequate oxygen supply may cause anaerobic metabolism and increased respiration in an attempt to increase oxygen intake whilst chronic hypoxia may give rise to angiogenesis and erythropoiesis in order to promote oxygen delivery to peripheral tissues. The effects of hypoxia on neuronal tissue are exacerbated by the release of many inflammatory agents from glia and neuronal cells. Cytokines, such as TNF-α, and IL-1β are known to be released during the early stages of hypoxia, causing either local or systemic inflammation, which can result in cell death. Another growing body of evidence suggests that inflammation can result in neuroprotection, such as preconditioning to cerebral ischemia, causing ischemic tolerance. In the following review we discuss the effects of acute and chronic hypoxia and the release of pro-inflammatory cytokines on synaptic transmission and plasticity in the central nervous system. Specifically we discuss the effects of the pro-inflammatory agent TNF-α during a hypoxic event.
Collapse
|
36
|
Cui XY, Tinholt M, Stavik B, Dahm AEA, Kanse S, Jin Y, Seidl S, Sahlberg KK, Iversen N, Skretting G, Sandset PM. Effect of hypoxia on tissue factor pathway inhibitor expression in breast cancer. J Thromb Haemost 2016; 14:387-96. [PMID: 26598923 DOI: 10.1111/jth.13206] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 11/04/2015] [Indexed: 12/28/2022]
Abstract
UNLABELLED ESSENTIALS: A hypoxic microenvironment is a common feature of tumors that may influence activation of coagulation. MCF-7 and SK-BR-3 breast cancer cells and breast cancer tissue samples were used. The results showed transcriptional repression of tissue factor pathway inhibitor expression in hypoxia. Hypoxia-inducible factor 1α may be a target for the therapy of cancer-related coagulation and thrombosis. BACKGROUND Activation of coagulation is a common finding in patients with cancer, and is associated with an increased risk of venous thrombosis. As a hypoxic microenvironment is a common feature of solid tumors, we investigated the role of hypoxia in the regulation of tissue factor (TF) pathway inhibitor (TFPI) expression in breast cancer. OBJECTIVES To explore the transcriptional regulation of TFPI by hypoxia-inducible factor (HIF)-1α in breast cancer cells and their correlation in breast cancer tissues. METHODS AND RESULTS MCF-7 and SK-BR-3 breast cancer cells were cultured in 1% oxygen or treated with cobalt chloride (CoCl2 ) to mimic hypoxia. Time-dependent and dose-dependent downregulation of TFPI mRNA (quantitative RT-PCR) and of free TFPI protein (ELISA) were observed in hypoxia. Western blotting showed parallel increases in the levels of HIF-1α protein and TF. HIF-1α inhibitor abolished or attenuated the hypoxia-induced downregulation of TFPI. Luciferase reporter assay showed that both hypoxia and HIF-1α overexpression caused strong repression of TFPI promoter activity. Subsequent chromatin immunoprecipitation and mutagenesis analysis demonstrated a functional hypoxia response element within the TFPI promoter, located at -1065 to -1060 relative to the transcriptional start point. In breast cancer tissue samples, gene expression analyses showed a positive correlation between the mRNA expression of TFPI and that of HIF-1α. CONCLUSIONS This study demonstrates that HIF-1α is involved in the transcriptional regulation of the TFPI gene, and suggests that a hypoxic microenvironment inside a breast tumor may induce a procoagulant state in breast cancer patients.
Collapse
Affiliation(s)
- X Y Cui
- Department of Hematology, Oslo University Hospital, Oslo, Norway
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - M Tinholt
- Department of Hematology, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - B Stavik
- Department of Hematology, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - A E A Dahm
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Hematology, Akershus University Hospital, Lørenskog, Norway
| | - S Kanse
- Department of Hematology, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Basal Medical Sciences, University of Oslo, Oslo, Norway
| | - Y Jin
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - S Seidl
- Department of Hematology, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Basal Medical Sciences, University of Oslo, Oslo, Norway
| | - K K Sahlberg
- Department of Research, Vestre Viken Hospital Trust, Drammen, Norway
- K. G. Jebsen Center for Breast Cancer Research, University of Oslo, Oslo, Norway
- Oslo Breast Cancer Research Consortium (OSBREAC), Oslo, Norway
| | - N Iversen
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - G Skretting
- Department of Hematology, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - P M Sandset
- Department of Hematology, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
37
|
Abstract
In this review, I summarize some of the recent insight into pharmacological targeting of hypoxia in disease models. Studies from cultured cell systems, animal models, and translation to human patients have revealed that posttranslational modifications of individual proteins within NF-κB and hypoxia-inducible factor pathways serve as ideal targets for analysis in disease models. Studies defining differences and similarities between these responses have taught us a number of important lessons about the complexity of the inflammatory response. A clearer definition of these pathways has provided new insight into disease pathogenesis and, importantly, the potential for new therapeutic targets.
Collapse
|
38
|
Xiao Z, Sperl B, Ullrich A, Knyazev P. Metformin and salinomycin as the best combination for the eradication of NSCLC monolayer cells and their alveospheres (cancer stem cells) irrespective of EGFR, KRAS, EML4/ALK and LKB1 status. Oncotarget 2015; 5:12877-90. [PMID: 25375092 PMCID: PMC4350329 DOI: 10.18632/oncotarget.2657] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 11/02/2014] [Indexed: 01/01/2023] Open
Abstract
The presence of cancer stem cells (CSCs) is linked to preexisting or acquired drug resistance and tumor relapse. Therefore, targeting both differentiated tumor cells and CSCs was suggested as an effective approach for non-small cell lung cancer (NSCLC) treatment. After screening of chemotherapeutic agents, tyrosine kinase inhibitors (TKIs) or monoclonal antibody in combination with the putative stem cell killer Salinomycin (SAL), we found Metformin (METF), which modestly exerted a growth inhibitory effect on monolayer cells and alveospheres/CSCs of 5 NSCLC cell lines regardless of their EGFR, KRAS, EML4/ALK and LKB1 status, interacted synergistically with SAL to effectively promote cell death. Inhibition of EGFR (AKT, ERK1/2) and mTOR (p70 s6k) signaling with the combination of METF and SAL can be augmented beyond that achieved using each agent individually. Phospho-kinase assay further suggested the multiple roles of this combination in reducing oncogenic effects of modules, such as ß-catenin, Src family kinases (Src, Lyn, Yes), Chk-2 and FAK. Remarkably, significant reduction of sphere formation was seen under combinatorial treatment in all investigated NSCLC cell lines. In conclusion, METF in combination with SAL could be a promising treatment option for patients with advanced NSCLC irrespective of their EGFR, KRAS, EML4/ALK and LKB1 status.
Collapse
Affiliation(s)
- Zhiguang Xiao
- Department of Molecular Biology, Max-Planck-Institute of Biochemistry, Am Klopferspitz, Martinsried, Germany
| | - Bianca Sperl
- Department of Molecular Biology, Max-Planck-Institute of Biochemistry, Am Klopferspitz, Martinsried, Germany
| | - Axel Ullrich
- Department of Molecular Biology, Max-Planck-Institute of Biochemistry, Am Klopferspitz, Martinsried, Germany
| | - Pjotr Knyazev
- Department of Molecular Biology, Max-Planck-Institute of Biochemistry, Am Klopferspitz, Martinsried, Germany
| |
Collapse
|
39
|
Tambuwala MM, Manresa MC, Cummins EP, Aversa V, Coulter IS, Taylor CT. Targeted delivery of the hydroxylase inhibitor DMOG provides enhanced efficacy with reduced systemic exposure in a murine model of colitis. J Control Release 2015; 217:221-7. [DOI: 10.1016/j.jconrel.2015.09.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/11/2015] [Accepted: 09/12/2015] [Indexed: 12/30/2022]
|
40
|
Zhdanov AV, Okkelman IA, Collins FW, Melgar S, Papkovsky DB. A novel effect of DMOG on cell metabolism: direct inhibition of mitochondrial function precedes HIF target gene expression. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1847:1254-66. [DOI: 10.1016/j.bbabio.2015.06.016] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 06/11/2015] [Accepted: 06/30/2015] [Indexed: 12/17/2022]
|
41
|
Habryka A, Gogler-Pigłowska A, Sojka D, Kryj M, Krawczyk Z, Scieglinska D. Cell type-dependent modulation of the gene encoding heat shock protein HSPA2 by hypoxia-inducible factor HIF-1: Down-regulation in keratinocytes and up-regulation in HeLa cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:1155-69. [PMID: 26164067 DOI: 10.1016/j.bbagrm.2015.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 06/24/2015] [Accepted: 07/07/2015] [Indexed: 11/26/2022]
Abstract
HSPA2 belongs to the multigene HSPA family, whose members encode chaperone proteins. Although expression and function of HSPA2 is mainly associated with spermatogenesis, recent studies demonstrated that in humans, the gene is active in various cancers, as well as in normal tissues, albeit in a cell type-specific manner. In the epidermis, HSPA2 is expressed in keratinocytes in the basal layer. Currently, the mechanisms underlying the regulation of HSPA2 expression remain unknown. This study was aimed at determining whether HIF-1 and its binding site, the hypoxia-response element (HRE) located in the HSPA2 promoter, are involved in HSPA2 regulation. As a model system, we used an immortal human keratinocyte line (HaCaT) and cervical cancer cells (HeLa) grown under control or hypoxic conditions. Using an in vitro gene reporter assay, we demonstrated that in keratinocytes HSPA2 promoter activity is reduced under conditions that facilitate stabilization of HIF-1α, whereas HIF-1 inhibitors abrogated the suppressive effect of hypoxia on promoter activity. Chromatin immunoprecipitation revealed that HIF-1α binds to the HSPA2 promoter. In keratinocytes, hypoxia or overexpression of a stable form of HIF-1α attenuated the expression of endogenous HSPA2, whereas targeted repression of HIF-1α by RNAi increased transcription of HSPA2 under hypoxia. Conversely, in HeLa cells, HSPA2 expression increased under conditions that stimulated HIF-1α activity, whereas inhibition of HIF-1α abrogated hypoxia-induced up-regulation of HSPA2 expression. Taken together, our results demonstrate that HIF-1 can exert differential, cell context-dependent regulatory control of the HSPA2 gene. Additionally, we also showed that HSPA2 expression can be stimulated during hypoxia/reoxygenation stress.
Collapse
Affiliation(s)
- Anna Habryka
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Agnieszka Gogler-Pigłowska
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Damian Sojka
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Mariusz Kryj
- The Oncologic and Reconstructive Surgery Clinic, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Zdzisław Krawczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Dorota Scieglinska
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland.
| |
Collapse
|
42
|
Abstract
Oxygen is the basic molecule which supports life and it truly is "god's gift to life." Despite its immense importance, research on "oxygen biology" has never received the light of the day and has been limited to physiological and biochemical studies. It seems that in modern day biology, oxygen research is summarized in one word "hypoxia." Scientists have focused on hypoxia-induced transcriptomics and molecular-cellular alterations exclusively in disease models. Interestingly, the potential of oxygen to control the basic principles of biology like homeostatic maintenance, transcription, replication, and protein folding among many others, at the molecular level, has been completely ignored. Here, we present a perspective on the crucial role played by oxygen in regulation of basic biological phenomena. Our conclusion highlights the importance of establishing novel research areas like oxygen biology, as there is great potential in this field for basic science discoveries and clinical benefits to the society.
Collapse
|
43
|
Caria CREP, Moscato CH, Tomé RBG, Pedrazzoli J, Ribeiro ML, Gambero A. Nitric oxide interferes with hypoxia signaling during colonic inflammation. ARQUIVOS DE GASTROENTEROLOGIA 2015; 51:302-8. [PMID: 25591158 DOI: 10.1590/s0004-28032014000400007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 07/08/2014] [Indexed: 01/24/2023]
Abstract
CONTEXT Intestinal inflammation can induce a local reduction in oxygen levels that triggers an adaptive response centered on the expression of hypoxia-inducible factors (HIFs). Nitric oxide, a well-described inflammatory mediator, may interfere with hypoxia signaling. OBJECTIVES We aimed to evaluate the role of nitric oxide in hypoxia signaling during colonic inflammation. METHODS Colitis was induced by single (acute) or repeated (reactivated colitis) trinitrobenzenosulfonic acid administration in rats. In addition, one group of rats with reactivated colitis was also treated with Nw-Nitro-L-arginine methyl ester hydrochloride to block nitric oxide synthase. Colitis was assessed by macroscopic score and myeloperoxidase activity in the colon samples. Hypoxia was determined using the oxygen-dependent probe, pimonidazole. The expression of HIF-1α and HIF-induced factors (vascular endothelial growth factor - VEGF and apelin) was assessed using Western blotting. RESULTS The single or repeated administration of trinitrobenzenosulfonic acid to rats induced colitis which was characterized by a high macroscopic score and myeloperoxidase activity. Hypoxia was observed with both protocols. During acute colitis, HIF-1α expression was not increased, but VEGF and apelin were increased. HIF-1α expression was inhibited during reactivated colitis, and VEGF and apelin were not increased. Nw-Nitro-L-arginine methyl ester hydrochloride blockade during reactivated colitis restored HIF-1α, VEGF and apelin expression. CONCLUSIONS Nitric oxide could interfere with hypoxia signaling during reactivated colitis inflammation modifying the expression of proteins regulated by HIF-1α.
Collapse
Affiliation(s)
- Cintia Rabelo e Paiva Caria
- Unidade Integrada de Farmacologia e Gastroenterologia (Unifag), Faculdade de Medicina da Universidade São Francisco, São Paulo, SP, Brasil
| | - Camila Henrique Moscato
- Unidade Integrada de Farmacologia e Gastroenterologia (Unifag), Faculdade de Medicina da Universidade São Francisco, São Paulo, SP, Brasil
| | - Renata Bortolin Guerra Tomé
- Unidade Integrada de Farmacologia e Gastroenterologia (Unifag), Faculdade de Medicina da Universidade São Francisco, São Paulo, SP, Brasil
| | - José Pedrazzoli
- Unidade Integrada de Farmacologia e Gastroenterologia (Unifag), Faculdade de Medicina da Universidade São Francisco, São Paulo, SP, Brasil
| | - Marcelo Lima Ribeiro
- Unidade Integrada de Farmacologia e Gastroenterologia (Unifag), Faculdade de Medicina da Universidade São Francisco, São Paulo, SP, Brasil
| | - Alessandra Gambero
- Unidade Integrada de Farmacologia e Gastroenterologia (Unifag), Faculdade de Medicina da Universidade São Francisco, São Paulo, SP, Brasil
| |
Collapse
|
44
|
Hu D, Yu Y, Wang C, Li D, Tai Y, Fang L. microRNA-98 mediated microvascular hyperpermeability during burn shock phase via inhibiting FIH-1. Eur J Med Res 2015; 20:51. [PMID: 25903459 PMCID: PMC4411771 DOI: 10.1186/s40001-015-0141-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 04/13/2015] [Indexed: 12/19/2022] Open
Abstract
Background microRNA is a small non-coding RNA molecule and functions in RNA silencing and post-transcriptional regulation of gene expression. This study was designed to evaluate the role of miR-98 in the development of microvascular permeability and its molecular pathogenesis. Methods Forty-eight healthy adult Wistar rats were divided into the control group (n = 8) and burn group (n = 40) that inflicted with 30% total body surface area third-degree burn. Groups were processed at 2, 4, 8, 12, and 24 h post-burn. Plasma for vascular endothelial cell culture was collected from control and 12 h post-burn rats. Organic microvascular permeability and serum miR-98 level were measured. In vitro, rat aorta endothelial cells were stimulated with burn serum. Level of miR-98 and protein of hypoxia-inducible factor-1 (HIF-1), factor inhibiting HIF-1α (FIH-1), and tight junction-associated proteins were determined. Results Organic microvascular permeability began to rise at 2 h post-burn and maintained the same character throughout the experiment except in lung tissue that was still rising at 12 h; the serum level of miR-98 was elevated (P < 0.05). In vitro, burn serum stimulation increased rat aorta endothelial monolayer cell permeability as well as upregulated miR-98 expression (P < 0.05). As shown in the result of transfection experiment, miR-98 negatively regulated FIH-1 and tight junction-associated protein expression (P < 0.05). Conclusions The findings of the present study suggest severe microvascular permeability due to burns; and the underlying mechanism bases on the promotion of miR-98 level to the extent that it activated HIF-1 gene expression, resulting in junction-associated protein deficiency.
Collapse
Affiliation(s)
- Delin Hu
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui Province, 230022, People's Republic of China.
| | - Youxin Yu
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui Province, 230022, People's Republic of China.
| | - Chunhua Wang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui Province, 230022, People's Republic of China.
| | - Denghui Li
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui Province, 230022, People's Republic of China.
| | - Yuncheng Tai
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui Province, 230022, People's Republic of China.
| | - Linsen Fang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui Province, 230022, People's Republic of China.
| |
Collapse
|
45
|
Kelly CJ, Zheng L, Campbell EL, Saeedi B, Scholz CC, Bayless AJ, Wilson KE, Glover LE, Kominsky DJ, Magnuson A, Weir TL, Ehrentraut SF, Pickel C, Kuhn KA, Lanis JM, Nguyen V, Taylor CT, Colgan SP. Crosstalk between Microbiota-Derived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. Cell Host Microbe 2015; 17:662-71. [PMID: 25865369 DOI: 10.1016/j.chom.2015.03.005] [Citation(s) in RCA: 1155] [Impact Index Per Article: 115.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 11/21/2014] [Accepted: 01/22/2015] [Indexed: 02/07/2023]
Abstract
Interactions between the microbiota and distal gut are fundamental determinants of human health. Such interactions are concentrated at the colonic mucosa and provide energy for the host epithelium through the production of the short-chain fatty acid butyrate. We sought to determine the role of epithelial butyrate metabolism in establishing the austere oxygenation profile of the distal gut. Bacteria-derived butyrate affects epithelial O2 consumption and results in stabilization of hypoxia-inducible factor (HIF), a transcription factor coordinating barrier protection. Antibiotic-mediated depletion of the microbiota reduces colonic butyrate and HIF expression, both of which are restored by butyrate supplementation. Additionally, germ-free mice exhibit diminished retention of O2-sensitive dyes and decreased stabilized HIF. Furthermore, the influences of butyrate are lost in cells lacking HIF, thus linking butyrate metabolism to stabilized HIF and barrier function. This work highlights a mechanism where host-microbe interactions augment barrier function in the distal gut.
Collapse
Affiliation(s)
- Caleb J Kelly
- Mucosal Inflammation Program, University of Colorado, Aurora, CO 80045, USA; Department of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Leon Zheng
- Mucosal Inflammation Program, University of Colorado, Aurora, CO 80045, USA; Department of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Eric L Campbell
- Mucosal Inflammation Program, University of Colorado, Aurora, CO 80045, USA; Department of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Bejan Saeedi
- Mucosal Inflammation Program, University of Colorado, Aurora, CO 80045, USA; Department of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Carsten C Scholz
- School of Medicine and Medical Science, Conway Institute, University College Dublin, Ireland
| | - Amanda J Bayless
- Mucosal Inflammation Program, University of Colorado, Aurora, CO 80045, USA; Department of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Kelly E Wilson
- Mucosal Inflammation Program, University of Colorado, Aurora, CO 80045, USA; Department of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Louise E Glover
- Mucosal Inflammation Program, University of Colorado, Aurora, CO 80045, USA; Department of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Douglas J Kominsky
- Mucosal Inflammation Program, University of Colorado, Aurora, CO 80045, USA; Department of Anesthesiology, University of Colorado, Aurora, CO 80045, USA
| | - Aaron Magnuson
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA
| | - Tiffany L Weir
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA
| | - Stefan F Ehrentraut
- Mucosal Inflammation Program, University of Colorado, Aurora, CO 80045, USA; Department of Medicine, University of Colorado, Aurora, CO 80045, USA; Department of Anesthesiology, University of Bonn, Bonn 53113, Germany
| | - Christina Pickel
- School of Medicine and Medical Science, Conway Institute, University College Dublin, Ireland
| | - Kristine A Kuhn
- Mucosal Inflammation Program, University of Colorado, Aurora, CO 80045, USA; Department of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Jordi M Lanis
- Mucosal Inflammation Program, University of Colorado, Aurora, CO 80045, USA; Department of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Vu Nguyen
- Department of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Cormac T Taylor
- School of Medicine and Medical Science, Conway Institute, University College Dublin, Ireland
| | - Sean P Colgan
- Mucosal Inflammation Program, University of Colorado, Aurora, CO 80045, USA; Department of Medicine, University of Colorado, Aurora, CO 80045, USA.
| |
Collapse
|
46
|
Colgan SP, Curtis VF, Lanis JM, Glover LE. Metabolic regulation of intestinal epithelial barrier during inflammation. Tissue Barriers 2015; 3:e970936. [PMID: 25838978 DOI: 10.4161/21688362.2014.970936] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/25/2014] [Indexed: 12/21/2022] Open
Abstract
The gastrointestinal mucosa has proven to be an interesting tissue for which to investigate disease-related metabolism. In this review, we outline some evidence that implicates metabolic signaling as important features of barrier in the healthy and disease. Studies from cultured cell systems, animal models and human patients have revealed that metabolites generated within the inflammatory microenvironment are central to barrier regulation. These studies have revealed a prominent role for hypoxia and hypoxia-inducible factor (HIF) at key steps in adenine nucleotide metabolism and within the creatine kinase pathway. Results from animal models of intestinal inflammation have demonstrated an almost uniformly beneficial influence of HIF stabilization on disease outcomes and barrier function. Studies underway to elucidate the contribution of immune responses will provide additional insight into how metabolic changes contribute to the complexity of the gastrointestinal tract and how such information might be harnessed for therapeutic benefit.
Collapse
Key Words
- AMP, adenosine monophosphate
- CK, creatine kinase
- ChIP, chromatin immunoprecipitation
- Colitis
- HIF, hypoxia-inducible factor
- PHD, prolyl hydroxylase
- PMN, polymorphonuclear leukcoyte, neutrophil
- TJ, tight junction
- VASP, vasodilator-stimulated
- ZO-1, zonula occludens-1
- creatine
- epithelium
- inflammation
- metabolism
- mucosa
- murine model
- neutrophil
- nucleoside
- nucleotidase
- nucleotide
- phosphocreatine
Collapse
Affiliation(s)
- Sean P Colgan
- Departments of Medicine and the Mucosal Inflammation Program; University of Colorado School of Medicine ; Aurora, CO USA
| | - Valerie F Curtis
- Departments of Medicine and the Mucosal Inflammation Program; University of Colorado School of Medicine ; Aurora, CO USA
| | - Jordi M Lanis
- Departments of Medicine and the Mucosal Inflammation Program; University of Colorado School of Medicine ; Aurora, CO USA
| | - Louise E Glover
- Departments of Medicine and the Mucosal Inflammation Program; University of Colorado School of Medicine ; Aurora, CO USA
| |
Collapse
|
47
|
Manresa MC, Godson C, Taylor CT. Hypoxia-sensitive pathways in inflammation-driven fibrosis. Am J Physiol Regul Integr Comp Physiol 2014; 307:R1369-80. [PMID: 25298511 DOI: 10.1152/ajpregu.00349.2014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tissue injury can occur for a variety of reasons, including physical damage, infection, and ischemia. The ability of tissues to effectively recover from injury is a cornerstone of human health. The healing response in tissues is conserved across organs and typically involves distinct but overlapping inflammatory, proliferative, and maturation/resolution phases. If the inflammatory phase is not successfully controlled and appropriately resolved, an excessive healing response characterized by scar formation can lead to tissue fibrosis, a major clinical complication in disorders such as Crohn's disease (CD). As a result of enhanced metabolic and inflammatory processes during chronic inflammation, profound changes in tissue oxygen levels occur leading to localized tissue hypoxia. Therefore, inflammation, fibrosis, and hypoxia are coincidental events during inflammation-driven fibrosis. Our current understanding of the mechanism(s) underpinning fibrosis is limited as are the therapeutic options available. In this review, we discuss what is known about the cellular and molecular mechanisms underpinning inflammation-driven fibrosis and how hypoxia may play a role in shaping this process.
Collapse
Affiliation(s)
- Mario C Manresa
- School of Medicine and Medical Science and the Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Catherine Godson
- School of Medicine and Medical Science and the Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Cormac T Taylor
- School of Medicine and Medical Science and the Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
48
|
Short SS, Wang J, Castle SL, Fernandez GE, Smiley N, Zobel M, Pontarelli EM, Papillon SC, Grishin AV, Ford HR. Low doses of celecoxib attenuate gut barrier failure during experimental peritonitis. J Transl Med 2013; 93:1265-75. [PMID: 24126890 PMCID: PMC3966546 DOI: 10.1038/labinvest.2013.119] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Revised: 09/13/2013] [Accepted: 09/14/2013] [Indexed: 02/08/2023] Open
Abstract
The intestinal barrier becomes compromised during systemic inflammation, leading to the entry of luminal bacteria into the host and gut origin sepsis. Pathogenesis and treatment of inflammatory gut barrier failure is an important problem in critical care. In this study, we examined the role of cyclooxygenase-2 (COX-2), a key enzyme in the production of inflammatory prostanoids, in gut barrier failure during experimental peritonitis in mice. I.p. injection of LPS or cecal ligation and puncture (CLP) increased the levels of COX-2 and its product prostaglandin E2 (PGE2) in the ileal mucosa, caused pathologic sloughing of the intestinal epithelium, increased passage of FITC-dextran and bacterial translocation across the barrier, and increased internalization of the tight junction (TJ)-associated proteins junction-associated molecule-A and zonula occludens-1. Luminal instillation of PGE2 in an isolated ileal loop increased transepithelial passage of FITC-dextran. Low doses (0.5-1 mg/kg), but not a higher dose (5 mg/kg) of the specific COX-2 inhibitor Celecoxib partially ameliorated the inflammatory gut barrier failure. These results demonstrate that high levels of COX-2-derived PGE2 seen in the mucosa during peritonitis contribute to gut barrier failure, presumably by compromising TJs. Low doses of specific COX-2 inhibitors may blunt this effect while preserving the homeostatic function of COX-2-derived prostanoids. Low doses of COX-2 inhibitors may find use as an adjunct barrier-protecting therapy in critically ill patients.
Collapse
Affiliation(s)
- Scott S. Short
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA,Department of Surgery, University of Southern California, Los Angeles, CA
| | - Jin Wang
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA
| | - Shannon L. Castle
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA,Department of Surgery, University of Southern California, Los Angeles, CA
| | | | - Nancy Smiley
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA
| | - Michael Zobel
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA
| | - Elizabeth M. Pontarelli
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA,Department of Surgery, University of Southern California, Los Angeles, CA
| | - Stephanie C. Papillon
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA,Department of Surgery, University of Southern California, Los Angeles, CA
| | - Anatoly V. Grishin
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA,Department of Surgery, University of Southern California, Los Angeles, CA
| | - Henri R. Ford
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA,Department of Surgery, University of Southern California, Los Angeles, CA
| |
Collapse
|
49
|
Van Welden S, Laukens D, Ferdinande L, De Vos M, Hindryckx P. Differential expression of prolyl hydroxylase 1 in patients with ulcerative colitis versus patients with Crohn's disease/infectious colitis and healthy controls. JOURNAL OF INFLAMMATION-LONDON 2013; 10:36. [PMID: 24257430 PMCID: PMC3842628 DOI: 10.1186/1476-9255-10-36] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 11/14/2013] [Indexed: 01/15/2023]
Abstract
BACKGROUND Inhibition of prolyl hydroxylases (PHDs) leads to the induction of a transcriptional program that, in the gut, promotes intestinal epithelial cell survival. PHD inhibitors have recently been suggested as a promising alternative treatment for inflammatory bowel disease (IBD). In this study, we explored the colonic mucosal expression of the different PHD-isoforms (PHD1, 2 and 3) in order to identify the key isoform(s) involved in the pathogenesis of IBD. METHODS The mRNA expression of inflammatory cytokines (IL-8 and TNF-α), an apoptosis marker (caspase 3) and PHD1, 2 and 3 was analysed in biopsies of IBD patients (UC and CD), patients with infectious colitis and healthy controls using qRT-PCR. PHD protein levels were evaluated using western blot. Cellular localization of PHD 1, 2 and 3 was determined by immunohistochemistry. RESULTS PHD1 was significantly up-regulated in IBD patients, both at the mRNA (UC: p < 0.0001 and CD: p < 0.05) and at the protein level (UC: p < 0.05 and CD: p < 0.05), and showed a very good correlation with the expression of the inflammatory cytokines IL-8 and TNF-α and the apoptosis marker caspase 3. Colonic mucosal PHD2 mRNA and protein expressions were not altered in IBD. PHD3 expression was increased in inflamed biopsies from UC patients (p < 0.0001), but only at the mRNA level. PHD1 and PHD2 expression was found both in the colonic lamina propria and the epithelium while PHD3 was mainly located in the endothelium of blood vessels. CONCLUSIONS In this exploratory expression analysis, PHD1 comes forward as the primary therapeutic target for UC and, to a lesser extent, for (colonic) CD.
Collapse
Affiliation(s)
| | | | | | | | - Pieter Hindryckx
- Department of Gastroenterology, Ghent University Hospital, De Pintelaan 185, 3K12-IE, Ghent B-9000, Belgium.
| |
Collapse
|
50
|
Fundamental role for HIF-1α in constitutive expression of human β defensin-1. Mucosal Immunol 2013; 6:1110-8. [PMID: 23462909 PMCID: PMC3740147 DOI: 10.1038/mi.2013.6] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 01/08/2013] [Indexed: 02/04/2023]
Abstract
Antimicrobial peptides are secreted by the intestinal epithelium to defend from microbial threats. The role of human β defensin-1 (hBD-1) is notable because its gene (beta-defensin 1 (DEFB1)) is constitutively expressed and its antimicrobial activity is potentiated in the low-oxygen environment that characterizes the intestinal mucosa. Hypoxia-inducible factor (HIF) is stabilized even in healthy intestinal mucosa, and we identified that epithelial HIF-1α maintains expression of murine defensins. Extension to a human model revealed that basal HIF-1α is critical for the constitutive expression of hBD-1. Chromatin immunoprecipitation identified HIF-1α binding to a hypoxia response element in the DEFB1 promoter whose importance was confirmed by site-directed mutagenesis. We used 94 human intestinal samples to identify a strong expression correlation between DEFB1 and the canonical HIF-1α target GLUT1. These findings indicate that basal HIF-1α is critical for constitutive expression of enteric DEFB1 and support targeting epithelial HIF for restoration and maintenance of intestinal integrity.
Collapse
|