1
|
Foti F, Schuler C, Ruiz PA, Perren L, Malagola E, de Vallière C, Seuwen K, Hausmann M, Rogler G. The Simultaneous Deletion of pH-Sensing Receptors GPR4 and OGR1 (GPR68) Ameliorates Colitis with Additive Effects on Multiple Parameters of Inflammation. Int J Mol Sci 2025; 26:1552. [PMID: 40004018 PMCID: PMC11855581 DOI: 10.3390/ijms26041552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
G protein-coupled receptors (GPRs), including pro-inflammatory GPR4 and ovarian cancer GPR1 (OGR1/GPR68), are involved in the pH sensing of the extracellular space and have been implicated in inflammatory bowel disease (IBD). Previous data show that a loss of GPR4 or OGR1 independently is associated with reduced intestinal inflammation in mouse models of experimental colitis. In the present manuscript, we investigated the impact of the simultaneous loss of GPR4 and OGR1 in animal models of IBD. To study the effects of combined loss of Gpr4 Ogr1 in IBD we used the well-established acute dextran sodium sulfate (DSS) and spontaneous Il10-/- murine colitis models. Disease severity was assessed using multiple clinical scores (e.g., body weight loss, disease activity score, murine endoscopic index of colitis severity (MEICS) and histological analyses). Real-time quantitative polymerase chain reaction (qPCR), Western blot, and flow cytometry were used to investigate changes in pro-inflammatory cytokines expression and immune cells infiltration. We found that a combined loss of GPR4 and OGR1 significantly reduces colon inflammation in IBD relative to single deficiencies as evidenced by reduced body weight loss, disease score, CD4/CD8 ratio, and Il1β, Il6, and Tnf in the colon. Similarly, in the II10 deficiency model, the inflammation was significantly ameliorated upon the simultaneous deletion of GPR4 and OGR1, evidenced by a reduction in the MEICS score, colon length, Tnf and Il1β measurements, and a decrease in the number of macrophages in the colon, as compared to single deletions. Importantly, hydroxyproline levels were decreased close to baseline in Il10-/- × Gpr4-/- × Ogr1-/- mice. Our findings demonstrate that the simultaneous loss of GRP4 and OGR1 functions exerts an additive effect on multiple parameters associated with colonic inflammation. These results further reinforce the hypothesis that chronic inflammatory acidosis is a driver of fibrosis and is dependent on GPR4 and OGR1 signaling. The inhibition of both GPR4 and OGR1 by pH-sensing receptor modulators may constitute as a potential therapeutic option for IBD, as both pH-sensing receptors appear to sustain inflammation by acting on complementary pro-inflammatory pathways.
Collapse
|
2
|
Wu J, Li L, Zhang T, Lu J, Tai Z, Zhu Q, Chen Z. The epidermal lipid-microbiome loop and immunity: Important players in atopic dermatitis. J Adv Res 2025; 68:359-374. [PMID: 38460775 PMCID: PMC11785582 DOI: 10.1016/j.jare.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 02/10/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024] Open
Abstract
BACKGROUND The promotion of epidermal barrier dysfunction is attributed to abnormalities in the lipid-microbiome positive feedback loop which significantly influences the imbalance of the epithelial immune microenvironment (EIME) in atopic dermatitis (AD). This imbalance encompasses impaired lamellar membrane integrity, heightened exposure to epidermal pathogens, and the regulation of innate and adaptive immunity. The lipid-microbiome loop is substantially influenced by intense adaptive immunity which is triggered by abnormal loop activity and affects the loop's integrity through the induction of atypical lipid composition and responses to dysregulated epidermal microbes. Immune responses participate in lipid abnormalities within the EIME by downregulating barrier gene expression and are further cascade-amplified by microbial dysregulation which is instigated by barrier impairment. AIM OF REVIEW This review examines the relationship between abnormal lipid composition, microbiome disturbances, and immune responses in AD while progressively substantiating the crosstalk mechanism among these factors. Based on this analysis, the "lipid-microbiome" positive feedback loop, regulated by immune responses, is proposed. KEY SCIENTIFIC CONCEPTS OF REVIEW The review delves into the impact of adaptive immune responses that regulate the EIME, driving AD, and investigates potential mechanisms by which lipid supplementation and probiotics may alleviate AD through the up-regulation of the epidermal barrier and modulation of immune signaling. This exploration offers support for targeting the EIME to attenuate AD.
Collapse
Affiliation(s)
- Junchao Wu
- School of Medicine, Shanghai University, Shanghai 200444, China; Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Lisha Li
- School of Medicine, Shanghai University, Shanghai 200444, China; Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Tingrui Zhang
- School of Medicine, Shanghai University, Shanghai 200444, China; Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Jiaye Lu
- School of Medicine, Shanghai University, Shanghai 200444, China; Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China; Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, 200443, China.
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China; Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, 200443, China.
| | - Zhongjian Chen
- School of Medicine, Shanghai University, Shanghai 200444, China; Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China; Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, 200443, China.
| |
Collapse
|
3
|
He X, Hawkins C, Lawley L, Phan TM, Park I, Joven N, Zhang J, Wunderlich M, Mizukawa B, Pei S, Patel A, VanOudenhove J, Halene S, Fang J. GPR68 supports AML cells through the calcium/calcineurin pro-survival pathway and confers chemoresistance by mediating glucose metabolic symbiosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167565. [PMID: 39522891 DOI: 10.1016/j.bbadis.2024.167565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/21/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Accumulating evidence demonstrates that the "Warburg effect" that glycolysis is enhanced even in the presence of oxygen existed in hematopoietic malignancies, contributing to extracellular acidosis. G-protein coupled receptor 68 (GPR68), as a proton sensing GPCR responding to extracellular acidosis, is expected to play a critical role in hematopoietic malignancies. In the present study, we found that GPR68 was overexpressed in acute myeloid leukemia (AML) cells, and GPR68 deficiency impaired AML cell survival in vitro and cell engraftment in vivo. Mechanistic studies revealed that unlike GPR68 regulates Calpain1 in myelodysplastic syndromes (MDS) cells, GPR68 deficiency reduced cytosolic Ca2+ levels and calcineurin (CaN) activity in AML cells through an NFAT-independent mechanism. Moreover, the decreased Ca2+ levels disturbed cellular respiration (i.e., oxidative phosphorylation, OxPhos) by inhibiting isocitrate dehydrogenase (IDH) activity; this was more pronounced when BCL2 was inhibited simultaneously. Interestingly, GPR68 inhibition also decreased aerobic glycolysis in AML cells in a Ca2+-independent manner, suggesting that GPR68 mediated glucose metabolic symbiosis. As glucose metabolic symbiosis and the heterogeneous dependencies on aerobic glycolysis and cellular respiration tremendously impact chemosensitivity, the inhibition of GPR68 potentiated the tumoricidal effect of first-line chemotherapeutic agents, including BCL-2 inhibitors targeting OxPhos and cytarabine (Ara-C) targeting glycolysis. Consistent with these in vitro observations, higher levels of GPR68 were associated with inferior clinical outcomes in AML patients who received chemotherapies. In short, GPR68 drives the Ca2+/CaN pro-survival pathway and mediates glucose metabolic pathways in AML cells. Targeting GPR68 eradicates AML cells and alleviates chemoresistance, which could be exploited as a therapeutic target.
Collapse
MESH Headings
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Humans
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Calcineurin/metabolism
- Calcium/metabolism
- Glucose/metabolism
- Animals
- Drug Resistance, Neoplasm
- Mice
- Cell Survival/drug effects
- Cell Line, Tumor
- Glycolysis
Collapse
Affiliation(s)
- Xiaofei He
- First Affliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Zhejiang Province, China; Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Caleb Hawkins
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Lauren Lawley
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Tra Mi Phan
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Isaac Park
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Nicole Joven
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Jiajia Zhang
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Mark Wunderlich
- Cancer and Blood Disease Institutes, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Benjamin Mizukawa
- Cancer and Blood Disease Institutes, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Shanshan Pei
- Division of Hematology, University of Colorado, Denver, CO 80045, USA
| | - Amisha Patel
- Section of Hematology, Yale Cancer Center and Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jennifer VanOudenhove
- Section of Hematology, Yale Cancer Center and Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Stephanie Halene
- Section of Hematology, Yale Cancer Center and Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Stem Cell Center and Yale RNA Center, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jing Fang
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA.
| |
Collapse
|
4
|
Chen YJN, Shi RC, Xiang YC, Fan L, Tang H, He G, Zhou M, Feng XZ, Tan JD, Huang P, Ye X, Zhao K, Fu WY, Li LL, Bian XT, Chen H, Wang F, Wang T, Zhang CK, Zhou BH, Chen W, Liang TT, Lv JT, Kang X, Shi YX, Kim E, Qin YH, Hettinghouse A, Wang KD, Zhao XL, Yang MY, Tang YZ, Piao HL, Guo L, Liu CJ, Miao HM, Tang KL. Malate initiates a proton-sensing pathway essential for pH regulation of inflammation. Signal Transduct Target Ther 2024; 9:367. [PMID: 39737965 PMCID: PMC11683149 DOI: 10.1038/s41392-024-02076-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 01/01/2025] Open
Abstract
Metabolites can double as a signaling modality that initiates physiological adaptations. Metabolism, a chemical language encoding biological information, has been recognized as a powerful principle directing inflammatory responses. Cytosolic pH is a regulator of inflammatory response in macrophages. Here, we found that L-malate exerts anti-inflammatory effect via BiP-IRF2BP2 signaling, which is a sensor of cytosolic pH in macrophages. First, L-malate, a TCA intermediate upregulated in pro-inflammatory macrophages, was identified as a potent anti-inflammatory metabolite through initial screening. Subsequent screening with DARTS and MS led to the isolation of L-malate-BiP binding. Further screening through protein‒protein interaction microarrays identified a L-malate-restrained coupling of BiP with IRF2BP2, a known anti-inflammatory protein. Interestingly, pH reduction, which promotes carboxyl protonation of L-malate, facilitates L-malate and carboxylate analogues such as succinate to bind BiP, and disrupt BiP-IRF2BP2 interaction in a carboxyl-dependent manner. Both L-malate and acidification inhibit BiP-IRF2BP2 interaction, and protect IRF2BP2 from BiP-driven degradation in macrophages. Furthermore, both in vitro and in vivo, BiP-IRF2BP2 signal is required for effects of both L-malate and pH on inflammatory responses. These findings reveal a previously unrecognized, proton/carboxylate dual sensing pathway wherein pH and L-malate regulate inflammatory responses, indicating the role of certain carboxylate metabolites as adaptors in the proton biosensing by interactions between macromolecules.
Collapse
Affiliation(s)
- Yu-Jia-Nan Chen
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China.
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA.
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases & Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China.
| | - Rong-Chen Shi
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - Yuan-Cai Xiang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Li Fan
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases & Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Hong Tang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Gang He
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Mei Zhou
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xin-Zhe Feng
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
| | - Jin-Dong Tan
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Pan Huang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xiao Ye
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Kun Zhao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - Wen-Yu Fu
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
- Department of Orthopedics and Rehabilitations, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Liu-Li Li
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
| | - Xu-Ting Bian
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Huan Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Feng Wang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Teng Wang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - Chen-Ke Zhang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Bing-Hua Zhou
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Wan Chen
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Tao-Tao Liang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Jing-Tong Lv
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xia Kang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - You-Xing Shi
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Ellen Kim
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
| | - Yin-Hua Qin
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University, Chongqing, 400038, China
| | - Aubryanna Hettinghouse
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
| | - Kai-di Wang
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
- Department of Medical Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266000, China
| | - Xiang-Li Zhao
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
- Department of Orthopedics and Rehabilitations, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Ming-Yu Yang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Yu-Zhen Tang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Hai-Long Piao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Lin Guo
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| | - Chuan-Ju Liu
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA.
- Department of Orthopedics and Rehabilitations, Yale University School of Medicine, New Haven, CT, 06519, USA.
| | - Hong-Ming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
| | - Kang-Lai Tang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
5
|
Pattison LA, Rickman RH, Hilton H, Dannawi M, Wijesinghe SN, Ladds G, Yang LV, Jones SW, Smith ESJ. Activation of the proton-sensing GPCR, GPR65 on fibroblast-like synoviocytes contributes to inflammatory joint pain. Proc Natl Acad Sci U S A 2024; 121:e2410653121. [PMID: 39661058 PMCID: PMC11665855 DOI: 10.1073/pnas.2410653121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
Inflammation is associated with localized acidosis, however, attributing physiological and pathological roles to proton-sensitive receptors is challenging due to their diversity and widespread expression. Here, agonists of the proton-sensing GPCR, GPR65, were systematically characterized. The synthetic agonist BTB09089 (BTB) recapitulated many proton-induced signaling events and demonstrated selectivity for GPR65. BTB was used to show that GPR65 activation on fibroblast-like synoviocytes (FLS), cells that line synovial joints, results in the secretion of proinflammatory mediators capable of recruiting immune cells and sensitizing sensory neurons. Intra-articular injection of BTB resulted in GPR65-dependent sensitization of knee-innervating neurons and nocifensive behaviors in mice. Stimulation of GPR65 on human FLS also triggered the release of inflammatory mediators and synovial fluid samples from human osteoarthritis patients were shown to activate GPR65. These results suggest a role of GPR65 in mediating cell-cell interactions that drive inflammatory joint pain in both mice and humans.
Collapse
Affiliation(s)
- Luke A. Pattison
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Rebecca H. Rickman
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Helen Hilton
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Maya Dannawi
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Susanne N. Wijesinghe
- Institute of Inflammation and Ageing, University of Birmingham, BirminghamB15 2TT, United Kingdom
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Li V. Yang
- Department of Internal Medicine, Brody School of Medicine at East Carolina University, Greenville, NC27834
| | - Simon W. Jones
- Institute of Inflammation and Ageing, University of Birmingham, BirminghamB15 2TT, United Kingdom
| | - Ewan St. John Smith
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| |
Collapse
|
6
|
Hurtado-Lorenzo A, Swantek JL. The landscape of new therapeutic opportunities for IBD. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 101:1-83. [PMID: 39521596 DOI: 10.1016/bs.apha.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
This chapter presents an overview of the emerging strategies to address the unmet needs in the management of inflammatory bowel diseases (IBD). IBD poses significant challenges, as over half of patients experience disease progression despite interventions, leading to irreversible complications, and a substantial proportion do not respond to existing therapies, such as biologics. To overcome these limitations, we describe a diverse array of novel therapeutic approaches. In the area of immune homeostasis restoration, the focus is on targeting cytokine networks, leukocyte trafficking, novel immune pathways, and cell therapies involving regulatory T cells and mesenchymal stem cells (MSC). Recognizing the critical role of impaired intestinal barrier integrity in IBD, we highlight therapies aimed at restoring barrier function and promoting mucosal healing, such as those targeting cell proliferation, tight junctions, and lipid mediators. Addressing the challenges posed by fibrosis and fistulas, we describe emerging targets for reversing fibrosis like kinase and cytokine inhibitors and nuclear receptor agonists, as well as the potential of MSC for fistulas. The restoration of a healthy gut microbiome, through strategies like fecal microbiota transplantation, rationally defined bacterial consortia, and targeted antimicrobials, is also highlighted. We also describe innovative approaches to gut-targeted drug delivery to enhance efficacy and minimize side effects. Reinforcing these advancements is the critical role of precision medicine, which emphasizes the use of multiomics analysis for the discovery of biomarkers to enable personalized IBD care. Overall, the emerging landscape of therapeutic opportunities for IBD holds great potential to surpass the therapeutic ceiling of current treatments.
Collapse
Affiliation(s)
- Andrés Hurtado-Lorenzo
- Translational Research & IBD Ventures, Research Department, Crohn's & Colitis Foundation, New York, NY, United States.
| | - Jennifer L Swantek
- Translational Research & IBD Ventures, Research Department, Crohn's & Colitis Foundation, New York, NY, United States
| |
Collapse
|
7
|
Dai SP, Yang CC, Chin Y, Sun WH. T cell death-associated gene 8-mediated distinct signaling pathways modulate the early and late phases of neuropathic pain. iScience 2024; 27:110955. [PMID: 39381739 PMCID: PMC11460492 DOI: 10.1016/j.isci.2024.110955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 05/01/2024] [Accepted: 09/10/2024] [Indexed: 10/10/2024] Open
Abstract
Peripheral nerve injury alters the transduction of nociceptive signaling. The coordination of neurons, glia, and immune cells results in persistent pain and inflammation. T cell death-associated gene 8 (TDAG8), located at nociceptors and immune cells, is involved in inflammatory pain and arthritis-induced pain. Here, we employed TDAG8-deficient mice, pharmacological approaches, and calcium/sodium imaging to elucidate how TDAG8-mediated signaling modulates neuron activities in a mouse model of chronic constriction injury-induced neuropathic pain. We demonstrated that TDAG8 participated alone in mechanical allodynia induced by constriction injury. (1) TDAG8-Nav1.8 signaling in small-diameter isolectin B4-positive [IB4(+)] neurons initiates mechanical allodynia; it also modulated substance P release from IB4(-) neurons to facilitate the development of early mechanical allodynia. (2) TDAG8-mediated signaling increased medium-to large-diameter IB4(-) neuron activity to maintain late mechanical allodynia; it also modulated substance P release in soma to reduce satellite glial number and Nav1.7 expression, thus attenuating chronic mechanical allodynia.
Collapse
Affiliation(s)
- Shih-Ping Dai
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Chieh Yang
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Yin Chin
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Hsin Sun
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| |
Collapse
|
8
|
Bagchi S, Yuan R, Huang HL, Zhang W, Chiu DKC, Kim H, Cha SL, Tolentino L, Lowitz J, Liu Y, Moshnikova A, Andreev O, Plevritis S, Engleman EG. The acid-sensing receptor GPR65 on tumor macrophages drives tumor growth in obesity. Sci Immunol 2024; 9:eadg6453. [PMID: 39423285 DOI: 10.1126/sciimmunol.adg6453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 09/19/2024] [Indexed: 10/21/2024]
Abstract
Multiple cancers, including colorectal cancer (CRC), are more frequent and often more aggressive in individuals with obesity. Here, we showed that macrophages accumulated within tumors of patients with obesity and CRC and in obese CRC mice and that they promoted accelerated tumor growth. These changes were initiated by oleic acid accumulation and subsequent tumor cell-derived acid production and were driven by macrophage signaling through the acid-sensing receptor GPR65. We found a similar role for GPR65 in hepatocellular carcinoma (HCC) in obese mice. Tumors in patients with obesity and CRC or HCC also exhibited increased GPR65 expression, suggesting that the mechanism revealed here may contribute to tumor growth in a range of obesity-associated cancers and represent a potential therapeutic target.
Collapse
Affiliation(s)
- Sreya Bagchi
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Robert Yuan
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Han-Li Huang
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
- TMU Research Center for Drug Discovery, Taipei Medical University, Taipei 11031, Taiwan
| | - Weiruo Zhang
- Department of Biological Data Science, Stanford University, Stanford, CA 94305, USA
| | | | - Hyungjoo Kim
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Sophia L Cha
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Lorna Tolentino
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | | | - Yilin Liu
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Anna Moshnikova
- Physics Department, University of Rhode Island, Kingston, RI 02881, USA
| | - Oleg Andreev
- Physics Department, University of Rhode Island, Kingston, RI 02881, USA
| | - Sylvia Plevritis
- Department of Biological Data Science, Stanford University, Stanford, CA 94305, USA
| | - Edgar G Engleman
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
9
|
Zhang SH, Yin J, Jing LJ, Cheng Y, Miao YL, Fan B, Zhang HF, Yang CH, Wang SS, Li Y, Jiao XY, Fan YY. Targeting astrocytic TDAG8 with delayed CO 2 postconditioning improves functional outcomes after controlled cortical impact injury in mice. Exp Neurol 2024; 380:114892. [PMID: 39047809 DOI: 10.1016/j.expneurol.2024.114892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/18/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
T-cell death-associated gene 8 (TDAG8), a G-protein-coupled receptor sensing physiological or weak acids, regulates inflammatory responses. However, its role in traumatic brain injury (TBI) remains unknown. Our recent study showed that delayed CO2 postconditioning (DCPC) has neuroreparative effects after TBI. We hypothesized that activating astrocytic TDAG8 is a key mechanism for DCPC. WT and TDAG8-/- mice received DCPC daily by transiently inhaling 10% CO2 after controlled cortical impact (CCI). HBAAV2/9-GFAP-m-TDAG8-3xflag-EGFP was used to overexpress TDAG8 in astrocytes. The beam walking test, mNSS, immunofluorescence and Golgi-Cox staining were used to evaluate motor function, glial activation and dendritic plasticity. DCPC significantly improved motor function; increased total dendritic length, neuronal complexity and spine density; inhibited overactivation of astrocytes and microglia; and promoted the expression of astrocytic brain-derived neurotrophic factor in WT but not TDAG8-/- mice. Overexpressing TDAG8 in astrocytes surrounding the lesion in TDAG8-/- mice restored the beneficial effects of DCPC. Although the effects of DCPC on Days 14-28 were much weaker than those of DCPC on Days 3-28 in WT mice, these effects were further enhanced by overexpressing astrocytic TDAG8. Astrocytic TDAG8 is a key target of DCPC for TBI rehabilitation. Its overexpression is a strategy that broadens the therapeutic window and enhances the effects of DCPC.
Collapse
Affiliation(s)
- Shu-Han Zhang
- Department of Pharmacology, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Jing Yin
- Department of Pharmacology, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Lian-Ju Jing
- Department of Pharmacology, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Yao Cheng
- Department of Pharmacology, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Yu-Lu Miao
- Department of Pharmacology, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Bo Fan
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Hui-Feng Zhang
- Department of Pharmacology, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Cai-Hong Yang
- Department of Pharmacology, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Shao-Shuai Wang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Yan Li
- Department of Pharmacology, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| | - Xiang-Ying Jiao
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| | - Yan-Ying Fan
- Department of Pharmacology, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
10
|
Justus CR, Marie MA, Sanderlin EJ, Yang LV. The Roles of Proton-Sensing G-Protein-Coupled Receptors in Inflammation and Cancer. Genes (Basel) 2024; 15:1151. [PMID: 39336742 PMCID: PMC11431078 DOI: 10.3390/genes15091151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
The precise regulation of pH homeostasis is crucial for normal physiology. However, in tissue microenvironments, it can be impacted by pathological conditions such as inflammation and cancer. Due to the overproduction and accumulation of acids (protons), the extracellular pH is characteristically more acidic in inflamed tissues and tumors in comparison to normal tissues. A family of proton-sensing G-protein-coupled receptors (GPCRs) has been identified as molecular sensors for cells responding to acidic tissue microenvironments. Herein, we review the current research progress pertaining to these proton-sensing GPCRs, including GPR4, GPR65 (TDAG8), and GPR68 (OGR1), in inflammation and cancer. Growing evidence suggests that GPR4 and GPR68 are mainly pro-inflammatory, whereas GPR65 is primarily anti-inflammatory, in various inflammatory disorders. Both anti- and pro-tumorigenic effects have been reported for this family of receptors. Moreover, antagonists and agonists targeting proton-sensing GPCRs have been developed and evaluated in preclinical models. Further research is warranted to better understand the roles of these proton-sensing GPCRs in pathophysiology and is required in order to exploit them as potential therapeutic targets for disease treatment.
Collapse
Affiliation(s)
- Calvin R Justus
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Mona A Marie
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Edward J Sanderlin
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Li V Yang
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
11
|
Fan J, Liu J, Zhang B, Wang X, Wang X, Liang J, Li Y, Zhang Y, Zhang C, Yu S, Li T, Yang X. GPR65 contributes to constructing immunosuppressive microenvironment in glioma. Neurosurg Rev 2024; 47:417. [PMID: 39123083 PMCID: PMC11315802 DOI: 10.1007/s10143-024-02633-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024]
Abstract
Glioma, especially glioblastoma patients, present highly heterogeneous and immunosuppressive microenvironment, leading to their poor response to treatment and survival. Targeting the tumor microenvironment is considered a promising therapeutic strategy. M2 macrophages are highly infiltrated in glioma tissue, even up to 50% of the total number of bulk tissue cells. Here, we identified GPR65 as the hub gene of the M2 macrophage-related module in glioma through WGCNA analysis. The expression and prognosis analysis suggested that GPR65 was positively correlated with the malignancy and poor prognosis of glioma, and the heterogeneity analysis found that GPR65 was highly expressed in the vascular proliferation area of glioma, which matched the spatial expression characteristics of M2 macrophages. We further verified that GPR65 was highly expressed in macrophages but not tumor cells in the glioma microenvironment by single-cell data analysis and immunofluorescence. Most importantly, we found that inhibition of GPR65 was sufficient to reduce macrophages' polarization response to glioma cell and break the malignant cooperation with glioma cells. Our study reports the expression characteristics and malignant behavior of GPR65 in the glioma microenvironment, which provides a new alternative target of treatment to glioma microenvironment.
Collapse
Affiliation(s)
- Jikang Fan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Jie Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Bin Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Xuya Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Xisen Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Jianshen Liang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Yiming Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Yu Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Chen Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Shengping Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Tao Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China.
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China.
| | - Xuejun Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China.
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China.
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, Beijing, 102200, China.
| |
Collapse
|
12
|
Neale I, Reddy C, Tan ZY, Li B, Nag PP, Park J, Park J, Carey KL, Graham DB, Xavier RJ. Small-molecule probe for IBD risk variant GPR65 I231L alters cytokine signaling networks through positive allosteric modulation. SCIENCE ADVANCES 2024; 10:eadn2339. [PMID: 39028811 PMCID: PMC11259170 DOI: 10.1126/sciadv.adn2339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/13/2024] [Indexed: 07/21/2024]
Abstract
The proton-sensing heterotrimeric guanine nucleotide-binding protein-coupled receptor GPR65 is expressed in immune cells and regulates tissue homeostasis in response to decreased extracellular pH, which occurs in the context of inflammation and tumorigenesis. Genome-wide association studies linked GPR65 to several autoimmune and inflammatory diseases such as multiple sclerosis and inflammatory bowel disease (IBD). The loss-of-function GPR65 I231L IBD risk variant alters cellular metabolism, impairs protective tissue functions, and increases proinflammatory cytokine production. Hypothesizing that a small molecule designed to potentiate GPR65 at subphysiological pH could decrease inflammatory responses, we found positive allosteric modulators of GPR65 that engage and activate both human and mouse orthologs of the receptor. We observed that the chemical probe BRD5075 alters cytokine and chemokine programs in dendritic cells, establishing that immune signaling can be modulated by targeting GPR65. Our investigation offers improved chemical probes to further interrogate the biology of human GPR65 and its clinically relevant genetic variants.
Collapse
Affiliation(s)
- Ilona Neale
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Clark Reddy
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Zher Yin Tan
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Bihua Li
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Partha P. Nag
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Joshua Park
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jihye Park
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Daniel B. Graham
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ramnik J. Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
13
|
Fan YY, Li Y, Tian XY, Wang YJ, Huo J, Guo BL, Chen R, Yang CH, Li Y, Zhang HF, Niu BL, Zhang MS. Delayed Chronic Acidic Postconditioning Improves Poststroke Motor Functional Recovery and Brain Tissue Repair by Activating Proton-Sensing TDAG8. Transl Stroke Res 2024; 15:620-635. [PMID: 36853417 DOI: 10.1007/s12975-023-01143-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/13/2022] [Accepted: 02/17/2023] [Indexed: 03/01/2023]
Abstract
Acidic postconditioning by transient CO2 inhalation applied within minutes after reperfusion has neuroprotective effects in the acute phase of stroke. However, the effects of delayed chronic acidic postconditioning (DCAPC) initiated during the subacute phase of stroke or other acute brain injuries are unknown. Mice received daily DCAPC by inhaling 5%/10%/20% CO2 for various durations (three cycles of 10- or 20-min CO2 inhalation/10-min break) at days 3-7, 7-21, or 3-21 after photothrombotic stroke. Grid-walk, cylinder, and gait tests were used to assess motor function. DCAPC with all CO2 concentrations significantly promoted motor functional recovery, even when DCAPC was delayed for 3-7 days. DCAPC enhanced the puncta density of GAP-43 (a marker of axon growth and regeneration) and synaptophysin (a marker of synaptogenesis) and reduced the amoeboid microglia number, glial scar thickness and mRNA expression of CD16 and CD32 (markers of proinflammatory M1 microglia) compared with those of the stroke group. Cerebral blood flow (CBF) increased in response to DCAPC. Furthermore, the mRNA expression of TDAG8 (a proton-activated G-protein-coupled receptor) was increased during the subacute phase of stroke, while DCAPC effects were blocked by systemic knockout of TDAG8, except for those on CBF. DCAPC reproduced the benefits by re-expressing TDAG8 in the peri-infarct cortex of TDAG8-/- mice infected with HBAAV2/9-CMV-TDAG8-3flag-ZsGreen. Taken together, we first showed that DCAPC promoted functional recovery and brain tissue repair after stroke with a wide therapeutic time window of at least 7 days after stroke. Brain-derived TDAG8 is a direct target of DCAPC that induces neuroreparative effects.
Collapse
Affiliation(s)
- Yan-Ying Fan
- Department of Pharmacology, Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, 030001, China.
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China.
| | - Yu Li
- Department of Pharmacology, Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Xiao-Ying Tian
- Department of Pharmacology, Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Ying-Jing Wang
- Department of Pharmacology, Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Jing Huo
- Department of Pharmacology, Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Bao-Lu Guo
- Department of Pharmacology, Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Ru Chen
- Department of Pharmacology, Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Cai-Hong Yang
- Department of Pharmacology, Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Yan Li
- Department of Pharmacology, Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Hui-Feng Zhang
- Department of Pharmacology, Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Bao-Long Niu
- Department of Pharmacology, Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, 030001, China.
- College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan, 030024, China.
| | - Ming-Sheng Zhang
- Department of Pharmacology, Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
14
|
Li MS, Wang XH, Wang H. Immunomodulation of Proton-activated G Protein-coupled Receptors in Inflammation. Curr Med Sci 2024; 44:475-484. [PMID: 38748372 DOI: 10.1007/s11596-024-2872-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/22/2024] [Indexed: 06/29/2024]
Abstract
Proton-activated G protein-coupled receptors (GPCRs), initially discovered by Ludwig in 2003, are widely distributed in various tissues. These receptors have been found to modulate the immune system in several inflammatory diseases, including inflammatory bowel disease, atopic dermatitis, and asthma. Proton-activated GPCRs belong to the G protein-coupled receptor family and can detect alternations in extracellular pH. This detection triggers downstream signaling pathways within the cells, ultimately influencing the function of immune cells. In this review, we specifically focused on investigating the immune response of proton-activated GPCRs under inflammatory conditions.
Collapse
Affiliation(s)
- Min-Shan Li
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan, 430030, China
| | - Xiang-Hong Wang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan, 430030, China
| | - Heng Wang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan, 430030, China.
| |
Collapse
|
15
|
Otsugu M, Mine A, Uchida I, Miyake Y, Tachihara R, Fujiwara K, Ichimura A, Sato K, Tomura H. Low pH modulates lipopolysaccharide-induced tumor necrosis factor-alpha expression and macropinocytotic activity in RAW264.7 cells. J Recept Signal Transduct Res 2024; 44:63-71. [PMID: 39175331 DOI: 10.1080/10799893.2024.2395310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/12/2024] [Accepted: 08/17/2024] [Indexed: 08/24/2024]
Abstract
Inflammation triggers various types of diseases that need to be addressed. Macrophages play important roles in the inflammatory responses. As atherosclerosis progresses, macrophages transform into foam cells. Extracellular acidification is observed at and around bacterial infection and atherosclerotic sites. However, the effects of acidification on the inflammatory response of macrophages and the progression of atherosclerosis have not been fully understood. This study investigates the impact of extracellular acidification on lipopolysaccharide-induced tumor necrosis factor-alpha (TNF-α) expression and macropinocytotic activity in RAW264.7 cells. TNF-α expression is measured by real-time polymerase chain reaction (relative value to glyceraldehyde-3-phosphate dehydrogenase expression). Macropinocytotic activity is measured by neutral red uptake (absorbance at 540 nm). Results show that TNF-α expression increased with decreasing extracellular pH in both un-foamed and foamed cells. Macropinocytotic activity was upregulated at pH 6.8 in un-foamed cells, but downregulated in foamed cells stimulated at low pH. Proton-sensing G protein-coupled receptors (GPCRs) were involved in the expression of TNF-α and in the macropinocytotic activity of foamed cells. In conclusion, this study reveals that extracellular acidification differently affect various inflammatory responses such as LPS-induced TNF-α expression and macropinocytotic activity of RAW264.7 cells and different proton-sensing GPCRs are involved in the different inflammatory responses.
Collapse
Affiliation(s)
- Miku Otsugu
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Ayumi Mine
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Izumi Uchida
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Yuta Miyake
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Ryo Tachihara
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Kurumi Fujiwara
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Ayako Ichimura
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Koichi Sato
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Hideaki Tomura
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
- Institute of Endocrinology, Meiji University, Kawasaki, Japan
| |
Collapse
|
16
|
Hausmann M, Seuwen K, de Vallière C, Busch M, Ruiz PA, Rogler G. Role of pH-sensing receptors in colitis. Pflugers Arch 2024; 476:611-622. [PMID: 38514581 PMCID: PMC11006753 DOI: 10.1007/s00424-024-02943-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/23/2024]
Abstract
Low pH in the gut is associated with severe inflammation, fibrosis, and colorectal cancer (CRC) and is a hallmark of active inflammatory bowel disease (IBD). Subsequently, pH-sensing mechanisms are of interest for the understanding of IBD pathophysiology. Tissue hypoxia and acidosis-two contributing factors to disease pathophysiology-are linked to IBD, and understanding their interplay is highly relevant for the development of new therapeutic options. One member of the proton-sensing G protein-coupled receptor (GPCR) family, GPR65 (T-cell death-associated gene 8, TDAG8), was identified as a susceptibility gene for IBD in a large genome-wide association study. In response to acidic extracellular pH, GPR65 induces an anti-inflammatory response, whereas the two other proton-sensing receptors, GPR4 and GPR68 (ovarian cancer G protein-coupled receptor 1, OGR1), mediate pro-inflammatory responses. Here, we review the current knowledge on the role of these proton-sensing receptors in IBD and IBD-associated fibrosis and cancer, as well as colitis-associated cancer (CAC). We also describe emerging small molecule modulators of these receptors as therapeutic opportunities for the treatment of IBD.
Collapse
Affiliation(s)
- Martin Hausmann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland.
| | - Klaus Seuwen
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Cheryl de Vallière
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Moana Busch
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Pedro A Ruiz
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091, Zurich, CH, Switzerland
| |
Collapse
|
17
|
Caratis F, Opiełka M, Hausmann M, Velasco-Estevez M, Rojek B, de Vallière C, Seuwen K, Rogler G, Karaszewski B, Rutkowska A. The proton-sensing receptors TDAG8 and GPR4 are differentially expressed in human and mouse oligodendrocytes: Exploring their role in neuroinflammation and multiple sclerosis. PLoS One 2024; 19:e0283060. [PMID: 38527054 PMCID: PMC10962805 DOI: 10.1371/journal.pone.0283060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 02/13/2024] [Indexed: 03/27/2024] Open
Abstract
Acidosis is one of the hallmarks of demyelinating central nervous system (CNS) lesions in multiple sclerosis (MS). The response to acidic pH is primarily mediated by a family of G protein-coupled proton-sensing receptors: OGR1, GPR4 and TDAG8. These receptors are inactive at alkaline pH, reaching maximal activation at acidic pH. Genome-wide association studies have identified a locus within the TDAG8 gene associated with several autoimmune diseases, including MS. Accordingly, we here found that expression of TDAG8, as opposed to GPR4 or OGR1, is upregulated in MS plaques. This led us to investigate the expression of TDAG8 in oligodendrocytes using mouse and human in vitro and in vivo models. We observed significant upregulation of TDAG8 in human MO3.13 oligodendrocytes during maturation and in response to acidic conditions. However, its deficiency did not impact normal myelination in the mouse CNS, and its expression remained unaltered under demyelinating conditions in mouse organotypic cerebellar slices. Notably, our data revealed no expression of TDAG8 in primary mouse oligodendrocyte progenitor cells (OPCs), in contrast to its expression in primary human OPCs. Our investigations have revealed substantial species differences in the expression of proton-sensing receptors in oligodendrocytes, highlighting the limitations of the employed experimental models in fully elucidating the role of TDAG8 in myelination and oligodendrocyte biology. Consequently, the study does not furnish robust evidence for the role of TDAG8 in such processes. Nonetheless, our findings tentatively point towards a potential association between TDAG8 and myelination processes in humans, hinting at a potential link between TDAG8 and the pathophysiology of MS and warrants further research.
Collapse
Affiliation(s)
- Fionä Caratis
- Brain Diseases Centre, Medical University of Gdansk, Gdansk, Poland
- Department of Anatomy and Neurobiology, Medical University of Gdansk, Gdansk, Poland
| | - Mikołaj Opiełka
- Brain Diseases Centre, Medical University of Gdansk, Gdansk, Poland
| | - Martin Hausmann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Maria Velasco-Estevez
- H12O-CNIO Hematological Malignancies Group, Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), Madrid, Spain
| | - Bartłomiej Rojek
- Department of Adult Neurology, Medical University of Gdansk & University Clinical Centre, Gdansk, Poland
| | - Cheryl de Vallière
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Klaus Seuwen
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Bartosz Karaszewski
- Brain Diseases Centre, Medical University of Gdansk, Gdansk, Poland
- Department of Adult Neurology, Medical University of Gdansk & University Clinical Centre, Gdansk, Poland
| | - Aleksandra Rutkowska
- Brain Diseases Centre, Medical University of Gdansk, Gdansk, Poland
- Department of Anatomy and Neurobiology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
18
|
Li G, Lin J, Gao X, Su H, Lin R, Gao H, Feng Z, Wu H, Feng B, Zuo K, Li Y, Wu W, Fang L, Liu Z. Intestinal epithelial pH-sensing receptor GPR65 maintains mucosal homeostasis via regulating antimicrobial defense and restrains gut inflammation in inflammatory bowel disease. Gut Microbes 2023; 15:2257269. [PMID: 37749885 PMCID: PMC10524779 DOI: 10.1080/19490976.2023.2257269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 09/06/2023] [Indexed: 09/27/2023] Open
Abstract
Intestinal epithelial cell (IEC) regulation of barrier function and mucosal homeostasis enables the establishment of a harmonious gut microenvironment. However, host-derived regulatory networks that modulate intestinal antimicrobial defenses have not been fully defined. Herein we generated mice with IEC-specific deletion of Gpr65 (Gpr65ΔIEC) and investigated the role of epithelial GPR65 using DSS- and C. rodentium-induced murine colitis models. RNA sequencing analysis was conducted on colonic IECs from Gpr65fl/fl and Gpr65ΔIEC mice, and colonoids and colonic epithelial cell lines were used to evaluate the pH-sensing effect of GPR65. The expression of GPR65 was determined in IECs from patients with inflammatory bowel disease (IBD) and DSS colitis mice by qRT-PCR, Western blot, and immunohistochemistry, respectively. We observed that the absence of GPR65 in IECs abrogated homeostatic antimicrobial programs, including the production of antimicrobial peptides (AMPs) and defense response-associated proteins. Gpr65ΔIEC mice displayed dysbiosis of the gut microbiota and were prone to DSS- and C. rodentium-induced colitis, as characterized by significantly disrupted epithelial antimicrobial responses, pathogen invasion, and increased inflammatory infiltrates in the inflamed colon. RNA sequencing analysis revealed that deletion of GPR65 in IECs provoked dramatic transcriptome changes with respect to the downregulation of immune and defense responses to bacteria. Forced AMP induction assays conducted in vivo or in ex vivo colonoids revealed that IEC-intrinsic GPR65 signaling drove antimicrobial defense. Mechanistically, GPR65 signaling promoted STAT3 phosphorylation to optimize mucosal defense responses. Epithelial cell line and colonoid assays further confirmed that epithelial GPR65 sensing pH synergized with IL-22 to facilitate antimicrobial responses. Finally, the expression of GPR65 was markedly decreased in the inflamed epithelia of IBD patients and DSS colitis mice. Our findings define an important role of epithelial GPR65 in regulating intestinal homeostasis and mucosal inflammation and point toward a potential therapeutic approach by targeting GPR65 in the treatment of IBD.
Collapse
Affiliation(s)
- Gengfeng Li
- Center for IBD Research, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jian Lin
- Center for IBD Research, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Gastroenterology, Affiliated Hospital of Putian University, Putian, China
| | - Xiang Gao
- Center for IBD Research, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huiling Su
- Department of Gastroenterology, Linfen Central Hospital of Shanxi Medical University, Linfen, China
| | - Ritian Lin
- Center for IBD Research, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Han Gao
- Center for IBD Research, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhongsheng Feng
- Center for IBD Research, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huili Wu
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Baisui Feng
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Keqiang Zuo
- Center for IBD Research, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yingchuan Li
- Center for IBD Research, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wei Wu
- Center for IBD Research, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Leilei Fang
- Center for IBD Research, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhanju Liu
- Center for IBD Research, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
19
|
Zhang K, Zhang MX, Meng XX, Zhu J, Wang JJ, He YF, Li YH, Zhao SC, Shi ZM, Zheng LN, Han T, Hong W. Targeting GPR65 alleviates hepatic inflammation and fibrosis by suppressing the JNK and NF-κB pathways. Mil Med Res 2023; 10:56. [PMID: 38001521 PMCID: PMC10675918 DOI: 10.1186/s40779-023-00494-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND G-protein coupled receptors (GPCRs) are recognized as attractive targets for drug therapy. However, it remains poorly understood how GPCRs, except for a few chemokine receptors, regulate the progression of liver fibrosis. Here, we aimed to reveal the role of GPR65, a proton-sensing receptor, in liver fibrosis and to elucidate the underlying mechanism. METHODS The expression level of GPR65 was evaluated in both human and mouse fibrotic livers. Furthermore, Gpr65-deficient mice were treated with either bile duct ligation (BDL) for 21 d or carbon tetrachloride (CCl4) for 8 weeks to investigate the role of GPR65 in liver fibrosis. A combination of experimental approaches, including Western blotting, quantitative real-time reverse transcription‑polymerase chain reaction (qRT-PCR), and enzyme-linked immunosorbent assay (ELISA), confocal microscopy and rescue studies, were used to explore the underlying mechanisms of GPR65's action in liver fibrosis. Additionally, the therapeutic potential of GPR65 inhibitor in the development of liver fibrosis was investigated. RESULTS We found that hepatic macrophages (HMs)-enriched GPR65 was upregulated in both human and mouse fibrotic livers. Moreover, knockout of Gpr65 significantly alleviated BDL- and CCl4-induced liver inflammation, injury and fibrosis in vivo, and mouse bone marrow transplantation (BMT) experiments further demonstrated that the protective effect of Gpr65 knockout is primarily mediated by bone marrow-derived macrophages (BMMs). Additionally, in vitro data demonstrated that Gpr65 silencing and GPR65 antagonist inhibited, while GPR65 overexpression and application of GPR65 endogenous and exogenous agonists enhanced the expression and release of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and transforming growth factor-β (TGF-β), all of which subsequently promoted the activation of hepatic stellate cells (HSCs) and the damage of hepatocytes (HCs). Mechanistically, GPR65 overexpression, the acidic pH and GPR65 exogenous agonist induced up-regulation of TNF-α and IL-6 via the Gαq-Ca2+-JNK/NF-κB pathways, while promoted the expression of TGF-β through the Gαq-Ca2+-MLK3-MKK7-JNK pathway. Notably, pharmacological GPR65 inhibition retarded the development of inflammation, HCs injury and fibrosis in vivo. CONCLUSIONS GPR65 is a major regulator that modulates the progression of liver fibrosis. Thus, targeting GPR65 could be an effective therapeutic strategy for the prevention of liver fibrosis.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Meng-Xia Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xiao-Xiang Meng
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jing Zhu
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jia-Jun Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yi-Fan He
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ye-Hua Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Si-Cong Zhao
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Zhe-Min Shi
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Li-Na Zheng
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Tao Han
- Department of Hepatology and Gastroenterology, Tianjin Union Medical Center, Tianjin Medical University, Tianjin Union Medical Center affiliated to Nankai University, Tianjin, 300000, China.
| | - Wei Hong
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
20
|
Perren L, Busch M, Schuler C, Ruiz PA, Foti F, Weibel N, de Vallière C, Morsy Y, Seuwen K, Hausmann M, Rogler G. OGR1 (GPR68) and TDAG8 (GPR65) Have Antagonistic Effects in Models of Colonic Inflammation. Int J Mol Sci 2023; 24:14855. [PMID: 37834303 PMCID: PMC10573511 DOI: 10.3390/ijms241914855] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/29/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
G-protein-coupled receptors (GPRs), including pro-inflammatory ovarian cancer GPR1 (OGR1/GPR68) and anti-inflammatory T cell death-associated gene 8 (TDAG8/GPR65), are involved in pH sensing and linked to inflammatory bowel disease (IBD). OGR1 and TDAG8 show opposite effects. To determine which effect is predominant or physiologically more relevant, we deleted both receptors in models of intestinal inflammation. Combined Ogr1 and Tdag8 deficiency was assessed in spontaneous and acute murine colitis models. Disease severity was assessed using clinical scores. Colon samples were analyzed using quantitative polymerase chain reaction (qPCR) and flow cytometry (FACS). In acute colitis, Ogr1-deficient mice showed significantly decreased clinical scores compared with wildtype (WT) mice, while Tdag8-deficient mice and double knockout (KO) mice presented similar scores to WT. In Il-10-spontaneous colitis, Ogr1-deficient mice presented significantly decreased, and Tdag8-deficient mice had increased inflammation. In the Il10-/- × Ogr1-/- × Tdag8-/- triple KO mice, inflammation was significantly decreased compared with Tdag8-/-. Absence of Ogr1 reduced pro-inflammatory cytokines in Tdag8-deficient mice. Tdag8-/- had significantly more IFNγ+ T-lymphocytes and IL-23 T-helper cells in the colon compared with WT. The absence of OGR1 significantly alleviates the intestinal damage mediated by the lack of functional TDAG8. Both OGR1 and TDAG8 represent potential new targets for therapeutic intervention.
Collapse
|
21
|
Hajjar S, Zhou X. pH sensing at the intersection of tissue homeostasis and inflammation. Trends Immunol 2023; 44:807-825. [PMID: 37714775 PMCID: PMC10543622 DOI: 10.1016/j.it.2023.08.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/13/2023] [Accepted: 08/13/2023] [Indexed: 09/17/2023]
Abstract
pH is tightly maintained at cellular, tissue, and systemic levels, and altered pH - particularly in the acidic range - is associated with infection, injury, solid tumors, and physiological and pathological inflammation. However, how pH is sensed and regulated and how it influences immune responses remain poorly understood at the tissue level. Applying conceptual frameworks of homeostatic and inflammatory circuitries, we categorize cellular and tissue components engaged in pH regulation, drawing parallels from established cases in physiology. By expressing various intracellular (pHi) and extracellular pH (pHe)-sensing receptors, the immune system may integrate information on tissue and cellular states into the regulation of homeostatic and inflammatory programs. We introduce the novel concept of resistance and adaptation responses to rationalize pH-dependent immunomodulation intertwined with homeostatic equilibrium and inflammatory control. We discuss emerging challenges and opportunities in understanding the immunological roles of pH sensing, which might reveal new strategies to combat inflammation and restore tissue homeostasis.
Collapse
Affiliation(s)
- Stephanie Hajjar
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, 300 Longwood Ave, Boston, MA 02115, USA
| | - Xu Zhou
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, 300 Longwood Ave, Boston, MA 02115, USA.
| |
Collapse
|
22
|
Whittington AM, Turner FS, Baark F, Templeman S, Kirwan DE, Roufosse C, Krishnan N, Robertson BD, Chong DLW, Porter JC, Gilman RH, Friedland JS. An acidic microenvironment in Tuberculosis increases extracellular matrix degradation by regulating macrophage inflammatory responses. PLoS Pathog 2023; 19:e1011495. [PMID: 37418488 PMCID: PMC10355421 DOI: 10.1371/journal.ppat.1011495] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 07/19/2023] [Accepted: 06/20/2023] [Indexed: 07/09/2023] Open
Abstract
Mycobacterium tuberculosis (M.tb) infection causes marked tissue inflammation leading to lung destruction and morbidity. The inflammatory extracellular microenvironment is acidic, however the effect of this acidosis on the immune response to M.tb is unknown. Using RNA-seq we show that acidosis produces system level transcriptional change in M.tb infected human macrophages regulating almost 4000 genes. Acidosis specifically upregulated extracellular matrix (ECM) degradation pathways with increased expression of Matrix metalloproteinases (MMPs) which mediate lung destruction in Tuberculosis. Macrophage MMP-1 and -3 secretion was increased by acidosis in a cellular model. Acidosis markedly suppresses several cytokines central to control of M.tb infection including TNF-α and IFN-γ. Murine studies demonstrated expression of known acidosis signaling G-protein coupled receptors OGR-1 and TDAG-8 in Tuberculosis which are shown to mediate the immune effects of decreased pH. Receptors were then demonstrated to be expressed in patients with TB lymphadenitis. Collectively, our findings show that an acidic microenvironment modulates immune function to reduce protective inflammatory responses and increase extracellular matrix degradation in Tuberculosis. Acidosis receptors are therefore potential targets for host directed therapy in patients.
Collapse
Affiliation(s)
| | - Frances S. Turner
- Edinburgh Genomics, University of Edinburgh, Edinburgh, United Kingdom
| | - Friedrich Baark
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Sam Templeman
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Daniela E. Kirwan
- Institute of Infection and Immunity, St. George’s, University of London, London, United Kingdom
| | - Candice Roufosse
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Nitya Krishnan
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Brian D. Robertson
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Deborah L. W. Chong
- Institute of Infection and Immunity, St. George’s, University of London, London, United Kingdom
| | - Joanna C. Porter
- Centre for Inflammation & Tissue Repair, Respiratory Medicine, University College London, London, United Kingdom
| | - Robert H. Gilman
- Department of International Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jon S. Friedland
- Institute of Infection and Immunity, St. George’s, University of London, London, United Kingdom
| |
Collapse
|
23
|
Xie L, Alam MJ, Marques FZ, Mackay CR. A major mechanism for immunomodulation: Dietary fibres and acid metabolites. Semin Immunol 2023; 66:101737. [PMID: 36857894 DOI: 10.1016/j.smim.2023.101737] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 01/17/2023] [Accepted: 02/09/2023] [Indexed: 03/01/2023]
Abstract
Diet and the gut microbiota have a profound influence on physiology and health, however, mechanisms are still emerging. Here we outline several pathways that gut microbiota products, particularly short-chain fatty acids (SCFAs), use to maintain gut and immune homeostasis. Dietary fibre is fermented by the gut microbiota in the colon, and large quantities of SCFAs such as acetate, propionate, and butyrate are produced. Dietary fibre and SCFAs enhance epithelial integrity and thereby limit systemic endotoxemia. Moreover, SCFAs inhibit histone deacetylases (HDAC), and thereby affect gene transcription. SCFAs also bind to 'metabolite-sensing' G-protein coupled receptors (GPCRs) such as GPR43, which promotes immune homeostasis. The enormous amounts of SCFAs produced in the colon are sufficient to lower pH, which affects the function of proton sensors such as GPR65 expressed on the gut epithelium and immune cells. GPR65 is an anti-inflammatory Gαs-coupled receptor, which leads to the inhibition of inflammatory cytokines. The importance of GPR65 in inflammatory diseases is underscored by genetics associated with the missense variant I231L (rs3742704), which is associated with human inflammatory bowel disease, atopic dermatitis, and asthma. There is enormous scope to manipulate these pathways using specialized diets that release very high amounts of specific SCFAs in the gut, and we believe that therapies that rely on chemically modified foods is a promising approach. Such an approach includes high SCFA-producing diets, which we have shown to decrease numerous inflammatory western diseases in mouse models. These diets operate at many levels - increased gut integrity, changes to the gut microbiome, and promotion of immune homeostasis, which represents a new and highly promising way to prevent or treat human disease.
Collapse
Affiliation(s)
- Liang Xie
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Hypertension Research Laboratory, School of Biological Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Md Jahangir Alam
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Clayton, VIC 3800, Australia; Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne,VIC 3004, Australia
| | - Charles R Mackay
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China.
| |
Collapse
|
24
|
von Breitenbuch P, Kurz B, Wallner S, Zeman F, Brochhausen C, Schlitt HJ, Schreml S. Expression of pH-Sensitive GPCRs in Peritoneal Carcinomatosis of Colorectal Cancer-First Results. J Clin Med 2023; 12:jcm12051803. [PMID: 36902589 PMCID: PMC10003041 DOI: 10.3390/jcm12051803] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/06/2023] [Accepted: 02/20/2023] [Indexed: 03/05/2023] Open
Abstract
Solid tumors have an altered metabolism with a so-called inside-out pH gradient (decreased pHe < increased pHi). This also signals back to tumor cells via proton-sensitive ion channels or G protein-coupled receptors (pH-GPCRs) to alter migration and proliferation. Nothing, however, is known about the expression of pH-GPCRs in the rare form of peritoneal carcinomatosis. Paraffin-embedded tissue samples of a series of 10 patients with peritoneal carcinomatosis of colorectal (including appendix) origin were used for immunohistochemistry to study the expression of GPR4, GPR65, GPR68, GPR132, and GPR151. GPR4 was just expressed weakly in 30% of samples and expression was significantly reduced as compared to GPR56, GPR132, and GPR151. Furthermore, GPR68 was only expressed in 60% of tumors and showed significantly reduced expression as compared to GPR65 and GPR151. This is the first study on pH-GPCRs in peritoneal carcinomatosis, which shows lower expression of GPR4 and GPR68 as compared to other pH-GPCRs in this type of cancer. It may give rise to future therapies targeting either the TME or these GPCRs directly.
Collapse
Affiliation(s)
| | - Bernadett Kurz
- Department of Dermatology, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Susanne Wallner
- Department of Dermatology, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Florian Zeman
- Center for Clinical Studies, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Christoph Brochhausen
- Institute of Pathology, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Hans-Jürgen Schlitt
- Department of Surgery, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Stephan Schreml
- Department of Dermatology, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
- Correspondence:
| |
Collapse
|
25
|
Stolwijk JA, Wallner S, Heider J, Kurz B, Pütz L, Michaelis S, Goricnik B, Erl J, Frank L, Berneburg M, Haubner F, Wegener J, Schreml S. GPR4 in the pH-dependent migration of melanoma cells in the tumor microenvironment. Exp Dermatol 2022; 32:479-490. [PMID: 36562556 DOI: 10.1111/exd.14735] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 11/17/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
Due to its high metastatic potential, malignant melanoma is one of the deadliest skin cancers. In melanoma as well as in other cancers, acidification of the tumor microenvironment (=TME, inverse pH-gradient) is a well-known driver of tumor progression and metastasis. Membrane-bound receptors, such as the proton-sensitive GPCR (pH-GPCR) GPR4, are considered as potential initiators of the signalling cascades relevant to malignant transformation. In this study, we investigated the pH-dependent migration of GPR4 wildtype/overexpressing SK-Mel-28 cells using an impedance-based electrical wounding and migration assay and classical Boyden chamber experiments. Migration of GPR4 overexpressing SK-Mel-28 cells was enhanced in a range of pH 6.5-7.5 as compared to controls in the impedance-based electrical wounding and migration assay. In Boyden chamber experiments, GPR4 overexpression only increased migration at pH 7.5 in a Matrigel-free setup, but not at pH 6.5. Results indicate that GPR4 is involved in the migration of melanoma cells, especially in the tumor periphery, and that this process is affected by pH in the TME.
Collapse
Affiliation(s)
- Judith Anthea Stolwijk
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany.,Faculty of Chemistry and Pharmacy, Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany
| | - Susanne Wallner
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| | - Judith Heider
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| | - Bernadett Kurz
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| | - Lisa Pütz
- Faculty of Chemistry and Pharmacy, Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany
| | - Stefanie Michaelis
- Faculty of Chemistry and Pharmacy, Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany.,Fraunhofer Research Institution for Microsystems and Solid State Technologies EMFT, Regensburg, Germany
| | - Barbara Goricnik
- Faculty of Chemistry and Pharmacy, Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany
| | - Julia Erl
- Faculty of Chemistry and Pharmacy, Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany
| | - Linda Frank
- Faculty of Chemistry and Pharmacy, Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany
| | - Mark Berneburg
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| | - Frank Haubner
- Department of Otorhinolaryngology, Ludwig Maximilians University Munich, Munich, Germany
| | - Joachim Wegener
- Faculty of Chemistry and Pharmacy, Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany.,Fraunhofer Research Institution for Microsystems and Solid State Technologies EMFT, Regensburg, Germany
| | - Stephan Schreml
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| |
Collapse
|
26
|
Araviiskaia E, Pincelli C, Sparavigna A, Luger T. The Role of a Novel Generation of Emollients, 'Emollients Plus', in Atopic Dermatitis. Clin Cosmet Investig Dermatol 2022; 15:2705-2719. [PMID: 36545500 PMCID: PMC9763050 DOI: 10.2147/ccid.s389697] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/26/2022] [Indexed: 12/15/2022]
Abstract
Emollients are the mainstay maintenance treatment for atopic dermatitis (AD). A novel generation of emollients, 'emollients plus', containing active, non-medicated substances, has softened the distinction between emollients and topical drugs. A literature search for selected key words was performed using PubMed. Additional papers were identified based on author expertise. Whilst the inclusion of five components of an ideal emollient has been proposed, no such consensus exists for emollients plus and they can vary markedly in their composition and modes of action for AD treatment. This could have a profound effect on their clinical efficacy. The efficacy of emollients plus in restoring and maintaining skin barrier function has been demonstrated on multiple levels, with evidence reported for their effects on the physical and biochemical, microbial, immunological, and neurosensory barriers. When selecting an appropriate AD treatment approach, the safety profiles of the available topical therapies must be carefully considered. There are several proposed treatment approaches for AD, including preventive, proactive, intermittent, and synergistic approaches. Emollients plus may be effective not only as maintenance therapy for AD, but also when used synergistically with anti-inflammatory pharmacological therapies.
Collapse
Affiliation(s)
- Elena Araviiskaia
- Department of Dermatology and Venereal Diseases, First Pavlov State Medical University of St Petersburg, St Petersburg, Russia
| | - Carlo Pincelli
- DermoLab, Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Adele Sparavigna
- Derming Clinical Research and Bioengineering Institute, Milan, Italy
| | - Thomas Luger
- Department of Dermatology, University of Munster, Munster, Germany,Correspondence: Thomas Luger, Dermatology Clinic, University of Münster, Von-Esmarch-Straẞe 58, Münster, 48149, Germany, Email
| |
Collapse
|
27
|
Deai M, Oya R, Saso N, Tanaka A, Uchida I, Miyake Y, Tachihara R, Otsugu M, Mine A, Sato K, Tomura H. Ethylenediaminetetraacetic acid (EDTA) enhances cAMP production in human TDAG8-expressing cells. Biochem Biophys Res Commun 2022; 626:15-20. [PMID: 35964552 DOI: 10.1016/j.bbrc.2022.07.110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/02/2022]
Abstract
Ethylenediaminetetraacetic acid (EDTA) is a chelating agent that binds tightly to metal ions. We found that cAMP response element (CRE)-driven promoter activity by protons was enhanced by EDTA in human T-cell death-associated gene 8 (TDAG8)-overexpressed HEK293T cells. The enhancing action by EDTA was also detected by proton-induced cAMP production that is located upstream from the CRE-driven promoter activity even at physiological proton concentration pH7.4. The proton-induced CRE-driven promoter activity was not enhanced by other chelating agents, ethylene glycol tetraacetic acid (EGTA) and sodium citrate. The enhanced CRE-driven promoter activity by EDTA was not attenuated by increasing the extracellular calcium ion concentration. These results indicate that the EDTA-enhancing action may not be due to its chelating action but might rather be another EDTA-specific effect. Enhanced cAMP production by EDTA was also detected in a human leukemia cell line HL-60, in which TDAG8 and OGR1 (ovarian cancer G-protein-coupled receptor 1) were endogenously expressed, suggesting that the medical use of EDTA would influence the physiological and pathophysiological functions of hematopoietic cells.
Collapse
Affiliation(s)
- Masahito Deai
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan
| | - Rin Oya
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan
| | - Naosi Saso
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan
| | - Asahi Tanaka
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan
| | - Izumi Uchida
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan
| | - Yuta Miyake
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan
| | - Ryo Tachihara
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan
| | - Miku Otsugu
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan
| | - Ayumi Mine
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan
| | - Koichi Sato
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, 371-8512, Japan
| | - Hideaki Tomura
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan; Institute of Endocrinology, Meiji University, Kawasaki, 214-8571, Japan.
| |
Collapse
|
28
|
Zha XM, Xiong ZG, Simon RP. pH and proton-sensitive receptors in brain ischemia. J Cereb Blood Flow Metab 2022; 42:1349-1363. [PMID: 35301897 PMCID: PMC9274858 DOI: 10.1177/0271678x221089074] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/11/2022] [Accepted: 02/28/2022] [Indexed: 01/01/2023]
Abstract
Extracellular proton concentration is at 40 nM when pH is 7.4. In disease conditions such as brain ischemia, proton concentration can reach µM range. To respond to this increase in extracellular proton concentration, the mammalian brain expresses at least three classes of proton receptors. Acid-sensing ion channels (ASICs) are the main neuronal cationic proton receptor. The proton-activated chloride channel (PAC), which is also known as (aka) acid-sensitive outwardly rectifying anion channel (ASOR; TMEM206), mediates acid-induced chloride currents. Besides proton-activated channels, GPR4, GPR65 (aka TDAG8, T-cell death-associated gene 8), and GPR68 (aka OGR1, ovarian cancer G protein-coupled receptor 1) function as proton-sensitive G protein-coupled receptors (GPCRs). Though earlier studies on these GPCRs mainly focus on peripheral cells, we and others have recently provided evidence for their functional importance in brain injury. Specifically, GPR4 shows strong expression in brain endothelium, GPR65 is present in a fraction of microglia, while GPR68 exhibits predominant expression in brain neurons. Here, to get a better view of brain acid signaling and its contribution to ischemic injury, we will review the recent findings regarding the differential contribution of proton-sensitive GPCRs to cerebrovascular function, neuroinflammation, and neuronal injury following acidosis and brain ischemia.
Collapse
Affiliation(s)
- Xiang-ming Zha
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Zhi-Gang Xiong
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Roger P Simon
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
29
|
Chen X, Jaiswal A, Costliow Z, Herbst P, Creasey EA, Oshiro-Rapley N, Daly MJ, Carey KL, Graham DB, Xavier RJ. pH sensing controls tissue inflammation by modulating cellular metabolism and endo-lysosomal function of immune cells. Nat Immunol 2022; 23:1063-1075. [PMID: 35668320 PMCID: PMC9720675 DOI: 10.1038/s41590-022-01231-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 04/26/2022] [Indexed: 02/08/2023]
Abstract
Extracellular acidification occurs in inflamed tissue and the tumor microenvironment; however, a systematic study on how pH sensing contributes to tissue homeostasis is lacking. In the present study, we examine cell type-specific roles of the pH sensor G protein-coupled receptor 65 (GPR65) and its inflammatory disease-associated Ile231Leu-coding variant in inflammation control. GPR65 Ile231Leu knock-in mice are highly susceptible to both bacterial infection-induced and T cell-driven colitis. Mechanistically, GPR65 Ile231Leu elicits a cytokine imbalance through impaired helper type 17 T cell (TH17 cell) and TH22 cell differentiation and interleukin (IL)-22 production in association with altered cellular metabolism controlled through the cAMP-CREB-DGAT1 axis. In dendritic cells, GPR65 Ile231Leu elevates IL-12 and IL-23 release at acidic pH and alters endo-lysosomal fusion and degradation capacity, resulting in enhanced antigen presentation. The present study highlights GPR65 Ile231Leu as a multistep risk factor in intestinal inflammation and illuminates a mechanism by which pH sensing controls inflammatory circuits and tissue homeostasis.
Collapse
Affiliation(s)
- Xiangjun Chen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Experimental Medicine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alok Jaiswal
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Paula Herbst
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Elizabeth A Creasey
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Noriko Oshiro-Rapley
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Experimental Medicine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark J Daly
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Institute for Molecular Medicine Finland, Helsinki, Finland
| | | | - Daniel B Graham
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Experimental Medicine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA.
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
30
|
Imenez Silva PH, Câmara NO, Wagner CA. Role of proton-activated G protein-coupled receptors in pathophysiology. Am J Physiol Cell Physiol 2022; 323:C400-C414. [PMID: 35759438 DOI: 10.1152/ajpcell.00114.2022] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Local acidification is a common feature of many disease processes such as inflammation, infarction, or solid tumor growth. Acidic pH is not merely a sequelae of disease but contributes to recruitment and regulation of immune cells, modifies metabolism of parenchymal, immune and tumor cells, modulates fibrosis, vascular permeability, oxygen availability and consumption, invasiveness of tumor cells, and impacts on cell survival. Thus, multiple pH-sensing mechanisms must exist in cells involved in these processes. These pH-sensors play important roles in normal physiology and pathophysiology, and hence might be attractive targets for pharmacological interventions. Among the pH-sensing mechanisms, OGR1 (GPR68), GPR4 (GPR4), and TDAG8 (GPR65) have emerged as important molecules. These G protein-coupled receptors are widely expressed, are upregulated in inflammation and tumors, sense changes in extracellular pH in the range between pH 8 and 6, and are involved in modulating key processes in inflammation, tumor biology, and fibrosis. This review discusses key features of these receptors and highlights important disease states and pathways affected by their activity.
Collapse
Affiliation(s)
- Pedro H Imenez Silva
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,National Center of Competence in Research NCCR Kidney.CH, Switzerland
| | - Niels Olsen Câmara
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,National Center of Competence in Research NCCR Kidney.CH, Switzerland
| |
Collapse
|
31
|
Christou H, Khalil RA. Mechanisms of pulmonary vascular dysfunction in pulmonary hypertension and implications for novel therapies. Am J Physiol Heart Circ Physiol 2022; 322:H702-H724. [PMID: 35213243 PMCID: PMC8977136 DOI: 10.1152/ajpheart.00021.2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 12/21/2022]
Abstract
Pulmonary hypertension (PH) is a serious disease characterized by various degrees of pulmonary vasoconstriction and progressive fibroproliferative remodeling and inflammation of the pulmonary arterioles that lead to increased pulmonary vascular resistance, right ventricular hypertrophy, and failure. Pulmonary vascular tone is regulated by a balance between vasoconstrictor and vasodilator mediators, and a shift in this balance to vasoconstriction is an important component of PH pathology, Therefore, the mainstay of current pharmacological therapies centers on pulmonary vasodilation methodologies that either enhance vasodilator mechanisms such as the NO-cGMP and prostacyclin-cAMP pathways and/or inhibit vasoconstrictor mechanisms such as the endothelin-1, cytosolic Ca2+, and Rho-kinase pathways. However, in addition to the increased vascular tone, many patients have a "fixed" component in their disease that involves altered biology of various cells in the pulmonary vascular wall, excessive pulmonary artery remodeling, and perivascular fibrosis and inflammation. Pulmonary arterial smooth muscle cell (PASMC) phenotypic switch from a contractile to a synthetic and proliferative phenotype is an important factor in pulmonary artery remodeling. Although current vasodilator therapies also have some antiproliferative effects on PASMCs, they are not universally successful in halting PH progression and increasing survival. Mild acidification and other novel approaches that aim to reverse the resident pulmonary vascular pathology and structural remodeling and restore a contractile PASMC phenotype could ameliorate vascular remodeling and enhance the responsiveness of PH to vasodilator therapies.
Collapse
Affiliation(s)
- Helen Christou
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
32
|
Imenez Silva PH, Wagner CA. Physiological relevance of proton-activated GPCRs. Pflugers Arch 2022; 474:487-504. [PMID: 35247105 PMCID: PMC8993716 DOI: 10.1007/s00424-022-02671-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022]
Abstract
The detection of H+ concentration variations in the extracellular milieu is accomplished by a series of specialized and non-specialized pH-sensing mechanisms. The proton-activated G protein-coupled receptors (GPCRs) GPR4 (Gpr4), TDAG8 (Gpr65), and OGR1 (Gpr68) form a subfamily of proteins capable of triggering intracellular signaling in response to alterations in extracellular pH around physiological values, i.e., in the range between pH 7.5 and 6.5. Expression of these receptors is widespread for GPR4 and OGR1 with particularly high levels in endothelial cells and vascular smooth muscle cells, respectively, while expression of TDAG8 appears to be more restricted to the immune compartment. These receptors have been linked to several well-studied pH-dependent physiological activities including central control of respiration, renal adaption to changes in acid-base status, secretion of insulin and peripheral responsiveness to insulin, mechanosensation, and cellular chemotaxis. Their role in pathological processes such as the genesis and progression of several inflammatory diseases (asthma, inflammatory bowel disease), and tumor cell metabolism and invasiveness, is increasingly receiving more attention and makes these receptors novel and interesting targets for therapy. In this review, we cover the role of these receptors in physiological processes and will briefly discuss some implications for disease processes.
Collapse
Affiliation(s)
- Pedro H Imenez Silva
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
- National Center of Competence in Research NCCR Kidney.CH, Zurich, Switzerland.
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
- National Center of Competence in Research NCCR Kidney.CH, Zurich, Switzerland.
| |
Collapse
|
33
|
IBD-associated G protein-coupled receptor 65 variant compromises signalling and impairs key functions involved in inflammation. Cell Signal 2022; 93:110294. [PMID: 35218908 PMCID: PMC9536022 DOI: 10.1016/j.cellsig.2022.110294] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/06/2022] [Accepted: 02/21/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Inflammatory bowel diseases (IBD) result in chronic inflammation of the gastrointestinal tract. Genetic studies have shown that the GPR65 gene, as well as its missense coding variant, GPR65*Ile231Leu, is associated with IBD. We aimed to define the signalling and biological pathways downstream of GPR65 activation and evaluate the impact of GPR65*231Leu on these. METHODS We used HEK 293 cells stably expressing GPR65 and deficient for either Gαs, Gαq/11 or Gα12/13, to define GPR65 signalling pathways, IBD patient biopsies and a panel of human tissues, primary immune cells and cell lines to determine biologic context, and genetic modulation of human THP-1-derived macrophages to examine the impact of GPR65 in bacterial phagocytosis and NLRP3 inflammasome activation. RESULTS We confirmed that GPR65 signals via the Gαs pathway, leading to cAMP accumulation. GPR65 can also signal via the Gα12/13 pathway leading to formation of stress fibers, actin remodeling and RhoA activation; all impaired by the IBD-associated GPR65*231Leu allele. Gene expression profiling revealed greater expression of GPR65 in biopsies from inflamed compared to non-inflamed tissues from IBD patients or control individuals, potentially explained by infiltration of inflammatory immune cells. Decreased GPR65 expression in THP-1-derived macrophages leads to impaired bacterial phagocytosis, increased NLRP3 inflammasome activation and IL-1β secretion in response to an inflammatory stimulus. CONCLUSIONS We demonstrate that GPR65 exerts its effects through Gαs- and Gα12/13-mediated pathways, that the IBD-associated GPR65*231Leu allele has compromised interactions with Gα12/13 and that KD of GPR65 leads to impaired bacterial phagocytosis and increased inflammatory signalling via the NLRP3 inflammasome. This work identifies a target for development of small molecule therapies.
Collapse
|
34
|
Jang KB, You MJ, Yang B, Rim C, Kim HJ, Kwon MS. Persistent Acidic Environment Induces Impaired Phagocytosis via ERK in Microglia. Neurochem Res 2022; 47:1341-1353. [PMID: 35103911 DOI: 10.1007/s11064-022-03533-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/23/2021] [Accepted: 01/17/2022] [Indexed: 12/28/2022]
Abstract
Acidic environment evoked by stroke, traumatic brain injury, and Alzheimer's disease may change the functional properties of microglia. Nevertheless, the underlying mechanisms of functional changes in microglia remain unclear. In this study, we found that acidic stimuli (pH 6.8) increased rapidly interleukin (IL)-1β and IL-6 mRNA levels and subsequently reduced IL-10, transforming growth factor (TGF)-β1, Cx3cr1, and P2ry12 as the exposure time to acidic environment increase in BV2 cells. In addition, persistent acidic environment (pH 6.8 for 6 h) induced impaired phagocytic function in BV2 cells. Short-term acidic exposure (pH 6.8 for 30 min) increased cyclic AMP (cAMP) and phospho-protein kinase A (PKA) but inhibited phospho-extracellular signal-regulated kinase (p-ERK). However, under persistent acidic environment (pH 6.8 for 6 h), cyclic AMP and PKA were normalized and p-ERK was increased with TDAG8 (T cell death associated gene 8; GPR65) reduction. FR 180,204, an ERK inhibitor, rescued the persistent acidic environment-induced functional changes in BV2 cells and its effect was recapitulated in primary neonatal microglia. Thus, we propose that ERK targeting may be an alternative strategy to restore microglial dysfunction in the central nervous system (CNS) acidic environment in various neurological disorders.
Collapse
Affiliation(s)
- Kyu-Beom Jang
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA BIO COMPLEX, CHA University, 335 Pangyo, Bundang-gu, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Min-Jung You
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA BIO COMPLEX, CHA University, 335 Pangyo, Bundang-gu, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Bohyun Yang
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA BIO COMPLEX, CHA University, 335 Pangyo, Bundang-gu, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Chan Rim
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA BIO COMPLEX, CHA University, 335 Pangyo, Bundang-gu, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Hui-Ju Kim
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA BIO COMPLEX, CHA University, 335 Pangyo, Bundang-gu, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Min-Soo Kwon
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA BIO COMPLEX, CHA University, 335 Pangyo, Bundang-gu, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea.
| |
Collapse
|
35
|
Manosalva C, Quiroga J, Hidalgo AI, Alarcón P, Ansoleaga N, Hidalgo MA, Burgos RA. Role of Lactate in Inflammatory Processes: Friend or Foe. Front Immunol 2022; 12:808799. [PMID: 35095895 PMCID: PMC8795514 DOI: 10.3389/fimmu.2021.808799] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022] Open
Abstract
During an inflammatory process, shift in the cellular metabolism associated with an increase in extracellular acidification are well-known features. This pH drop in the inflamed tissue is largely attributed to the presence of lactate by an increase in glycolysis. In recent years, evidence has accumulated describing the role of lactate in inflammatory processes; however, there are differences as to whether lactate can currently be considered a pro- or anti-inflammatory mediator. Herein, we review these recent advances on the pleiotropic effects of lactate on the inflammatory process. Taken together, the evidence suggests that lactate could exert differential effects depending on the metabolic status, cell type in which the effects of lactate are studied, and the pathological process analyzed. Additionally, various targets, including post-translational modifications, G-protein coupled receptor and transcription factor activation such as NF-κB and HIF-1, allow lactate to modulate signaling pathways that control the expression of cytokines, chemokines, adhesion molecules, and several enzymes associated with immune response and metabolism. Altogether, this would explain its varied effects on inflammatory processes beyond its well-known role as a waste product of metabolism.
Collapse
Affiliation(s)
- Carolina Manosalva
- Faculty of Sciences, Institute of Pharmacy, Universidad Austral de Chile, Valdivia, Chile
| | - John Quiroga
- Laboratory of Immunometabolism, Faculty of Veterinary Sciences, Institute of Pharmacology and Morphophysiology, Universidad Austral de Chile, Valdivia, Chile
- Graduate School, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Alejandra I. Hidalgo
- Laboratory of Immunometabolism, Faculty of Veterinary Sciences, Institute of Pharmacology and Morphophysiology, Universidad Austral de Chile, Valdivia, Chile
| | - Pablo Alarcón
- Laboratory of Immunometabolism, Faculty of Veterinary Sciences, Institute of Pharmacology and Morphophysiology, Universidad Austral de Chile, Valdivia, Chile
| | - Nicolás Ansoleaga
- Laboratory of Immunometabolism, Faculty of Veterinary Sciences, Institute of Pharmacology and Morphophysiology, Universidad Austral de Chile, Valdivia, Chile
- Graduate School, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - María Angélica Hidalgo
- Laboratory of Immunometabolism, Faculty of Veterinary Sciences, Institute of Pharmacology and Morphophysiology, Universidad Austral de Chile, Valdivia, Chile
| | - Rafael Agustín Burgos
- Laboratory of Immunometabolism, Faculty of Veterinary Sciences, Institute of Pharmacology and Morphophysiology, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
36
|
Weder B, Schefer F, van Haaften WT, Patsenker E, Stickel F, Mueller S, Hutter S, Schuler C, Baebler K, Wang Y, Mamie C, Dijkstra G, de Vallière C, Imenez Silva PH, Wagner CA, Frey-Wagner I, Ruiz PA, Seuwen K, Rogler G, Hausmann M. New Therapeutic Approach for Intestinal Fibrosis Through Inhibition of pH-Sensing Receptor GPR4. Inflamm Bowel Dis 2022; 28:109-125. [PMID: 34320209 DOI: 10.1093/ibd/izab140] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Patients suffering from inflammatory bowel diseases (IBDs) express increased mucosal levels of pH-sensing receptors compared with non-IBD controls. Acidification leads to angiogenesis and extracellular matrix remodeling. We aimed to determine the expression of pH-sensing G protein-coupled receptor 4 (GPR4) in fibrotic lesions in Crohn's disease (CD) patients. We further evaluated the effect of deficiency in Gpr4 or its pharmacologic inhibition. METHODS Paired samples from fibrotic and nonfibrotic terminal ileum were obtained from CD patients undergoing ileocaecal resection. The effects of Gpr4 deficiency were assessed in the spontaneous Il-10-/- and the chronic dextran sodium sulfate (DSS) murine colitis model. The effects of Gpr4 deficiency and a GPR4 antagonist (39c) were assessed in the heterotopic intestinal transplantation model. RESULTS In human terminal ileum, increased expression of fibrosis markers was accompanied by an increase in GPR4 expression. A positive correlation between the expression of procollagens and GPR4 was observed. In murine disease models, Gpr4 deficiency was associated with a decrease in angiogenesis and fibrogenesis evidenced by decreased vessel length and expression of Edn, Vegfα, and procollagens. The heterotopic animal model for intestinal fibrosis, transplanted with terminal ileum from Gpr4-/- mice, revealed a decrease in mRNA expression of fibrosis markers and a decrease in collagen content and layer thickness compared with grafts from wild type mice. The GPR4 antagonist decreased collagen deposition. The GPR4 expression was also observed in human and murine intestinal fibroblasts. The GPR4 inhibition reduced markers of fibroblast activation stimulated by low pH, notably Acta2 and cTgf. CONCLUSIONS Expression of GPR4 positively correlates with the expression of profibrotic genes and collagen. Deficiency of Gpr4 is associated with a decrease in angiogenesis and fibrogenesis. The GPR4 antagonist decreases collagen deposition. Targeting GPR4 with specific inhibitors may constitute a new treatment option for IBD-associated fibrosis.
Collapse
Affiliation(s)
- Bruce Weder
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Fabian Schefer
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Wouter Tobias van Haaften
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Eleonora Patsenker
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Felix Stickel
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Sebastian Mueller
- Department of Internal Medicine and Center for Alcohol Research, Salem Medical Center University Hospital Heidelberg, Heidelberg, Germany
| | - Senta Hutter
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Cordelia Schuler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Katharina Baebler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Yu Wang
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Céline Mamie
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Cheryl de Vallière
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Pedro H Imenez Silva
- Institute of Physiology, University of Zurich, Zurich, Switzerland and National Center of Competence in Research Kidney Control of Homeostasis, Switzerland
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland and National Center of Competence in Research Kidney Control of Homeostasis, Switzerland
| | - Isabelle Frey-Wagner
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Pedro A Ruiz
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Klaus Seuwen
- Novartis Institutes for Biomedical Research, Forum1 Novartis Campus, Basel, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Martin Hausmann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
37
|
GPR65 (TDAG8) inhibits intestinal inflammation and colitis-associated colorectal cancer development in experimental mouse models. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166288. [PMID: 34628032 PMCID: PMC8629932 DOI: 10.1016/j.bbadis.2021.166288] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 02/06/2023]
Abstract
GPR65 (TDAG8) is a proton-sensing G protein-coupled receptor predominantly expressed in immune cells. Genome-wide association studies (GWAS) have identified GPR65 gene polymorphisms as an emerging risk factor for the development of inflammatory bowel disease (IBD). Patients with IBD have an elevated risk of developing colorectal cancer when compared to the general population. To study the role of GPR65 in intestinal inflammation and colitis-associated colorectal cancer (CAC), colitis and CAC were induced in GPR65 knockout (KO) and wild-type (WT) mice using dextran sulfate sodium (DSS) and azoxymethane (AOM)/DSS, respectively. Disease severity parameters such as fecal score, colon shortening, histopathology, and mesenteric lymph node enlargement were aggravated in GPR65 KO mice compared to WT mice treated with DSS. Elevated leukocyte infiltration and fibrosis were observed in the inflamed colon of GPR65 KO when compared to WT mice which may represent a cellular mechanism for the observed exacerbation of intestinal inflammation. In line with high expression of GPR65 in infiltrated leukocytes, GPR65 gene expression was increased in inflamed intestinal tissue samples of IBD patients compared to normal intestinal tissues. Moreover, colitis-associated colorectal cancer development was higher in GPR65 KO mice than WT mice when treated with AOM/DSS. Altogether, our data demonstrate that GPR65 suppresses intestinal inflammation and colitis-associated tumor development in murine colitis and CAC models, suggesting potentiation of GPR65 with agonists may have an anti-inflammatory therapeutic effect in IBD and reduce the risk of developing colitis-associated colorectal cancer.
Collapse
|
38
|
Park I, Phan TM, Fang J. Novel Molecular Mechanism of Lenalidomide in Myeloid Malignancies Independent of Deletion of Chromosome 5q. Cancers (Basel) 2021; 13:5084. [PMID: 34680233 PMCID: PMC8534127 DOI: 10.3390/cancers13205084] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 09/30/2021] [Accepted: 10/08/2021] [Indexed: 12/26/2022] Open
Abstract
Lenalidomide as well as other immunomodulatory drugs (IMiDs) have achieved clinical efficacies in certain sub-types of hematologic malignancies, such as multiple myeloma, lower-risk myelodysplastic syndromes (MDS) with a single deletion of chromosome 5q (del(5q)) and others. Despite superior clinical response to lenalidomide in hematologic malignancies, relapse and resistance remains a problem in IMiD-based therapy. The last ten years have witnessed the discovery of novel molecular mechanism of IMiD-based anti-tumor therapy. IMiDs bind human cereblon (CRBN), the substrate receptor of the CRL4 E3 ubiquitin ligase complex. Binding of CRBN with IMiDs leads to degradation of the Ikaros family zinc finger proteins 1 and 3 (IKZF1 and IKZF3) and casein kinase 1 alpha. We have found that lenalidomide-mediated degradation of IKZF1 leads to activation of the G protein-coupled receptor 68 (GPR68)/calcium/calpain pro-apoptotic pathway and inhibition of the regulator of calcineurin 1 (RCAN1)/calcineurin pro-survival pathway in MDS and acute myeloid leukemia (AML). Calcineurin inhibitor Cyclosporin-A potentiates the anti-leukemia activity of lenalidomide in MDS/AML with or without del(5q). These findings broaden the therapeutic potential of IMiDs. This review summarizes novel molecular mechanism of lenalidomide in myeloid malignancies, especially without del(5q), in the hope to highlight novel therapeutic targets.
Collapse
Affiliation(s)
| | | | - Jing Fang
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA; (I.P.); (T.M.P.)
| |
Collapse
|
39
|
Sisignano M, Fischer MJM, Geisslinger G. Proton-Sensing GPCRs in Health and Disease. Cells 2021; 10:cells10082050. [PMID: 34440817 PMCID: PMC8392051 DOI: 10.3390/cells10082050] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 12/17/2022] Open
Abstract
The group of proton-sensing G-protein coupled receptors (GPCRs) consists of the four receptors GPR4, TDAG8 (GPR65), OGR1 (GPR68), and G2A (GPR132). These receptors are cellular sensors of acidification, a property that has been attributed to the presence of crucial histidine residues. However, the pH detection varies considerably among the group of proton-sensing GPCRs and ranges from pH of 5.5 to 7.8. While the proton-sensing GPCRs were initially considered to detect acidic cellular environments in the context of inflammation, recent observations have expanded our knowledge about their physiological and pathophysiological functions and many additional individual and unique features have been discovered that suggest a more differentiated role of these receptors in health and disease. It is known that all four receptors contribute to different aspects of tumor biology, cardiovascular physiology, and asthma. However, apart from their overlapping functions, they seem to have individual properties, and recent publications identify potential roles of individual GPCRs in mechanosensation, intestinal inflammation, oncoimmunological interactions, hematopoiesis, as well as inflammatory and neuropathic pain. Here, we put together the knowledge about the biological functions and structural features of the four proton-sensing GPCRs and discuss the biological role of each of the four receptors individually. We explore all currently known pharmacological modulators of the four receptors and highlight potential use. Finally, we point out knowledge gaps in the biological and pharmacological context of proton-sensing GPCRs that should be addressed by future studies.
Collapse
Affiliation(s)
- Marco Sisignano
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany;
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Correspondence:
| | - Michael J. M. Fischer
- Center for Physiology and Pharmacology, Institute of Physiology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria;
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany;
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
40
|
Xie L, McKenzie CI, Qu X, Mu Y, Wang Q, Bing N, Naidoo K, Alam MJ, Yu D, Gong F, Ang C, Robert R, Marques FZ, Furlotte N, Hinds D, Gasser O, Xavier RJ, Mackay CR. pH and Proton Sensor GPR65 Determine Susceptibility to Atopic Dermatitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:101-109. [PMID: 34135065 PMCID: PMC8674371 DOI: 10.4049/jimmunol.2001363] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/15/2021] [Indexed: 12/15/2022]
Abstract
pH sensing by GPR65 regulates various inflammatory conditions, but its role in skin remains unknown. In this study, we performed a phenome-wide association study and report that the T allele of GPR65-intronic single-nucleotide polymorphism rs8005161, which reduces GPR65 signaling, showed a significant association with atopic dermatitis, in addition to inflammatory bowel diseases and asthma, as previously reported. Consistent with this genetic association in humans, we show that deficiency of GPR65 in mice resulted in markedly exacerbated disease in the MC903 experimental model of atopic dermatitis. Deficiency of GPR65 also increased neutrophil migration in vitro. Moreover, GPR65 deficiency in mice resulted in higher expression of the inflammatory cytokine TNF-α by T cells. In humans, CD4+ T cells from rs8005161 heterozygous individuals expressed higher levels of TNF-α after PMA/ionomycin stimulation, particularly under pH 6 conditions. pH sensing by GPR65 appears to be important for regulating the pathogenesis of atopic dermatitis.
Collapse
Affiliation(s)
- Liang Xie
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| | - Craig I McKenzie
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Xinyan Qu
- School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology, Shandong Academy of Sciences, Jinan, China
| | - Yan Mu
- School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology, Shandong Academy of Sciences, Jinan, China
| | - Quanbo Wang
- School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology, Shandong Academy of Sciences, Jinan, China
| | | | - Karmella Naidoo
- Malaghan Institute of Medical Research, Victoria University of Wellington, Wellington, New Zealand
| | - Md Jahangir Alam
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Di Yu
- School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology, Shandong Academy of Sciences, Jinan, China
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Fang Gong
- Department of Laboratory Medicine, Wuxi Hospital of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Caroline Ang
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Remy Robert
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Clayton, Victoria, Australia
- Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | | | - Olivier Gasser
- Malaghan Institute of Medical Research, Victoria University of Wellington, Wellington, New Zealand
| | - Ramnik J Xavier
- Broad Institute, MA
- Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA; and
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA
| | - Charles R Mackay
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia;
- School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology, Shandong Academy of Sciences, Jinan, China
| |
Collapse
|
41
|
Sato K, Tobo A, Mogi C, Tobo M, Yamane N, Tosaka M, Tomura H, Im DS, Okajima F. The protective role of proton-sensing TDAG8 in the brain injury in a mouse ischemia reperfusion model. Sci Rep 2020; 10:17193. [PMID: 33057165 PMCID: PMC7566628 DOI: 10.1038/s41598-020-74372-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 09/30/2020] [Indexed: 01/09/2023] Open
Abstract
Extracellular acidification in the brain has been observed in ischemia; however, the physiological and pathophysiological implications of the pH reduction remain largely unknown. Here, we analyzed the roles of proton-sensing G protein-coupled receptors, including T-cell death-associated gene 8 (TDAG8), ovarian cancer G protein-coupled receptor 1 (OGR1), and G protein-coupled receptor 4 (GPR4) in a mouse ischemia reperfusion model. Cerebral infarction and dysfunctional behavior with transient middle cerebral artery occlusion (tMCAO) and subsequent reperfusion were exacerbated by the deficiency of TDAG8, whereas no significant effect was observed with the deficiency of OGR1 or GPR4. We confirmed that the pH of the predicted infarction region was 6.5. TDAG8 mRNA was observed in Iba1-positive microglia in the mouse brain. The tMCAO increased the mRNA expression of tumor necrosis factor-α in the ipsilateral cerebral hemisphere and evoked morphological changes in microglia in an evolving cerebral injury. These tMCAO-induced actions were significantly enhanced by the TDAG8 deficiency. Administration of minocycline, which is known to inhibit microglial activation, improved the cerebral infarction and dysfunctional behavior induced by tMCAO in the TDAG8-deficient mouse. Thus, acidic pH/TDAG8 protects against cerebral infarction caused by tMCAO, at least due to the mechanism involving the inhibition of microglial functions.
Collapse
Affiliation(s)
- Koichi Sato
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, 371-8512, Japan.
| | - Ayaka Tobo
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, 371-8512, Japan
| | - Chihiro Mogi
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, 371-8512, Japan
| | - Masayuki Tobo
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, 371-8512, Japan
| | - Nobuhiro Yamane
- Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Masahiko Tosaka
- Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Hideaki Tomura
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan
| | - Dong-Soon Im
- College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Fumikazu Okajima
- Laboratory of Signal Transduction, Faculty of Pharmaceutical Sciences, Aomori University, Aomori, 030-0943, Japan
| |
Collapse
|
42
|
Klatt W, Wallner S, Brochhausen C, Stolwijk JA, Schreml S. Expression profiles of proton-sensing G-protein coupled receptors in common skin tumors. Sci Rep 2020; 10:15327. [PMID: 32948783 PMCID: PMC7501253 DOI: 10.1038/s41598-020-71700-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 08/19/2020] [Indexed: 12/18/2022] Open
Abstract
The proton-sensing GPCRs (pH-GPCRs) GPR4 (GPR19), TDAG8 (GPR65, T-cell death associated gene 8), OGR1 (GPR68, ovarian cancer GPCR1), and G2A (GPR132, G2 accumulation protein) are involved in sensing and transducing changes in extracellular pH (pHe). Extracellular acidification is a central hallmark of solid cancer. pH-GPCR function has been associated with cancer cell proliferation, adhesion, migration and metastasis, as well as with modulation of the immune system. Little is known about the expression levels and role of pH-GPCRs in skin cancer. To better understand the functions of pH-GPCRs in skin cancer in vivo, we examined the expression-profiles of GPR4, TDAG8, OGR1 and G2A in four common skin tumors, i.e. squamous cell carcinoma (SCC), malignant melanoma (MM), compound nevus cell nevi (NCN), basal cell carcinoma (BCC). We performed immunohistochemistry and immunofluorescence staining on paraffin-embedded tissue samples acquired from patients suffering from SCC, MM, NCN or BCC. We show the expression of pH-GPCRs in four common skin cancers. Different expression patterns in the investigated skin cancer types indicate that the different pH-GPCRs may have distinct functions in tumor progression and serve as novel therapeutic targets.
Collapse
Affiliation(s)
- Wybke Klatt
- Department of Dermatology, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Susanne Wallner
- Department of Dermatology, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Christoph Brochhausen
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Judith A Stolwijk
- Department of Dermatology, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
- Institute of Analytical Chemistry, Chemo- and Biosensors, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Stephan Schreml
- Department of Dermatology, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
43
|
Dai SP, Hsieh WS, Chen CH, Lu YH, Huang HS, Chang DM, Huang SL, Sun WH. TDAG8 deficiency reduces satellite glial number and pro-inflammatory macrophage number to relieve rheumatoid arthritis disease severity and chronic pain. J Neuroinflammation 2020; 17:170. [PMID: 32471455 PMCID: PMC7257243 DOI: 10.1186/s12974-020-01851-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 05/21/2020] [Indexed: 11/12/2022] Open
Abstract
Background The autoimmune disease rheumatoid arthritis (RA) affects approximately 1% of the global population. RA is characterized with chronic joint inflammation and often associated with chronic pain. The imbalance of pro-inflammatory and anti-inflammatory macrophages is a feature of RA progression. Glial cells affecting neuronal sensitivity at both peripheral and central levels may also be important for RA progression and associated pain. Genetic variants in the T cell death-associated gene 8 (TDAG8) locus are found to associate with spondyloarthritis. TDAG8 was also found involved in RA disease progression and associated hyperalgesia in the RA mouse model. However, its modulation in RA remains unclear. Methods To address this question, we intra-articularly injected complete Freund’s adjuvant (CFA) into TDAG8+/+, TDAG8−/− or wild-type mice, followed by pain behavioral tests. Joints and dorsal root ganglia were taken, sectioned, and stained with antibodies to observe the number of immune cells, macrophages, and satellite glial cells (SGCs). For compound treatments, compounds were intraperitoneally or orally administered weekly for 9 consecutive weeks after CFA injection. Results We demonstrated that TDAG8 deletion slightly reduced RA pain in the early phase but dramatically attenuated RA progression and pain in the chronic phase (> 7 weeks). TDAG8 deletion inhibited an increase in SGC number and inhibition of SGC function attenuated chronic phase of RA pain, so TDAG8 could regulate SGC number to control chronic pain. TDAG8 deletion also reduced M1 pro-inflammatory macrophage number at 12 weeks, contributing to the attenuation of chronic RA pain. Such results were further confirmed by using salicylanilide derivatives, CCL-2d or LCC-09, to suppress TDAG8 expression and function. Conclusions This study demonstrates that TDAG8 deletion reduced SGC and M1 macrophage number to relieve RA disease severity and associated chronic pain. M1 macrophages are critical for the development and maintenance of RA disease and pain, but glial activation is also required for the chronic phase of RA pain.
Collapse
Affiliation(s)
- Shih-Ping Dai
- Department of Life Sciences, National Central University, Jhongli, Taoyuan City, Taiwan
| | - Wei-Shan Hsieh
- Department of Life Sciences, National Central University, Jhongli, Taoyuan City, Taiwan
| | - Chien-Hua Chen
- Department of Life Sciences, National Central University, Jhongli, Taoyuan City, Taiwan
| | - Yueh-Hao Lu
- Department of Life Sciences, National Central University, Jhongli, Taoyuan City, Taiwan
| | - Hsu-Shan Huang
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Der-Ming Chang
- Division of Allergy, Immunology, Rheumatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shir-Ly Huang
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Wei-Hsin Sun
- Department of Life Sciences, National Central University, Jhongli, Taoyuan City, Taiwan. .,Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
44
|
Kung CC, Dai SP, Chiang H, Huang HS, Sun WH. Temporal expression patterns of distinct cytokines and M1/M2 macrophage polarization regulate rheumatoid arthritis progression. Mol Biol Rep 2020; 47:3423-3437. [PMID: 32277445 DOI: 10.1007/s11033-020-05422-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 04/03/2020] [Indexed: 02/06/2023]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammation of synovial joints and often associated with chronic pain. Chronic joint inflammation is attributed to severe proliferation of synoviocytes and resident macrophages and infiltration of immune cells. These cells secrete pro-inflammatory cytokines such as tumor necrosis factor α (TNF-α), interleukin 6 (IL-6) and IL-17 to overcome actions of anti-inflammatory cytokines, thereby maintaining chronic inflammation and pain. The imbalance between pro-inflammatory cytokines (produced by M1 macrophages) and anti-inflammatory cytokines (produced by M2 macrophages) is a feature of RA progression, but the switch time of M1/M2 polarization and which receptor regulates the switch remain unsolved. Here we used an established RA mouse model to demonstrate that TNF-α expression was responsible for the initial acute stage of inflammation and pain (1-4 weeks), IL-17 expression the transition stage (4-12 weeks), and IL-6 expression the later maintenance stage (> 12 weeks). The switch time of M1/M2 polarization occurred at 4-8 weeks. We also identified a potential compound, anthra[2,1-c][1,2,5] thiadiazole-6,11-dione (NSC745885), that specifically inhibited T-cell death-associated gene 8 (TDAG8) function and expression. NSC745885 decreased joint inflammation and destruction and attenuated pain by reducing cytokine production and regulating the M1/M2 polarization switch. TDAG8 may participate in regulating the M1/M2 polarization and temporal expression of distinct cytokines to control RA progression.
Collapse
Affiliation(s)
- Chia-Chi Kung
- Department of Life Sciences, Zhongli District, National Central University, Taoyuan City, Taiwan.,Division of Anesthesiology, Fu Jen Catholic University Hospital, New Taipei City, Taiwan.,School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Shih-Ping Dai
- Department of Life Sciences, Zhongli District, National Central University, Taoyuan City, Taiwan
| | - Hao Chiang
- Department of Life Sciences, Zhongli District, National Central University, Taoyuan City, Taiwan
| | - Hsu-Shan Huang
- Graduated Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei City, Taiwan
| | - Wei-Hsin Sun
- Department of Life Sciences, Zhongli District, National Central University, Taoyuan City, Taiwan. .,Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, No. 155, Sec2, Linong Street, Taipei, 112, Taiwan.
| |
Collapse
|
45
|
Ward C, Meehan J, Gray ME, Murray AF, Argyle DJ, Kunkler IH, Langdon SP. The impact of tumour pH on cancer progression: strategies for clinical intervention. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:71-100. [PMID: 36046070 PMCID: PMC9400736 DOI: 10.37349/etat.2020.00005] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Dysregulation of cellular pH is frequent in solid tumours and provides potential opportunities for therapeutic intervention. The acidic microenvironment within a tumour can promote migration, invasion and metastasis of cancer cells through a variety of mechanisms. Pathways associated with the control of intracellular pH that are under consideration for intervention include carbonic anhydrase IX, the monocarboxylate transporters (MCT, MCT1 and MCT4), the vacuolar-type H+-ATPase proton pump, and the sodium-hydrogen exchanger 1. This review will describe progress in the development of inhibitors to these targets.
Collapse
Affiliation(s)
- Carol Ward
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| | - James Meehan
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| | - Mark E Gray
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG Midlothian, UK
| | - Alan F Murray
- School of Engineering, Institute for Integrated Micro and Nano Systems, EH9 3JL Edinburgh, UK
| | - David J Argyle
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG Midlothian, UK
| | - Ian H Kunkler
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| | - Simon P Langdon
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| |
Collapse
|
46
|
Zeng Z, Mukherjee A, Varghese AP, Yang XL, Chen S, Zhang H. Roles of G protein-coupled receptors in inflammatory bowel disease. World J Gastroenterol 2020; 26:1242-1261. [PMID: 32256014 PMCID: PMC7109274 DOI: 10.3748/wjg.v26.i12.1242] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/18/2020] [Accepted: 03/05/2020] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a complex disease with multiple pathogenic factors. Although the pathogenesis of IBD is still unclear, a current hypothesis suggests that genetic susceptibility, environmental factors, a dysfunctional immune system, the microbiome, and the interactions of these factors substantially contribute to the occurrence and development of IBD. Although existing and emerging drugs have been proven to be effective in treating IBD, none can cure IBD permanently. G protein-coupled receptors (GPCRs) are critical signaling molecules implicated in the immune response, cell proliferation, inflammation regulation and intestinal barrier maintenance. Breakthroughs in the understanding of the structures and functions of GPCRs have provided a driving force for exploring the roles of GPCRs in the pathogenesis of diseases, thereby leading to the development of GPCR-targeted medication. To date, a number of GPCRs have been shown to be associated with IBD, significantly advancing the drug discovery process for IBD. The associations between GPCRs and disease activity, disease severity, and disease phenotypes have also paved new avenues for the precise management of patients with IBD. In this review, we mainly focus on the roles of the most studied proton-sensing GPCRs, cannabinoid receptors, and estrogen-related GPCRs in the pathogenesis of IBD and their potential clinical values in IBD and some other diseases.
Collapse
Affiliation(s)
- Zhen Zeng
- Department of Gastroenterology, Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 410061, Sichuan Province, China
| | - Arjudeb Mukherjee
- West China School of Medicine, Sichuan University, Chengdu 410061, Sichuan Province, China
| | | | - Xiao-Li Yang
- Department of Gastroenterology, Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 410061, Sichuan Province, China
| | - Sha Chen
- Department of Gastroenterology, Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 410061, Sichuan Province, China
| | - Hu Zhang
- Department of Gastroenterology, Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 410061, Sichuan Province, China
| |
Collapse
|
47
|
Maeyashiki C, Melhem H, Hering L, Baebler K, Cosin-Roger J, Schefer F, Weder B, Hausmann M, Scharl M, Rogler G, de Vallière C, Ruiz PA. Activation of pH-Sensing Receptor OGR1 (GPR68) Induces ER Stress Via the IRE1α/JNK Pathway in an Intestinal Epithelial Cell Model. Sci Rep 2020; 10:1438. [PMID: 31996710 PMCID: PMC6989664 DOI: 10.1038/s41598-020-57657-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 12/31/2019] [Indexed: 12/15/2022] Open
Abstract
Proton-sensing ovarian cancer G-protein coupled receptor (OGR1) plays an important role in pH homeostasis. Acidosis occurs at sites of intestinal inflammation and can induce endoplasmic reticulum (ER) stress and the unfolded protein response (UPR), an evolutionary mechanism that enables cells to cope with stressful conditions. ER stress activates autophagy, and both play important roles in gut homeostasis and contribute to the pathogenesis of inflammatory bowel disease (IBD). Using a human intestinal epithelial cell model, we investigated whether our previously observed protective effects of OGR1 deficiency in experimental colitis are associated with a differential regulation of ER stress, the UPR and autophagy. Caco-2 cells stably overexpressing OGR1 were subjected to an acidic pH shift. pH-dependent OGR1-mediated signalling led to a significant upregulation in the ER stress markers, binding immunoglobulin protein (BiP) and phospho-inositol required 1α (IRE1α), which was reversed by a novel OGR1 inhibitor and a c-Jun N-terminal kinase (JNK) inhibitor. Proton-activated OGR1-mediated signalling failed to induce apoptosis, but triggered accumulation of total microtubule-associated protein 1 A/1B-light chain 3, suggesting blockage of late stage autophagy. Our results show novel functions for OGR1 in the regulation of ER stress through the IRE1α-JNK signalling pathway, as well as blockage of autophagosomal degradation. OGR1 inhibition might represent a novel therapeutic approach in IBD.
Collapse
Affiliation(s)
- Chiaki Maeyashiki
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Hassan Melhem
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Larissa Hering
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Katharina Baebler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Jesus Cosin-Roger
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Fabian Schefer
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Bruce Weder
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Martin Hausmann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, Zurich, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, Zurich, Switzerland
| | - Cheryl de Vallière
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland.
| | - Pedro A Ruiz
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
48
|
Pattison LA, Callejo G, St John Smith E. Evolution of acid nociception: ion channels and receptors for detecting acid. Philos Trans R Soc Lond B Biol Sci 2019; 374:20190291. [PMID: 31544616 PMCID: PMC6790391 DOI: 10.1098/rstb.2019.0291] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2019] [Indexed: 12/13/2022] Open
Abstract
Nociceptors, i.e. sensory neurons tuned to detect noxious stimuli, are found in numerous phyla of the Animalia kingdom and are often polymodal, responding to a variety of stimuli, e.g. heat, cold, pressure and chemicals, such as acid. Owing to the ability of protons to have a profound effect on ionic homeostasis and damage macromolecular structures, it is no wonder that the ability to detect acid is conserved across many species. To detect changes in pH, nociceptors are equipped with an assortment of different acid sensors, some of which can detect mild changes in pH, such as the acid-sensing ion channels, proton-sensing G protein-coupled receptors and several two-pore potassium channels, whereas others, such as the transient receptor potential vanilloid 1 ion channel, require larger shifts in pH. This review will discuss the evolution of acid sensation and the different mechanisms by which nociceptors can detect acid. This article is part of the Theo Murphy meeting issue 'Evolution of mechanisms and behaviour important for pain'.
Collapse
Affiliation(s)
| | | | - Ewan St John Smith
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| |
Collapse
|
49
|
McMurray KMJ, Vollmer LL, Ahlbrand R, Thomas J, Winter A, Lewkowich IP, Sah R. Immunomodulatory T cell death associated gene-8 (TDAG8) receptor in depression-associated behaviors. Physiol Behav 2019; 209:112598. [PMID: 31271833 DOI: 10.1016/j.physbeh.2019.112598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/26/2019] [Accepted: 06/29/2019] [Indexed: 12/27/2022]
Abstract
Converging evidence supports neuroimmune factors in depression psychopathology. We previously reported reduced depression-like behavior in immunomodulatory G-protein-coupled receptor, T cell death-associated gene-8 (TDAG8) deficient mice. Here, we expand on those findings by investigating depression- and anxiety-associated behaviors, and cytokine profiles in TDAG8-deficient mice. TDAG8-deficiency reduced depression- and anxiety-associated behaviors in the forced swim test (FST), open-field test and elevated zero maze. Interestingly, cytokine expression, particularly IL-6, was attenuated within hippocampus and spleen in TDAG8-deficient mice following the FST. There were no differences in immune-cell frequencies. Collectively, these data suggest a contributory role of TDAG8 in neuroimmune regulation and depression-associated physiology.
Collapse
Affiliation(s)
- Katherine M J McMurray
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Lauren Larke Vollmer
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rebecca Ahlbrand
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joshua Thomas
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Andrew Winter
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Neuroscience Graduate Program, University of Cincinnati, Medical Sciences Building, Room 1058B, 231 Albert Sabin Way, Cincinnati, OH 45237, USA
| | - Ian P Lewkowich
- Division of Immunobiology, Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Renu Sah
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Neuroscience Graduate Program, University of Cincinnati, Medical Sciences Building, Room 1058B, 231 Albert Sabin Way, Cincinnati, OH 45237, USA; VA Medical Center, Cincinnati, OH 45237, USA
| |
Collapse
|
50
|
Hudalla H, Michael Z, Christodoulou N, Willis GR, Fernandez-Gonzalez A, Filatava EJ, Dieffenbach P, Fredenburgh LE, Stearman RS, Geraci MW, Kourembanas S, Christou H. Carbonic Anhydrase Inhibition Ameliorates Inflammation and Experimental Pulmonary Hypertension. Am J Respir Cell Mol Biol 2019; 61:512-524. [PMID: 30951642 PMCID: PMC6775956 DOI: 10.1165/rcmb.2018-0232oc] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 04/02/2019] [Indexed: 01/07/2023] Open
Abstract
Inflammation and vascular smooth muscle cell (VSMC) phenotypic switching are causally linked to pulmonary arterial hypertension (PAH) pathogenesis. Carbonic anhydrase inhibition induces mild metabolic acidosis and exerts protective effects in hypoxic pulmonary hypertension. Carbonic anhydrases and metabolic acidosis are further known to modulate immune cell activation. To evaluate if carbonic anhydrase inhibition modulates macrophage activation, inflammation, and VSMC phenotypic switching in severe experimental pulmonary hypertension, pulmonary hypertension was assessed in Sugen 5416/hypoxia (SU/Hx) rats after treatment with acetazolamide or ammonium chloride (NH4Cl). We evaluated pulmonary and systemic inflammation and characterized the effect of carbonic anhydrase inhibition and metabolic acidosis in alveolar macrophages and bone marrow-derived macrophages (BMDMs). We further evaluated the treatment effects on VSMC phenotypic switching in pulmonary arteries and pulmonary artery smooth muscle cells (PASMCs) and corroborated some of our findings in lungs and pulmonary arteries of patients with PAH. Both patients with idiopathic PAH and SU/Hx rats had increased expression of lung inflammatory markers and signs of PASMC dedifferentiation in pulmonary arteries. Acetazolamide and NH4Cl ameliorated SU/Hx-induced pulmonary hypertension and blunted pulmonary and systemic inflammation. Expression of carbonic anhydrase isoform 2 was increased in alveolar macrophages from SU/Hx animals, classically (M1) and alternatively (M2) activated BMDMs, and lungs of patients with PAH. Carbonic anhydrase inhibition and acidosis had distinct effects on M1 and M2 markers in BMDMs. Inflammatory cytokines drove PASMC dedifferentiation, and this was inhibited by acetazolamide and acidosis. The protective antiinflammatory effect of acetazolamide in pulmonary hypertension is mediated by a dual mechanism of macrophage carbonic anhydrase inhibition and systemic metabolic acidosis.
Collapse
MESH Headings
- Acetazolamide/therapeutic use
- Acidosis/chemically induced
- Acidosis/complications
- Acidosis/immunology
- Ammonium Chloride/therapeutic use
- Animals
- Carbonic Anhydrase Inhibitors/therapeutic use
- Carbonic Anhydrases/physiology
- Cell Differentiation/drug effects
- Contractile Proteins/biosynthesis
- Contractile Proteins/genetics
- Drug Evaluation, Preclinical
- Humans
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/enzymology
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/pathology
- Hypoxia/complications
- Inflammation
- Macrophages/drug effects
- Macrophages/enzymology
- Macrophages, Alveolar/drug effects
- Macrophages, Alveolar/enzymology
- Male
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Protein Isoforms/antagonists & inhibitors
- Pulmonary Artery/pathology
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Rats
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- Hannes Hudalla
- Department of Pediatric Newborn Medicine and
- Department of Neonatology, Heidelberg University Children’s Hospital, Heidelberg, Germany
- Harvard Medical School, Boston, Massachusetts
| | - Zoe Michael
- Department of Pediatric Newborn Medicine and
- Harvard Medical School, Boston, Massachusetts
| | | | - Gareth R. Willis
- Harvard Medical School, Boston, Massachusetts
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, Massachusetts; and
| | - Angeles Fernandez-Gonzalez
- Harvard Medical School, Boston, Massachusetts
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, Massachusetts; and
| | | | - Paul Dieffenbach
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Laura E. Fredenburgh
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Robert S. Stearman
- Division of Pulmonary, Critical Care Medicine, Sleep, and Occupational Medicine, Department of Medicine, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Mark W. Geraci
- Division of Pulmonary, Critical Care Medicine, Sleep, and Occupational Medicine, Department of Medicine, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Stella Kourembanas
- Department of Pediatric Newborn Medicine and
- Harvard Medical School, Boston, Massachusetts
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, Massachusetts; and
| | - Helen Christou
- Department of Pediatric Newborn Medicine and
- Harvard Medical School, Boston, Massachusetts
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, Massachusetts; and
| |
Collapse
|