1
|
Pu D, Wang P, Wang X, Tian Y, Gong H, Ma X, Li M, Zhang D. Focusing on non-responders to infliximab with ulcerative colitis, what can we do first and next? Int Immunopharmacol 2024; 141:112943. [PMID: 39191122 DOI: 10.1016/j.intimp.2024.112943] [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: 05/20/2024] [Revised: 07/31/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic immune-mediated inflammation of the colorectum, for which infliximab (IFX) is currently the mainstay of treatment. However, one-third of patients with UC still fail to benefit from the IFX therapy, and early exposure to IFX impairs the efficacy of other subsequent biologics. Therefore, personalized therapeutic system is urgently needed to assist in clinical decision-making and precision treatment. METHODS Four microarray datasets of colonic biopsies from UC patients treated with IFX were obtained from the GEO database to form the Training Cohort and Validation Cohort. Differentially expressed genes (DEGs) in Training Cohort were identified and enriched for GO, KEGG and immune cell infiltration analysis. A prediction model for IFX efficacy was developed based on the LASSO and Logistic regression. The predictive accuracy of the model was verified by the Validation Cohort, and the model-genes/proteins were validated by immunohistochemistry. Gene-drug, gene-ncRNA interaction analysis were performed to identify drugs or non-coding RNAs (ncRNAs) that potentially interacted with the model-genes. Homology Modeling and Molecular Docking were conducted to filter the optimal candidate as the subsequent adjuvant or alternative for IFX in predicted non-responders. At last, the down-regulation of the key model-gene/protein CYP24A1 by the drug candidate Deferasirox was verified by Western Blot and qRT-PCR Assay based on cellular experiments. RESULTS A total of 113 DEGs were identified in the Training Cohort, mainly enriched in inflammatory cell chemotaxis, migration, and response to molecules derived from intestinal microbiota. Activated pro-inflammatory innate immune cells, including neutrophils, M1 macrophages, activated dendritic cells and mast cells, were significantly enriched in colons of non-responders. The prediction model based on three model-genes (IFI44L, CYP24A1, and RGS1) exhibited strong predictive efficacy, with AUC values of 0.901 and 0.80 in the Training and Validation Cohorts, respectively. Higher expression of the three model-genes/proteins in colons of non-responders to IFX was confirmed by clinical colonic mucosal biopsies. 4 Drugs (Calcitriol, Lunacalcipol, Deferasirox, Telaprevir), 15 miRNAs and 66 corresponding lnRNAs interacting with model-genes were identified. The protein 3D structure of the key model-gene/protein (human-derived CYP24A1) was developed. Through the Molecular Docking and cellular experimental validation, Deferasirox, which significantly down-regulated both the RNA and protein expression of CYP24A1, was identified as the optimal adjuvant or alternative for IFX in predicted non-responders with UC. CONCLUSION This study developed a novel prediction model for pre-assessing the efficacy of IFX in patients with UC, as the first step towards personalized therapy. Meanwhile, drugs and non-coding RNAs were provided as potential candidates to develop the next-step precise treatment for the predicted non-responders. In particular, Defeasirox appears to hold promise as an adjuvant or alternative to IFX for the optimization of UC therapy.
Collapse
Affiliation(s)
- Dan Pu
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Pengfei Wang
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Xiang Wang
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Yonggang Tian
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Hang Gong
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Xueni Ma
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Muyang Li
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Dekui Zhang
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China.
| |
Collapse
|
2
|
Jiang Y, Zhang X, Wang B, Tang L, Liu X, Ding X, Dong Y, Lei H, Wang D, Feng H. Single-cell transcriptomic analysis reveals a decrease in the frequency of macrophage-RGS1 high subsets in patients with osteoarticular tuberculosis. Mol Med 2024; 30:118. [PMID: 39123125 PMCID: PMC11316427 DOI: 10.1186/s10020-024-00886-9] [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: 04/29/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Cell subsets differentially modulate host immune responses to Mycobacterium tuberculosis (MTB) infection. However, the nature and functions of these subsets against osteoarticular tuberculosis (OTB) are unclear. Here, we aimed to understand the phenotypes and functions of immune cell subsets in patients with OTB using single-cell RNA sequencing (scRNA-Seq). METHODS Pathological and healthy adjacent tissues were isolated from patients with OTB and subjected to scRNA-Seq. Unsupervised clustering of cells was performed based on gene expression profiles, and uniform manifold approximation and projection was used for clustering visualization. RESULTS Thirteen cell subsets were identified in OTB tissues. scRNA-seq datasets of patients and healthy controls (HCs) showed that infection changed the frequency of immune cell subsets in OTB tissues. Myeloid cell examination revealed nine subsets. The frequency of macrophage-RGS1high subsets decreased in OTB tissues; this increased MTB susceptibility in an SLC7A11/ferroptosis-dependent manner. Immunohistochemistry assays and flow cytometry for patients with OTB and osteoarticular bacterial infection (OBI) and HCs verified that the frequency of macrophage-RGS1high subset decreased in OTB tissues and blood samples, thereby distinguishing patients with OTB from HCs and patients with OBI. CONCLUSION The macrophage-RGS1high subset levels were decreased in patients with OTB, and would be up-regulated after effective treatment. Therefore, the clinical significance of this study is to discover that macrophage-RGS1high subset may serve as a potential biomarker for OTB diagnosis and treatment efficacy monitoring.
Collapse
Affiliation(s)
- Ying Jiang
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, People's Republic of China
| | - Xinqiang Zhang
- Emergency Department, Chengde Central Hospital, Chengde, 067000, Hebei, People's Republic of China
| | - Bo Wang
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, People's Republic of China
| | - Liping Tang
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, People's Republic of China
| | - Xin Liu
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, People's Republic of China
| | - Xiudong Ding
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, People's Republic of China
| | - Yueming Dong
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, People's Republic of China
| | - Hong Lei
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, People's Republic of China.
| | - Di Wang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China.
| | - Huicheng Feng
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, People's Republic of China.
| |
Collapse
|
3
|
Wang K, Xiao Y, Zhang W, Yang H, Li C, Wang J, Li G. Elucidating key immunological biomarkers and immune microenvironment dynamics in aging-related intracranial aneurysm through integrated multi-omics analysis. ENVIRONMENTAL TOXICOLOGY 2024; 39:2642-2654. [PMID: 38214030 DOI: 10.1002/tox.24117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/17/2023] [Accepted: 12/25/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND The exact cause of intracranial aneurysms (IA) is still unclear. However, pro-inflammatory factors are known to contribute to IA progression. The specific changes in the immune microenvironment of IAs remain largely unexplored. METHODS This study analyzed single-cell sequencing data from a male mouse model of brain aneurysm, focusing on samples before and after elastase-induced Willis aneurysms. The data helped identify eight distinct cell subpopulations: fibroblasts, macrophages, NK cells, endothelial cells, B cells, granulocytes, and monocytes. The study also involved bulk RNA sequencing of 97 IA samples, utilizing ssGSEA and CIBERSORT algorithms for analysis. Intercellular communication among these cells was inferred to understand the immune dynamics in IA. RESULTS The study found that fibroblasts and macrophages are predominant in various disease states of IA. Notably, the onset of IA was marked by a significant increase in fibroblasts and a decrease in macrophages. There was a marked increase in cellular interactions, especially involving macrophages, at the onset of the disease. Through enrichment analysis, 12 potential immunogenic biomarkers were identified. Of these, Rgs1 emerged as a critical molecule in IA formation, confirmed through secondary validation in a single-cell sequencing dataset. CONCLUSION This comprehensive analysis of immune cell composition and intercellular communication in IA tissues highlights the significant roles of macrophages and the molecule Rgs1. These findings shed light on the physiological and pathological conditions of IA, offering new insights into its immune microenvironment.
Collapse
Affiliation(s)
- Kai Wang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yangyang Xiao
- Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, China
| | - Wenjia Zhang
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haiguang Yang
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chaoqun Li
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jia Wang
- Department of Obstetrics and Gynecology, Shanghai Putuo District Liqun Hospital, Shanghai, China
| | - Guoshu Li
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Onisiforou A, Christodoulou CC, Zamba-Papanicolaou E, Zanos P, Georgiou P. Transcriptomic analysis reveals sex-specific patterns in the hippocampus in Alzheimer's disease. Front Endocrinol (Lausanne) 2024; 15:1345498. [PMID: 38689734 PMCID: PMC11058985 DOI: 10.3389/fendo.2024.1345498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
Background The hippocampus, vital for memory and learning, is among the first brain regions affected in Alzheimer's Disease (AD) and exhibits adult neurogenesis. Women face twice the risk of developing AD compare to men, making it crucial to understand sex differences in hippocampal function for comprehending AD susceptibility. Methods We conducted a comprehensive analysis of bulk mRNA postmortem samples from the whole hippocampus (GSE48350, GSE5281) and its CA1 and CA3 subfields (GSE29378). Our aim was to perform a comparative molecular signatures analysis, investigating sex-specific differences and similarities in the hippocampus and its subfields in AD. This involved comparing the gene expression profiles among: (a) male controls (M-controls) vs. female controls (F-controls), (b) females with AD (F-AD) vs. F-controls, (c) males with AD (M-AD) vs. M-controls, and (d) M-AD vs. F-AD. Furthermore, we identified AD susceptibility genes interacting with key targets of menopause hormone replacement drugs, specifically the ESR1 and ESR2 genes, along with GPER1. Results The hippocampal analysis revealed contrasting patterns between M-AD vs. M-controls and F-AD vs. F-controls, as well as M-controls vs. F-controls. Notably, BACE1, a key enzyme linked to amyloid-beta production in AD pathology, was found to be upregulated in M-controls compared to F-controls in both CA1 and CA3 hippocampal subfields. In M-AD vs. M-controls, the GABAergic synapse was downregulated, and the Estrogen signaling pathway was upregulated in both subfields, unlike in F-AD vs. F-controls. Analysis of the whole hippocampus also revealed upregulation of the GABAergic synapse in F-AD vs. F-controls. While direct comparison of M-AD vs. F-AD, revealed a small upregulation of the ESR1 gene in the CA1 subfield of males. Conversely, F-AD vs. F-controls exhibited downregulation of the Dopaminergic synapse in both subfields, while the Calcium signaling pathway showed mixed regulation, being upregulated in CA1 but downregulated in CA3, unlike in M-AD vs. M-controls. The upregulated Estrogen signaling pathway in M-AD, suggests a compensatory response to neurodegenerative specifically in males with AD. Our results also identified potential susceptibility genes interacting with ESR1 and ESR2, including MAPK1, IGF1, AKT1, TP53 and CD44. Conclusion These findings underscore the importance of sex-specific disease mechanisms in AD pathogenesis. Region-specific analysis offers a more detailed examination of localized changes in the hippocampus, enabling to capture sex-specific molecular patterns in AD susceptibility and progression.
Collapse
Affiliation(s)
- Anna Onisiforou
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, Nicosia, Cyprus
| | | | | | - Panos Zanos
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, Nicosia, Cyprus
| | - Polymnia Georgiou
- Laboratory of Epigenetics and Gene Regulation, Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
- Psychoneuroendocrinology Laboratory, Department of Psychology, University of Wisconsin Milwaukee, Milwaukee, WI, United States
| |
Collapse
|
5
|
Wu M, Xu X, Yang C, An Q, Zhang J, Zhao Z, Feng Y, Liang W, Fu Y, Zhang G, Jiang T. Regulator of G protein signaling 1 is a potential target in gastric cancer and impacts tumor-associated macrophages. Cancer Sci 2024; 115:1085-1101. [PMID: 38287908 PMCID: PMC11006993 DOI: 10.1111/cas.16083] [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: 08/30/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/31/2024] Open
Abstract
Regulator of G protein signaling 1 (RGS1) is closely associated with the tumor immune microenvironment and is highly expressed in various tumors and immune cells. The specific effects of RGS1 in the dynamic progression from chronic gastritis to gastric cancer have not been reported, and the role of tumor-associated macrophages (TAMs) is also unclear. In the present study, RGS1 was identified as an upregulated gene in different pathological stages ranging from chronic gastritis to gastric cancer by using Gene Expression Omnibus (GEO) screening together with pancancer analysis of The Cancer Genome Atlas and clinical prognostic analysis. The results indicated that RGS1 is highly expressed in gastric cancer and has potential prognostic value. We confirmed through in vivo experiments that RGS1 inhibited the proliferation of gastric cancer cells and promoted apoptosis, which was further corroborated by in vitro experiments. Additionally, RGS1 influenced cell migration and invasion. In our subsequent investigation of RGS1, we discovered its role in the immune response. Through analyses of single-cell and GEO database data, we confirmed its involvement in immune cell regulation, specifically TAM activation. Subsequently, we conducted in vivo and in vitro experiments to confirm the involvement of RGS1 in polarizing M1 macrophages while indirectly regulating M2 macrophages through tumor cells. In conclusion, RGS1 could be a potential target for the transformation of chronic gastritis into gastric cancer and has a measurable impact on TAMs, which warrants further in-depth research.
Collapse
Affiliation(s)
- Mengting Wu
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Labortory of Blood‐Stasis‐Toxin Syndrome of Zhejiang ProvinceHangzhouChina
- Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of ZhejiangHangzhouChina
| | - Xuefei Xu
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Labortory of Blood‐Stasis‐Toxin Syndrome of Zhejiang ProvinceHangzhouChina
- Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of ZhejiangHangzhouChina
| | - Chuqi Yang
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Labortory of Blood‐Stasis‐Toxin Syndrome of Zhejiang ProvinceHangzhouChina
- Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of ZhejiangHangzhouChina
| | - Qingwen An
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Labortory of Blood‐Stasis‐Toxin Syndrome of Zhejiang ProvinceHangzhouChina
- Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of ZhejiangHangzhouChina
| | - Jingcheng Zhang
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Labortory of Blood‐Stasis‐Toxin Syndrome of Zhejiang ProvinceHangzhouChina
- Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of ZhejiangHangzhouChina
| | - Zhengqi Zhao
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Labortory of Blood‐Stasis‐Toxin Syndrome of Zhejiang ProvinceHangzhouChina
- Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of ZhejiangHangzhouChina
| | - Yewen Feng
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Labortory of Blood‐Stasis‐Toxin Syndrome of Zhejiang ProvinceHangzhouChina
- Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of ZhejiangHangzhouChina
| | - Weiyu Liang
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Labortory of Blood‐Stasis‐Toxin Syndrome of Zhejiang ProvinceHangzhouChina
- Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of ZhejiangHangzhouChina
| | - Yufei Fu
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Labortory of Blood‐Stasis‐Toxin Syndrome of Zhejiang ProvinceHangzhouChina
- Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of ZhejiangHangzhouChina
| | - Guangji Zhang
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Labortory of Blood‐Stasis‐Toxin Syndrome of Zhejiang ProvinceHangzhouChina
- Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of ZhejiangHangzhouChina
| | - Tao Jiang
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Labortory of Blood‐Stasis‐Toxin Syndrome of Zhejiang ProvinceHangzhouChina
- Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of ZhejiangHangzhouChina
| |
Collapse
|
6
|
Wang Y, Liu Z, Song S, Wang J, Jin C, Jia L, Ma Y, Yuan T, Cai Z, Xiang M. IRF5 governs macrophage adventitial infiltration to fuel abdominal aortic aneurysm formation. JCI Insight 2024; 9:e171488. [PMID: 38175709 PMCID: PMC11143966 DOI: 10.1172/jci.insight.171488] [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: 04/14/2023] [Accepted: 12/26/2023] [Indexed: 01/05/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a chronic inflammatory disease characterized by the expansion of the aortic wall. One of the most significant features is the infiltration of macrophages in the adventitia, which drives vasculature remodeling. The role of macrophage-derived interferon regulatory factor 5 (IRF5) in macrophage infiltration and AAA formation remains unknown. RNA sequencing of AAA adventitia identified Irf5 as the top significantly increased transcription factor that is predominantly expressed in macrophages. Global and myeloid cell-specific deficiency of Irf5 reduced AAA progression, with a marked reduction in macrophage infiltration. Further cellular investigations indicated that IRF5 promotes macrophage migration by direct regulation of downstream phosphoinositide 3-kinase γ (PI3Kγ, Pik3cg). Pik3cg ablation hindered AAA progression, and myeloid cell-specific salvage of Pik3cg restored AAA progression and macrophage infiltration derived from Irf5 deficiency. Finally, we found that IRF5 and PI3Kγ expression in the adventitia is significantly increased in patients with AAA. These findings reveal that the IRF5-dependent regulation of PI3Kγ is essential for AAA formation.
Collapse
Affiliation(s)
- Yidong Wang
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| | - Zhenjie Liu
- Department of Vascular Surgery, The second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shen Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianfang Wang
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| | - Chunna Jin
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| | - Liangliang Jia
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| | - Yuankun Ma
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| | - Tan Yuan
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| | - Zhejun Cai
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| | - Meixiang Xiang
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| |
Collapse
|
7
|
Eley L, Richardson RV, Alqahtani A, Chaudhry B, Henderson DJ. eNOS plays essential roles in the developing heart and aorta linked to disruption of Notch signalling. Dis Model Mech 2024; 17:dmm050265. [PMID: 38111957 PMCID: PMC10846539 DOI: 10.1242/dmm.050265] [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: 04/24/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023] Open
Abstract
eNOS (NOS3) is the enzyme that generates nitric oxide, a signalling molecule and regulator of vascular tone. Loss of eNOS function is associated with increased susceptibility to atherosclerosis, hypertension, thrombosis and stroke. Aortopathy and cardiac hypertrophy have also been found in eNOS null mice, but their aetiology is unclear. We evaluated eNOS nulls before and around birth for cardiac defects, revealing severe abnormalities in the ventricular myocardium and pharyngeal arch arteries. Moreover, in the aortic arch, there were fewer baroreceptors, which sense changes in blood pressure. Adult eNOS null survivors showed evidence of cardiac hypertrophy, aortopathy and cartilaginous metaplasia in the periductal region of the aortic arch. Notch1 and neuregulin were dysregulated in the forming pharyngeal arch arteries and ventricles, suggesting that these pathways may be relevant to the defects observed. Dysregulation of eNOS leads to embryonic and perinatal death, suggesting mutations in eNOS are candidates for causing congenital heart defects in humans. Surviving eNOS mutants have a deficiency of baroreceptors that likely contributes to high blood pressure and may have relevance to human patients who suffer from hypertension associated with aortic arch abnormalities.
Collapse
Affiliation(s)
- Lorraine Eley
- Bioscience Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Rachel V. Richardson
- Bioscience Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Ahlam Alqahtani
- Bioscience Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Bill Chaudhry
- Bioscience Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Deborah J. Henderson
- Bioscience Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| |
Collapse
|
8
|
Song J, Li M, Chen C, Zhou J, Wang L, Yan Y, She J, Tong L, Song Y. Regulator of G protein signaling protein 6 alleviates acute lung injury by inhibiting inflammation and promoting cell self-renewal in mice. Cell Mol Biol Lett 2023; 28:102. [PMID: 38066447 PMCID: PMC10709870 DOI: 10.1186/s11658-023-00488-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 09/08/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a disease with high mortality and morbidity. Regulator of G protein signaling protein 6 (RGS6), identified as a tumor suppressor gene, has received increasing attention owing to its close relationship with oxidative stress and inflammation. However, the association between ARDS and RGS6 has not been reported. METHODS Congruously regulated G protein-coupled receptor (GPCR)-related genes and differentially expressed genes (DEGs) in an acute lung injury (ALI) model were identified, and functional enrichment analysis was conducted. In an in vivo study, the effects of RGS6 knockout were studied in a mouse model of ALI induced by lipopolysaccharide (LPS). HE staining, ELISA, and immunohistochemistry were used to evaluate pathological changes and the degree of inflammation. In vitro, qRT‒PCR, immunofluorescence staining, and western blotting were used to determine the dynamic changes in RGS6 expression in cells. The RGS6 overexpression plasmid was constructed for transfection. qRT‒PCR was used to assess proinflammatory factors transcription. Western blotting and flow cytometry were used to evaluate apoptosis and reactive oxygen species (ROS) production. Organoid culture was used to assess the stemness and self-renewal capacity of alveolar epithelial type II cells (AEC2s). RESULTS A total of 110 congruously regulated genes (61 congruously upregulated and 49 congruously downregulated genes) were identified among GPCR-related genes and DEGs in the ALI model. RGS6 was downregulated in vivo and in vitro in the ALI model. RGS6 was expressed in the cytoplasm and accumulated in the nucleus after LPS stimulation. Compared with the control group, we found higher mortality, more pronounced body weight changes, more serious pulmonary edema and pathological damage, and more neutrophil infiltration in the RGS6 knockout group upon LPS stimulation in vivo. Moreover, AEC2s loss was significantly increased upon RGS6 knockout. Organoid culture assays showed slower alveolar organoid formation, fewer alveolar organoids, and impaired development of new structures after passaging upon RGS6 knockout. In addition, RGS6 overexpression decreased ROS production as well as proinflammatory factor transcription in macrophages and decreased apoptosis in epithelial cells. CONCLUSIONS RGS6 plays a protective role in ALI not only in early inflammatory responses but also in endogenous lung stem cell regeneration.
Collapse
Affiliation(s)
- Juan Song
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Pulmonary Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361000, China
- Shanghai Respiratory Research Institute, Shanghai, 200032, China
| | - Miao Li
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Respiratory Research Institute, Shanghai, 200032, China
| | - Cuicui Chen
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Respiratory Research Institute, Shanghai, 200032, China
| | - Jian Zhou
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Respiratory Research Institute, Shanghai, 200032, China
| | - Linlin Wang
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Respiratory Research Institute, Shanghai, 200032, China
| | - Yu Yan
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Respiratory Research Institute, Shanghai, 200032, China
| | - Jun She
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Respiratory Research Institute, Shanghai, 200032, China.
| | - Lin Tong
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Department of Pulmonary Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361000, China.
- Shanghai Respiratory Research Institute, Shanghai, 200032, China.
| | - Yuanlin Song
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Department of Pulmonary Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361000, China.
- Shanghai Respiratory Research Institute, Shanghai, 200032, China.
| |
Collapse
|
9
|
Xu Q, Yao M, Tang C. RGS2 and female common diseases: a guard of women's health. J Transl Med 2023; 21:583. [PMID: 37649067 PMCID: PMC10469436 DOI: 10.1186/s12967-023-04462-3] [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: 07/05/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023] Open
Abstract
Currently, women around the world are still suffering from various female common diseases with the high incidence, such as ovarian cancer, uterine fibroids and preeclampsia (PE), and some diseases are even with the high mortality rate. As a negative feedback regulator in G Protein-Coupled Receptor signaling (GPCR), the Regulator of G-protein Signaling (RGS) protein family participates in regulating kinds of cell biological functions by destabilizing the enzyme-substrate complex through the transformation of hydrolysis of G Guanosine Triphosphate (GTP). Recent work has indicated that, the Regulator of G-protein Signaling 2 (RGS2), a member belonging to the RGS protein family, is closely associated with the occurrence and development of certain female diseases, providing with the evidence that RGS2 functions in sustaining women's health. In this review paper, we summarize the current knowledge of RGS2 in female common diseases, and also tap and discuss its therapeutic potential by targeting multiple mechanisms.
Collapse
Affiliation(s)
- Qiang Xu
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd, Hangzhou, 310052, People's Republic of China
| | - Mukun Yao
- Department of Gynecology, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| | - Chao Tang
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd, Hangzhou, 310052, People's Republic of China.
| |
Collapse
|
10
|
Jin B, Fei G, Sang S, Zhong C. Identification of biomarkers differentiating Alzheimer's disease from other neurodegenerative diseases by integrated bioinformatic analysis and machine-learning strategies. Front Mol Neurosci 2023; 16:1152279. [PMID: 37234685 PMCID: PMC10205980 DOI: 10.3389/fnmol.2023.1152279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Background Alzheimer's disease (AD) is the most common neurodegenerative disease, imposing huge mental and economic burdens on patients and society. The specific molecular pathway(s) and biomarker(s) that distinguish AD from other neurodegenerative diseases and reflect the disease progression are still not well studied. Methods Four frontal cortical datasets of AD were integrated to conduct differentially expressed genes (DEGs) and functional gene enrichment analyses. The transcriptional changes after the integrated frontal cortical datasets subtracting the cerebellar dataset of AD were further compared with frontal cortical datasets of frontotemporal dementia and Huntingdon's disease to identify AD-frontal-associated gene expression. Integrated bioinformatic analysis and machine-learning strategies were applied for screening and determining diagnostic biomarkers, which were further validated in another two frontal cortical datasets of AD by receiver operating characteristic (ROC) curves. Results Six hundred and twenty-six DEGs were identified as AD frontal associated, including 580 downregulated genes and 46 upregulated genes. The functional enrichment analysis revealed that immune response and oxidative stress were enriched in AD patients. Decorin (DCN) and regulator of G protein signaling 1 (RGS1) were screened as diagnostic biomarkers in distinguishing AD from frontotemporal dementia and Huntingdon's disease of AD. The diagnostic effects of DCN and RGS1 for AD were further validated in another two datasets of AD: the areas under the curve (AUCs) reached 0.8148 and 0.8262 in GSE33000, and 0.8595 and 0.8675 in GSE44770. There was a better value for AD diagnosis when combining performances of DCN and RGS1 with the AUCs of 0.863 and 0.869. Further, DCN mRNA level was correlated to CDR (Clinical Dementia Rating scale) score (r = 0.5066, p = 0.0058) and Braak staging (r = 0.3348, p = 0.0549). Conclusion DCN and RGS1 associated with the immune response may be useful biomarkers for diagnosing AD and distinguishing the disease from frontotemporal dementia and Huntingdon's disease. DCN mRNA level reflects the development of the disease.
Collapse
Affiliation(s)
- Boru Jin
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Guoqiang Fei
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Shaoming Sang
- Shanghai Raising Pharmaceutical Technology Co., Ltd., Shanghai, China
| | - Chunjiu Zhong
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Loick P, Mohammad GH, Cassimjee I, Chandrashekar A, Lapolla P, Carrington A, Vera-Aviles M, Handa A, Lee R, Lakhal-Littleton S. Protective Role for Smooth Muscle Cell Hepcidin in Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2023; 43:713-725. [PMID: 36951059 PMCID: PMC10125116 DOI: 10.1161/atvbaha.123.319224] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 03/10/2023] [Indexed: 03/24/2023]
Abstract
BACKGROUND Hepcidin is a liver-derived hormone that controls systemic iron homeostasis, by inhibiting the iron exporter ferroportin in the gut and spleen, respective sites of iron absorption and recycling. Hepcidin is also expressed ectopically in the context of cardiovascular disease. However, the precise role of ectopic hepcidin in underlying pathophysiology is unknown. In patients with abdominal aortic aneurysm (AAA), hepcidin is markedly induced in smooth muscle cells (SMCs) of the aneurysm wall and inversely correlated with the expression of LCN2 (lipocalin-2), a protein implicated in AAA pathology. In addition, plasma hepcidin levels were inversely correlated with aneurysm growth, suggesting hepcidin has a potential disease-modifying role. METHODS To probe the role of SMC-derived hepcidin in the setting of AAA, we applied AngII (Angiotensin-II)-induced AAA model to mice harbouring an inducible, SMC-specific deletion of hepcidin. To determine whether SMC-derived hepcidin acted cell-autonomously, we also used mice harboring an inducible SMC-specific knock-in of hepcidin-resistant ferroportinC326Y. The involvement of LCN2 was established using a LCN2-neutralizing antibody. RESULTS Mice with SMC-specific deletion of hepcidin or knock-in of hepcidin-resistant ferroportinC326Y had a heightened AAA phenotype compared with controls. In both models, SMCs exhibited raised ferroportin expression and reduced iron retention, accompanied by failure to suppress LCN2, impaired autophagy in SMCs, and greater aortic neutrophil infiltration. Pretreatment with LCN2-neutralizing antibody restored autophagy, reduced neutrophil infiltration, and prevented the heightened AAA phenotype. Finally, plasma hepcidin levels were consistently lower in mice with SMC-specific deletion of hepcidin than in controls, indicating that SMC-derived hepcidin contributes to the circulating pool in AAA. CONCLUSIONS Hepcidin elevation in SMCs plays a protective role in the setting of AAA. These findings are the first demonstration of a protective rather than deleterious role for hepcidin in cardiovascular disease. They highlight the need to further explore the prognostic and therapeutic value of hepcidin outside disorders of iron homeostasis.
Collapse
Affiliation(s)
- Paul Loick
- Department of Anesthesiology, Intensive Care and Pain Medicine, Universitätsklinikum Münster, Germany (P. Loick)
| | - Goran Hamid Mohammad
- Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (G.H.M., A. Carrington, M.V.-A., S.L.-L.)
| | - Ismail Cassimjee
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, United Kingdom (I.C., A. Chandrashekar, P. Lapolla, A.H., R.L.)
| | - Anirudh Chandrashekar
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, United Kingdom (I.C., A. Chandrashekar, P. Lapolla, A.H., R.L.)
| | - Pierfrancesco Lapolla
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, United Kingdom (I.C., A. Chandrashekar, P. Lapolla, A.H., R.L.)
| | - Alison Carrington
- Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (G.H.M., A. Carrington, M.V.-A., S.L.-L.)
| | - Mayra Vera-Aviles
- Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (G.H.M., A. Carrington, M.V.-A., S.L.-L.)
| | - Ashok Handa
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, United Kingdom (I.C., A. Chandrashekar, P. Lapolla, A.H., R.L.)
| | - Regent Lee
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, United Kingdom (I.C., A. Chandrashekar, P. Lapolla, A.H., R.L.)
| | - Samira Lakhal-Littleton
- Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (G.H.M., A. Carrington, M.V.-A., S.L.-L.)
| |
Collapse
|
12
|
Yuan G, Yang ST, Yang S. Regulator of G protein signaling 12 drives inflammatory arthritis by activating synovial fibroblasts through MYCBP2/KIF2A signaling. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:197-210. [PMID: 36700049 PMCID: PMC9843488 DOI: 10.1016/j.omtn.2022.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Synovial fibroblasts are the active and aggressive drivers in the progression of arthritis, but the cellular and molecular mechanisms remain unknown. Here, our results showed that regulator of G protein signaling 12 (RGS12) maintained ciliogenesis in synovial fibroblasts, which is critical for the development of inflammatory arthritis. Deletion of RGS12 led to a significant decrease in ciliogenesis, adhesion, migration, and secretion of synovial fibroblasts. Mechanistically, RGS12 overexpression in synovial fibroblasts increased the length and number of cilia but decreased the protein level of kinesin family member 2A (KIF2A). The results of LC-MS analyses showed that RGS12 interacted with MYC binding protein 2 to enhance its ubiquitination activity, through which the KIF2A protein was degraded in synovial fibroblasts. Moreover, overexpression of KIF2A blocked the increases in cilia length and number. Mice with RGS12 deficiency or treatment of RGS12 shRNA nanoparticles significantly decreased the clinical score, paw swelling, synovitis, and cartilage destruction during inflammatory arthritis by inhibiting the activation of synovial fibroblasts. Therefore, this study provides evidence that RGS12 activates synovial fibroblasts' pathological function via promoting MCYBP2-mediated degradation of KIF2A and ciliogenesis. Our data suggest that RGS12 may be a potential drug target for the treatment of inflammatory arthritis.
Collapse
Affiliation(s)
- Gongsheng Yuan
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shu-ting Yang
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shuying Yang
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Penn Center for Musculoskeletal Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Innovation & Precision Dentistry, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
13
|
Munshaw S, Redpath AN, Pike BT, Smart N. Thymosin β4 preserves vascular smooth muscle phenotype in atherosclerosis via regulation of low density lipoprotein related protein 1 (LRP1). Int Immunopharmacol 2023; 115:109702. [PMID: 37724952 PMCID: PMC10666903 DOI: 10.1016/j.intimp.2023.109702] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 12/29/2022] [Indexed: 01/21/2023]
Abstract
Atherosclerosis is a progressive, degenerative vascular disease and a leading cause of morbidity and mortality. In response to endothelial damage, platelet derived growth factor (PDGF)-BB induced phenotypic modulation of medial smooth muscle cells (VSMCs) promotes atherosclerotic lesion formation and destabilisation of the vessel wall. VSMC sensitivity to PDGF-BB is determined by endocytosis of Low density lipoprotein receptor related protein 1 (LRP1)-PDGFR β complexes to balance receptor recycling with lysosomal degradation. Consequently, LRP1 is implicated in various arterial diseases. Having identified Tβ4 as a regulator of LRP1-mediated endocytosis to protect against aortic aneurysm, we sought to determine whether Tβ4 may additionally function to protect against atherosclerosis, by regulating LRP1-mediated growth factor signalling. By single cell transcriptomic analysis, Tmsb4x, encoding Tβ4, strongly correlated with contractile gene expression and was significantly down-regulated in cells that adopted a modulated phenotype in atherosclerosis. We assessed susceptibility to atherosclerosis of global Tβ4 knockout mice using the ApoE-/- hypercholesterolaemia model. Inflammation, elastin integrity, VSMC phenotype and signalling were analysed in the aortic root and descending aorta. Tβ4KO; ApoE-/- mice develop larger atherosclerotic plaques than control mice, with medial layer degeneration characterised by accelerated VSMC phenotypic modulation. Defects in Tβ4KO; ApoE-/- mice phenocopied those in VSMC-specific LRP1 nulls and, moreover, were underpinned by hyperactivated LRP1-PDGFRβ signalling. We identify an atheroprotective role for endogenous Tβ4 in maintaining differentiated VSMC phenotype via LRP1-mediated PDGFRβ signalling.
Collapse
Affiliation(s)
- Sonali Munshaw
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3PT, UK
| | - Andia N Redpath
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3PT, UK; Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Oxford OX3 7TY, UK
| | - Benjamin T Pike
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3PT, UK
| | - Nicola Smart
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3PT, UK; Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Oxford OX3 7TY, UK.
| |
Collapse
|
14
|
von Werdt D, Gungor B, Barreto de Albuquerque J, Gruber T, Zysset D, Kwong Chung CKC, Corrêa-Ferreira A, Berchtold R, Page N, Schenk M, Kehrl JH, Merkler D, Imhof BA, Stein JV, Abe J, Turchinovich G, Finke D, Hayday AC, Corazza N, Mueller C. Regulator of G-protein signaling 1 critically supports CD8 + T RM cell-mediated intestinal immunity. Front Immunol 2023; 14:1085895. [PMID: 37153600 PMCID: PMC10158727 DOI: 10.3389/fimmu.2023.1085895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/13/2023] [Indexed: 05/09/2023] Open
Abstract
Members of the Regulator of G-protein signaling (Rgs) family regulate the extent and timing of G protein signaling by increasing the GTPase activity of Gα protein subunits. The Rgs family member Rgs1 is one of the most up-regulated genes in tissue-resident memory (TRM) T cells when compared to their circulating T cell counterparts. Functionally, Rgs1 preferentially deactivates Gαq, and Gαi protein subunits and can therefore also attenuate chemokine receptor-mediated immune cell trafficking. The impact of Rgs1 expression on tissue-resident T cell generation, their maintenance, and the immunosurveillance of barrier tissues, however, is only incompletely understood. Here we report that Rgs1 expression is readily induced in naïve OT-I T cells in vivo following intestinal infection with Listeria monocytogenes-OVA. In bone marrow chimeras, Rgs1 -/- and Rgs1 +/+ T cells were generally present in comparable frequencies in distinct T cell subsets of the intestinal mucosa, mesenteric lymph nodes, and spleen. After intestinal infection with Listeria monocytogenes-OVA, however, OT-I Rgs1 +/+ T cells outnumbered the co-transferred OT-I Rgs1- /- T cells in the small intestinal mucosa already early after infection. The underrepresentation of the OT-I Rgs1 -/- T cells persisted to become even more pronounced during the memory phase (d30 post-infection). Remarkably, upon intestinal reinfection, mice with intestinal OT-I Rgs1 +/+ TRM cells were able to prevent the systemic dissemination of the pathogen more efficiently than those with OT-I Rgs1 -/- TRM cells. While the underlying mechanisms are not fully elucidated yet, these data thus identify Rgs1 as a critical regulator for the generation and maintenance of tissue-resident CD8+ T cells as a prerequisite for efficient local immunosurveillance in barrier tissues in case of reinfections with potential pathogens.
Collapse
Affiliation(s)
- Diego von Werdt
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Bilgi Gungor
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | | | - Thomas Gruber
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Daniel Zysset
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Cheong K. C. Kwong Chung
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Department of Gastrointestinal Health, Immunology, Nestlé Research, Lausanne, Switzerland
| | - Antonia Corrêa-Ferreira
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Regina Berchtold
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Nicolas Page
- Department of Pathology, Division of Clinical Pathology, University & University Hospitals of Geneva, Geneva, Switzerland
| | - Mirjam Schenk
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - John H. Kehrl
- National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Doron Merkler
- Department of Pathology, Division of Clinical Pathology, University & University Hospitals of Geneva, Geneva, Switzerland
| | - Beat A. Imhof
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Department of Pathology and Immunology, Centre Medical Universitaire, University of Geneva, Geneva, Switzerland
| | - Jens V. Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Jun Abe
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Gleb Turchinovich
- Department of Biomedicine, and University Children’s Hospital Basel, University of Basel, Basel, Switzerland
| | - Daniela Finke
- Department of Biomedicine, and University Children’s Hospital Basel, University of Basel, Basel, Switzerland
| | - Adrian C. Hayday
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| | - Nadia Corazza
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- *Correspondence: Christoph Mueller, ; Nadia Corazza,
| | - Christoph Mueller
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Department of Biomedicine, and University Children’s Hospital Basel, University of Basel, Basel, Switzerland
- *Correspondence: Christoph Mueller, ; Nadia Corazza,
| |
Collapse
|
15
|
Xue W, He W, Yan M, Zhao H, Pi J. Exploring Shared Biomarkers of Myocardial Infarction and Alzheimer's Disease via Single-Cell/Nucleus Sequencing and Bioinformatics Analysis. J Alzheimers Dis 2023; 96:705-723. [PMID: 37840493 PMCID: PMC10657707 DOI: 10.3233/jad-230559] [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] [Accepted: 09/04/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND Patients are at increased risk of dementia, including Alzheimer's disease (AD), after myocardial infarction (MI), but the biological link between MI and AD is unclear. OBJECTIVE To understand the association between the pathogenesis of MI and AD and identify common biomarkers of both diseases. METHODS Using public databases, we identified common biomarkers of MI and AD. Least absolute shrinkage and selection operator (LASSO) regression and protein-protein interaction (PPI) network were performed to further screen hub biomarkers. Functional enrichment analyses were performed on the hub biomarkers. Single-cell/nucleus analysis was utilized to further analyze the hub biomarkers at the cellular level in carotid atherosclerosis and AD datasets. Motif enrichment analysis was used to screen key transcription factors. RESULTS 26 common differentially expressed genes were screened between MI and AD. Function enrichment analyses showed that these differentially expressed genes were mainly associated with inflammatory pathways. A key gene, Regulator of G-protein Signaling 1 (RGS1), was obtained by LASSO regression and PPI network. RGS1 was confirmed to mainly express in macrophages and microglia according to single-cell/nucleus analysis. The difference in expression of RGS1 in macrophages and microglia between disease groups and controls was statistically significant (p < 0.0001). The expression of RGS1 in the disease groups was upregulated with the differentiation of macrophages and microglia. RelA was a key transcription factor regulating RGS1. CONCLUSION Macrophages and microglia are involved in the inflammatory response of MI and AD. RGS1 may be a key biomarker in this process.
Collapse
Affiliation(s)
- Weiqi Xue
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Weifeng He
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Mengyuan Yan
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huanyi Zhao
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianbin Pi
- Department of Cardiovascular Disease, The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| |
Collapse
|
16
|
Weighted Gene Coexpression Network Analysis Identifies Crucial Genes Involved in Coronary Atherosclerotic Heart Disease. DISEASE MARKERS 2022; 2022:6971238. [PMID: 35958279 PMCID: PMC9363224 DOI: 10.1155/2022/6971238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/31/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022]
Abstract
Background Coronary atherosclerotic heart disease (CHD) is a lethal disease with an unstated pathogenic mechanism. Therefore, it is urgent to develop innovative strategies to ameliorate the outcome of CHD patients and explore novel biomarkers connected to the pathogenicity of CHD. Methods The weighted gene coexpression network analysis (WGCNA) was carried out on a coronary atherosclerosis dataset GSE90074 to determine the crucial modules and hub genes for their prospective relationship to CHD. After the different modules associated with CHD have been identified, the Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enriched pathway analyses were conducted. The protein-protein interaction (PPI) network was thereafter performed for the critical module using STRING and Cytoscape. Results The yellow module was recognized as the most critical module associated with CHD. The enriched pathways in the yellow module included those related to inflammatory response, positive regulation of extracellular signal-regulated kinase1/2 (ERK1/2) cascade, lipid catabolic process, cellular response to oxidative stress, apoptotic pathway, and NF-kappa B pathway. Further CytoHubba analysis revealed the top five hub genes (MMP14, CD28, CaMK4, RGS1, and DDAH1) associated with CHD development. Conclusions The current study provides the prognosis, novel hub genes, and signaling pathways for treating coronary atherosclerosis. However, their potential biological roles require deeper investigation.
Collapse
|
17
|
Lymperopoulos A, Suster MS, Borges JI. Cardiovascular GPCR regulation by regulator of G protein signaling proteins. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 193:145-166. [PMID: 36357075 DOI: 10.1016/bs.pmbts.2022.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
G protein-coupled receptors (GPCRs) play pivotal roles in regulation of cardiovascular homeostasis across all vertebrate species, including humans. In terms of normal cellular function, termination of GPCR signaling via the heterotrimeric G proteins is equally (if not more) important to its stimulation. The Regulator of G protein Signaling (RGS) protein superfamily are indispensable for GPCR signaling cessation at the cell membrane, and thus, for cellular control of GPCR signaling and function. Perturbations in both activation and termination of G protein signaling underlie many examples of cardiovascular dysfunction and heart disease pathogenesis. Despite the plethora of over 30 members comprising the mammalian RGS protein superfamily, each member interacts with a specific set of second messenger pathways and GPCR types/subtypes in a tissue/cell type-specific manner. An increasing number of studies over the past two decades have provided compelling evidence for the involvement of various RGS proteins in physiological regulation of cardiovascular GPCRs and, consequently, also in the pathophysiology of several cardiovascular ailments. This chapter summarizes the current understanding of the functional roles of RGS proteins as they pertain to cardiovascular, i.e., heart, blood vessel, and platelet GPCR function, with a particular focus on their implications for chronic heart failure pathophysiology and therapy.
Collapse
Affiliation(s)
- Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL, United States.
| | - Malka S Suster
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL, United States
| | - Jordana I Borges
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL, United States
| |
Collapse
|
18
|
Chan KYY, Chung PY, Zhang C, Poon ENY, Leung AWK, Leung KT. R4 RGS proteins as fine tuners of immature and mature hematopoietic cell trafficking. J Leukoc Biol 2022; 112:785-797. [PMID: 35694792 DOI: 10.1002/jlb.1mr0422-475r] [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: 11/30/2021] [Revised: 04/28/2022] [Indexed: 11/08/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) are the largest and most diverse group of membrane receptors. They are involved in almost every physiologic process and consequently have a pivotal role in an extensive number of pathologies, including genetic, neurologic, and immune system disorders. Indeed, the vast array of GPCRs mechanisms have led to the development of a tremendous number of drug therapies and already account for about a third of marketed drugs. These receptors mediate their downstream signals primarily via G proteins. The regulators of G-protein signaling (RGS) proteins are now in the spotlight as the critical modulatory factors of active GTP-bound Gα subunits of heterotrimeric G proteins to fine-tune the biologic responses driven by the GPCRs. Also, they possess noncanonical functions by multiple mechanisms, such as protein-protein interactions. Essential roles and impacts of these RGS proteins have been revealed in physiology, including hematopoiesis and immunity, and pathologies, including asthma, cancers, and neurologic disorders. This review focuses on the largest subfamily of R4 RGS proteins and provides a brief overview of their structures and G-proteins selectivity. With particular interest, we explore and highlight, their expression in the hematopoietic system and the regulation in the engraftment of hematopoietic stem/progenitor cells (HSPCs). Distinct expression patterns of R4 RGS proteins in the hematopoietic system and their pivotal roles in stem cell trafficking pave the way for realizing new strategies for enhancing the clinical performance of hematopoietic stem cell transplantation. Finally, we discuss the exciting future trends in drug development by targeting RGS activity and expression with small molecules inhibitors and miRNA approaches.
Collapse
Affiliation(s)
- Kathy Yuen Yee Chan
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Po Yee Chung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Chi Zhang
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Ellen Ngar Yun Poon
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Alex Wing Kwan Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong, China.,Department of Paediatrics & Adolescent Medicine, Hong Kong Children's Hospital, Hong Kong SAR, China
| | - Kam Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong, China.,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| |
Collapse
|
19
|
Agrafioti P, Morin-Baxter J, Tanagala KKK, Dubey S, Sims P, Lalla E, Momen-Heravi F. Decoding the role of macrophages in periodontitis and type 2 diabetes using single-cell RNA-sequencing. FASEB J 2022; 36:e22136. [PMID: 35032412 PMCID: PMC8881186 DOI: 10.1096/fj.202101198r] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/14/2021] [Accepted: 12/17/2021] [Indexed: 02/03/2023]
Abstract
Macrophages are resident myeloid cells in the gingival tissue which control homeostasis and play a pivotal role in orchestrating the immune response in periodontitis. Cell heterogeneity and functional phenotypes of macrophage subpopulations in periodontitis remain elusive. Here, we isolated gingival tissue from periodontitis-affected and healthy sites of patients with and without type 2 diabetes mellitus (T2DM). We then used single-cell RNA-sequencing (scRNA-seq) to define the heterogeneity of tissue-resident macrophages in gingival tissue in health vs. periodontitis. scRNA-seq demonstrated an unforeseen gene expression heterogeneity among macrophages in periodontitis and showed transcriptional and signaling heterogeneity of identified subsets in an independent cohort of patients with periodontitis and T2DM. Our bioinformatic inferences indicated divergent expression profiles in macrophages driven by transcriptional regulators CIITA, RELA, RFX5, and RUNX2. Macrophages in periodontitis expressed both pro-inflammatory and anti-inflammatory markers and their polarization was not mutually exclusive. The majority of macrophages in periodontitis expressed the monocyte lineage marker CD14, indicating their bone marrow lineage. We also found high expression and activation of RELA, a subunit of the NF-κB transcription factor complex, in gingival macrophages of periodontitis patients with T2DM. Our data suggested that heterogeneity and hyperinflammatory activation of macrophages may be relevant to the pathogenesis and outcomes of periodontitis, and may be further augmented in patients with T2DM.
Collapse
Affiliation(s)
- Panagiota Agrafioti
- Division of Periodontics, Section of Oral, Diagnostic and Rehabilitation Sciences, College of Dental Medicine, Columbia University, New York, New York, USA,Cancer Biology and Immunology Laboratory, Columbia University Irving Medical Center, New York, New York, USA
| | - Joshua Morin-Baxter
- Cancer Biology and Immunology Laboratory, Columbia University Irving Medical Center, New York, New York, USA,Fu Foundation School of Engineering and Applied Science, Columbia University, New York, New York, USA
| | - Kranthi K. K. Tanagala
- Division of Periodontics, Section of Oral, Diagnostic and Rehabilitation Sciences, College of Dental Medicine, Columbia University, New York, New York, USA,Cancer Biology and Immunology Laboratory, Columbia University Irving Medical Center, New York, New York, USA
| | - Sunil Dubey
- Division of Periodontics, Section of Oral, Diagnostic and Rehabilitation Sciences, College of Dental Medicine, Columbia University, New York, New York, USA,Cancer Biology and Immunology Laboratory, Columbia University Irving Medical Center, New York, New York, USA
| | - Peter Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York, USA,Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, New York, USA
| | - Evanthia Lalla
- Division of Periodontics, Section of Oral, Diagnostic and Rehabilitation Sciences, College of Dental Medicine, Columbia University, New York, New York, USA
| | - Fatemeh Momen-Heravi
- Division of Periodontics, Section of Oral, Diagnostic and Rehabilitation Sciences, College of Dental Medicine, Columbia University, New York, New York, USA,Cancer Biology and Immunology Laboratory, Columbia University Irving Medical Center, New York, New York, USA,Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
20
|
Feng D, Yu J, Bao L, Fan D, Zhang B. Inhibiting RGS1 attenuates secondary inflammation response and tissue degradation via the TLR/TRIF/NF-κB pathway in macrophage post spinal cord injury. Neurosci Lett 2022; 768:136374. [PMID: 34852285 DOI: 10.1016/j.neulet.2021.136374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 11/25/2021] [Indexed: 12/13/2022]
Abstract
Macrophage-dominated inflammation by the activation of Toll-like receptor (TLR) pathway leads to neurological disruption after spinal cord injury (SCI). Regulator of G-protein signaling 1 (RGS1) is reported to be a regulator in inflammation. The present study thus purposes to identify the unknown role of RGS1 mediating TLR on inflammation post SCI. A mouse model of traumatic SCI was established by a mechanical trauma at T10. The mice underwent SCI and a macrophage line activated by lipopolysaccharide (LPS) were treated with shRNA-RGS1 to elucidate the role of RGS1 in inflammatory progression. The inflammatory factors were measured, and the degree of histology and function protection were determined. The expression levels of RGS1, myeloid differentiation primary response protein 88 (Myd88), (TIR-domain-containing adaptor inducing interferon-β (TRIF), p38, metalloproteinase (MMP)-2, and MMP-9 were determined. RGS1 was robustly increased both in LPS-activated macrophage and SCI mice. The TLR signaling pathway-induced inflammation was suppressed by RGS1 knockdown. shRNA-mediated silence of RGS1 was exhibited a prominent decrease in TNF-α, IL-1β and IL-6 via TLR/TRIF/ nuclear factor kappa-B (NF-κB) axis. Depletion of RGS1 also inhibited MMP-induced tissue degradation via MAPK-p38 pathway in SCI mice. Moreover, suppression of RGS1 improved spinal cord histology and function recovery. These findings suggest that RGS1 regulates inflammation and tissue disruption via TLR/TRIF/NF-κB signaling pathway in mice with SCI, thereby explaining a novel target that regulates macrophage inflammation post SCI.
Collapse
Affiliation(s)
- Dongqian Feng
- Department of Orthopedics, Shuyang Hospital Affiliated to Medical College of Yangzhou University, Shuyang Hospital of Traditional Chinese Medicine, Shuyang 223600, China; Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Jiasheng Yu
- Department of Orthopedics, Shuyang Hospital Affiliated to Medical College of Yangzhou University, Shuyang Hospital of Traditional Chinese Medicine, Shuyang 223600, China
| | - Lei Bao
- Department of Orthopedics, Shuyang Hospital Affiliated to Medical College of Yangzhou University, Shuyang Hospital of Traditional Chinese Medicine, Shuyang 223600, China
| | - Daobo Fan
- Department of Orthopedics, Shuyang Hospital Affiliated to Medical College of Yangzhou University, Shuyang Hospital of Traditional Chinese Medicine, Shuyang 223600, China
| | - Bin Zhang
- Department of Orthopedics, Shuyang Hospital Affiliated to Medical College of Yangzhou University, Shuyang Hospital of Traditional Chinese Medicine, Shuyang 223600, China.
| |
Collapse
|
21
|
Affiliation(s)
| | - Leo M Carlin
- Cancer Research UK Beatson Institute, Glasgow, UK.
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
22
|
Huang D, Chen X, Zeng X, Lao L, Li J, Xing Y, Lu Y, Ouyang Q, Chen J, Yang L, Su F, Yao H, Liu Q, Su S, Song E. Targeting regulator of G protein signaling 1 in tumor-specific T cells enhances their trafficking to breast cancer. Nat Immunol 2021; 22:865-879. [PMID: 34140678 DOI: 10.1038/s41590-021-00939-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 04/22/2021] [Indexed: 02/05/2023]
Abstract
Reduced infiltration of anti-tumor lymphocytes remains a major cause of tumor immune evasion and is correlated with poor cancer survival. Here, we found that upregulation of regulator of G protein signaling (RGS)1 in helper TH1 cells and cytotoxic T lymphocytes (CTLs) reduced their trafficking to and survival in tumors and was associated with shorter survival of patients with breast and lung cancer. RGS1 was upregulated by type II interferon (IFN)-signal transducer and activator of transcription (STAT)1 signaling and impaired trafficking of circulating T cells to tumors by inhibiting calcium influx and suppressing activation of the kinases ERK and AKT. RGS1 knockdown in adoptively transferred tumor-specific CTLs significantly increased their infiltration and survival in breast and lung tumor grafts and effectively inhibited tumor growth in vivo, which was further improved when combined with programmed death ligand (PD-L)1 checkpoint inhibition. Our findings reveal RGS1 is important for tumor immune evasion and suggest that targeting RGS1 may provide a new strategy for tumor immunotherapy.
Collapse
MESH Headings
- Animals
- Apoptosis
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Breast Neoplasms/therapy
- Carcinoma, Ductal, Breast/immunology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/therapy
- Cell Line, Tumor
- Chemokines/metabolism
- Chemotaxis, Leukocyte
- Coculture Techniques
- Cytotoxicity, Immunologic
- Female
- Humans
- Immunotherapy, Adoptive
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/transplantation
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Microscopy, Fluorescence
- Microscopy, Video
- RGS Proteins/genetics
- RGS Proteins/metabolism
- Signal Transduction
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Time Factors
- Time-Lapse Imaging
- Tumor Cells, Cultured
- Tumor Escape
- Mice
Collapse
Affiliation(s)
- Di Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Xueman Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Xin Zeng
- Bioland Laboratory, Guangzhou, China
- Program of Molecular Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Liyan Lao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Jiaqian Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Yue Xing
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Yiwen Lu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Qian Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Jianing Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Linbin Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Fengxi Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qiang Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Shicheng Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Bioland Laboratory, Guangzhou, China.
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Bioland Laboratory, Guangzhou, China.
- Program of Molecular Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
- Fountain-Valley Institute for Life Sciences, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
| |
Collapse
|
23
|
Munshaw S, Bruche S, Redpath AN, Jones A, Patel J, Dubé KN, Lee R, Hester SS, Davies R, Neal G, Handa A, Sattler M, Fischer R, Channon KM, Smart N. Thymosin β4 protects against aortic aneurysm via endocytic regulation of growth factor signaling. J Clin Invest 2021; 131:127884. [PMID: 33784254 PMCID: PMC8121525 DOI: 10.1172/jci127884] [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: 02/01/2019] [Accepted: 03/23/2021] [Indexed: 01/06/2023] Open
Abstract
Vascular stability and tone are maintained by contractile smooth muscle cells (VSMCs). However, injury-induced growth factors stimulate a contractile-synthetic phenotypic modulation which increases susceptibility to abdominal aortic aneurysm (AAA). As a regulator of embryonic VSMC differentiation, we hypothesized that Thymosin β4 (Tβ4) may function to maintain healthy vasculature throughout postnatal life. This was supported by the identification of an interaction with low density lipoprotein receptor related protein 1 (LRP1), an endocytic regulator of platelet-derived growth factor BB (PDGF-BB) signaling and VSMC proliferation. LRP1 variants have been implicated by genome-wide association studies with risk of AAA and other arterial diseases. Tβ4-null mice displayed aortic VSMC and elastin defects that phenocopy those of LRP1 mutants, and their compromised vascular integrity predisposed them to Angiotensin II-induced aneurysm formation. Aneurysmal vessels were characterized by enhanced VSMC phenotypic modulation and augmented PDGFR-β signaling. In vitro, enhanced sensitivity to PDGF-BB upon loss of Tβ4 was associated with dysregulated endocytosis, with increased recycling and reduced lysosomal targeting of LRP1-PDGFR-β. Accordingly, the exacerbated aneurysmal phenotype in Tβ4-null mice was rescued upon treatment with the PDGFR-β antagonist Imatinib. Our study identifies Tβ4 as a key regulator of LRP1 for maintaining vascular health, and provides insights into the mechanisms of growth factor-controlled VSMC phenotypic modulation underlying aortic disease progression.
Collapse
MESH Headings
- Angiotensin II/adverse effects
- Angiotensin II/pharmacology
- Animals
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/prevention & control
- Becaplermin/genetics
- Becaplermin/metabolism
- Low Density Lipoprotein Receptor-Related Protein-1/genetics
- Low Density Lipoprotein Receptor-Related Protein-1/metabolism
- Male
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/metabolism
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Thymosin/genetics
- Thymosin/metabolism
- Thymosin/pharmacology
Collapse
Affiliation(s)
- Sonali Munshaw
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| | - Susann Bruche
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| | - Andia N. Redpath
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| | - Alisha Jones
- Institute of Structural Biology, Helmholtz Zentrum München, Neuherberg, Munich, Germany
- Biomolecular NMR and Center for Integrated Protein Science Munich at Chemistry Department, Technical University of Munich, Garching, Munich, Germany
| | - Jyoti Patel
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | | | - Regent Lee
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Svenja S. Hester
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom
| | - Rachel Davies
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| | - Giles Neal
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| | - Ashok Handa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Michael Sattler
- Institute of Structural Biology, Helmholtz Zentrum München, Neuherberg, Munich, Germany
- Biomolecular NMR and Center for Integrated Protein Science Munich at Chemistry Department, Technical University of Munich, Garching, Munich, Germany
| | - Roman Fischer
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom
| | - Keith M. Channon
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Nicola Smart
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| |
Collapse
|
24
|
Li J, Peng L, Bai W, Peng P, Chen W, Yang W, Shao J. Biliverdin Protects Against Cerebral Ischemia/Reperfusion Injury by Regulating the miR-27a-3p/Rgs1 Axis. Neuropsychiatr Dis Treat 2021; 17:1165-1181. [PMID: 33911865 PMCID: PMC8075361 DOI: 10.2147/ndt.s300773] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/02/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND We have previously demonstrated that biliverdin has neuroprotective effects that ameliorate cerebral ischemia/reperfusion (I/R) injury in rats. However, the underlying mechanism is unknown. This study aimed at elucidating on the modulatory role of miR-27a-3p on Rgs1 as a mechanism by which biliverdin affects cerebral I/R injury. METHODS Middle cerebral artery occlusion/reperfusion (MCAO/R) was used to establish I/R rat models while oxygen glucose deprivation/reoxygenation (OGD/R) was used to induce hippocampal neurons to establish I/R models in vitro. Infarct volume was assessed by TTC staining. Apoptotic analyses of ischemic cortical neurons and cells were performed by TUNEL staining and flow cytometry, respectively. Cell viability was assessed by the CCK-8 assay while the target of miR-27a-3p was determined by double luciferase reporter assay. Relative expression levels of miR-27a-3p and Rgs1 (in vivo and in vitro) as well as markers of inflammation and apoptosis (in vitro) were detected by RT-qPCR. Then, Elisa and western blot were used to assess protein expression levels of inflammatory and apoptotic markers in vitro. RESULTS Biliverdin suppressed inflammation and apoptosis in hippocampal neurons upon OGD/R, and reduced cerebral infarction volume as well as apoptosis in the MCAO/R rat model. Furthermore, biliverdin upregulated miR-27a-3p and downregulated hippocampal neuron Rgs1 after OGD/R as well as in rat brain tissues after cerebral I/R. Bioinformatic analysis revealed an miR-27a-3p docking site in the 3'-UTR region of Rgs1. Luciferase reporter assays showed that Rgs1 is an miR-27a-3p target. Moreover, miR-27a-3p upregulation inhibited OGD/R-triggered inflammation and suppressed neuronal apoptosis. Rgs1 knockdown suppressed OGD/R-triggered inflammation and decreased neuronal apoptosis while miR-27a-3p downregulation reversed the protective effect of Rgs1 knockdown. Moreover, miR-27a-3p overexpression and Rgs1 silencing suppressed NF-κB (p65) expression. CONCLUSION Biliverdin protects against cerebral I/R injury by regulating the miR-27a-3p/Rgs1 axis, thereby inhibiting inflammation and apoptosis.
Collapse
Affiliation(s)
- Junjie Li
- Department of Anesthesiology, First Affiliated Hospital, Kunming Medical University, Kunming City, 650032, People's Republic of China
| | - Lijia Peng
- Department of Anesthesiology, First Affiliated Hospital, Kunming Medical University, Kunming City, 650032, People's Republic of China
| | - Wenya Bai
- Department of Anesthesiology, First Affiliated Hospital, Kunming Medical University, Kunming City, 650032, People's Republic of China
| | - Peihua Peng
- Department of Anesthesiology, First Affiliated Hospital, Kunming Medical University, Kunming City, 650032, People's Republic of China
| | - Wendong Chen
- Department of Anesthesiology, First Affiliated Hospital, Kunming Medical University, Kunming City, 650032, People's Republic of China
| | - Wei Yang
- Department of Anesthesiology, First Affiliated Hospital, Kunming Medical University, Kunming City, 650032, People's Republic of China
| | - Jianlin Shao
- Department of Anesthesiology, First Affiliated Hospital, Kunming Medical University, Kunming City, 650032, People's Republic of China
| |
Collapse
|
25
|
Maia J, Otake AH, Poças J, Carvalho AS, Beck HC, Magalhães A, Matthiesen R, Strano Moraes MC, Costa-Silva B. Transcriptome Reprogramming of CD11b + Bone Marrow Cells by Pancreatic Cancer Extracellular Vesicles. Front Cell Dev Biol 2020; 8:592518. [PMID: 33330473 PMCID: PMC7729189 DOI: 10.3389/fcell.2020.592518] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancers (PC) are highly metastatic with poor prognosis, mainly due to delayed detection. We previously showed that PC-derived extracellular vesicles (EVs) act on macrophages residing in the liver, eliciting extracellular matrix remodeling in this organ and marked hepatic accumulation of CD11b+ bone marrow (BM) cells, which support PC liver metastasis. We here show that PC-EVs also bind to CD11b+ BM cells and induce the expansion of this cell population. Transcriptomic characterization of these cells shows that PC-EVs upregulate IgG and IgA genes, which have been linked to the presence of monocytes/macrophages in tumor microenvironments. We also report here the transcriptional downregulation of genes linked to monocyte/macrophage activation, trafficking, and expression of inflammatory molecules. Together, these results show for the first time the existence of a PC-BM communication axis mediated by EVs with a potential role in PC tumor microenvironments.
Collapse
Affiliation(s)
- Joana Maia
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Lisbon, Portugal
- Graduate Program in Areas of Basic and Applied Biology, University of Porto, Porto, Portugal
| | - Andreia Hanada Otake
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Lisbon, Portugal
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Juliana Poças
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP – Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Ana Sofia Carvalho
- Computational and Experimental Biology Group, CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Hans Christian Beck
- Centre for Clinical Proteomics, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Ana Magalhães
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP – Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Rune Matthiesen
- Computational and Experimental Biology Group, CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | | | - Bruno Costa-Silva
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Lisbon, Portugal
| |
Collapse
|
26
|
Lindquist Liljeqvist M, Hultgren R, Bergman O, Villard C, Kronqvist M, Eriksson P, Roy J. Tunica-Specific Transcriptome of Abdominal Aortic Aneurysm and the Effect of Intraluminal Thrombus, Smoking, and Diameter Growth Rate. Arterioscler Thromb Vasc Biol 2020; 40:2700-2713. [PMID: 32907367 DOI: 10.1161/atvbaha.120.314264] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE There is no medical treatment to prevent abdominal aortic aneurysm (AAA) growth and rupture, both of which are linked to smoking. Our objective was to map the tunica-specific pathophysiology of AAA with consideration of the intraluminal thrombus, age, and sex, and to subsequently identify which mechanisms were linked to smoking and diameter growth rate. Approach and Results: Microarray analyses were performed on 246 samples from 76 AAA patients and 13 controls. In media and adventitia, there were 5889 and 2701 differentially expressed genes, respectively. Gene sets related to adaptive and innate immunity were upregulated in both tunicas. Media-specific gene sets included increased matrix disassembly and angiogenesis, as well as decreased muscle cell development, contraction, and differentiation. Genes implicated in previous genome-wide association studies were dysregulated in media. The intraluminal thrombus had a pro-proteolytic and proinflammatory effect on the underlying media. Active smoking resulted in increased inflammation, oxidative stress, and angiogenesis in all tissues and enriched lipid metabolism in adventitia. Processes enriched with active smoking in control aortas overlapped to a high extent with those differentially expressed between AAAs and controls. The AAA diameter growth rate (n=24) correlated with T- and B-cell expression in media, as well as lipid-related processes in the adventitia. CONCLUSIONS This tunica-specific analysis of gene expression in a large study enabled the detection of features not previously described in AAA disease. Smoking was associated with increased expression of aneurysm-related processes, of which adaptive immunity and lipid metabolism correlated with growth rate.
Collapse
Affiliation(s)
- Moritz Lindquist Liljeqvist
- Department of Molecular Medicine and Surgery (M.L.L., R.H., C.V., M.K., J.R.), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Rebecka Hultgren
- Department of Molecular Medicine and Surgery (M.L.L., R.H., C.V., M.K., J.R.), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Department of Vascular Surgery, Karolinska University Hospital, Stockholm, Sweden (R.H., J.R.)
| | - Otto Bergman
- Department of Medicine (O.B., P.E.), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Christina Villard
- Department of Molecular Medicine and Surgery (M.L.L., R.H., C.V., M.K., J.R.), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Malin Kronqvist
- Department of Molecular Medicine and Surgery (M.L.L., R.H., C.V., M.K., J.R.), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Per Eriksson
- Department of Medicine (O.B., P.E.), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Joy Roy
- Department of Molecular Medicine and Surgery (M.L.L., R.H., C.V., M.K., J.R.), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Department of Vascular Surgery, Karolinska University Hospital, Stockholm, Sweden (R.H., J.R.)
| |
Collapse
|
27
|
Yuan G, Yang S, Ng A, Fu C, Oursler MJ, Xing L, Yang S. RGS12 Is a Novel Critical NF-κB Activator in Inflammatory Arthritis. iScience 2020; 23:101172. [PMID: 32512384 PMCID: PMC7281782 DOI: 10.1016/j.isci.2020.101172] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/15/2019] [Accepted: 05/12/2020] [Indexed: 11/21/2022] Open
Abstract
Rheumatoid arthritis (RA) is the most common inflammatory disease, which currently lacks effective treatment. Here, we discovered that the Regulator of G Protein Signaling 12 (RGS12) plays a key role in regulating inflammation. Transcriptional and protein analysis revealed that RGS12 was upregulated in human and mouse RA macrophages. Deletion of RGS12 in myeloid lineage or globally inhibits the development of collagen-induced arthritis including joint swelling and bone destruction. Mechanistically, RGS12 associates with NF-κB(p65) to activate its phosphorylation and nuclear translocation through PTB domain, and NF-κB(p65) regulates RGS12 expression in a transcriptional manner. The nuclear translocation ability of NF-κB(p65) and RGS12 can both be enhanced by cyclooxygenase-2 (COX2). Furthermore, ablation of RGS12 via RNA interference significantly blocks the inflammatory process in vivo and in vitro. These results demonstrate that RGS12 plays a critical role in the pathogenesis of inflammatory arthritis.
Collapse
Affiliation(s)
- Gongsheng Yuan
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shuting Yang
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Ng
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY, USA
| | - Chuanyun Fu
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Merry Jo Oursler
- Department of Medicine, Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Shuying Yang
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA; The Penn Center for Musculoskeletal Disorders, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
28
|
He Z, Xu X, Chen C, Li H, Wang DW. Adenosine 2A Receptor Activation Contributes to Ang II–Induced Aortic Remodeling by Promoting Macrophage Retention. Hypertension 2020; 75:119-130. [DOI: 10.1161/hypertensionaha.119.13709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The A
2A
R (adenosine 2A receptor) plays a crucial role in the pathophysiological process of cardiovascular diseases, yet its effect on aortic remodeling remains unclear. We observed elevated adenosine and A
2A
R levels following infusion of mice with Ang II (angiotensin II), suggesting a potential role for the adenosine-A
2A
R system in macrophage accumulation and subsequent aortic remodeling. The effects and mechanisms of A
2A
R on macrophage dynamics during aortic remodeling were further investigated using mice with macrophage knockout of A
2A
R and by transplantation of A
2A
R
−/−
bone marrow. We demonstrated that macrophage knockout of A
2A
R inhibited macrophage accumulation and subsequent aortic remodeling by inhibiting macrophage retention. This was shown to occur via promotion of macrophage emigration to the draining lymph node. These effects correlated with restoration of the expression and surface content of CCR7 (CC chemokine receptor 7). Consistently, A
2A
R
−/−
bone marrow transplantation relieved Ang II–induced aortic remodeling, macrophage retention, and CCR7 downregulation and internalization, all of which were rescued by A
2A
R
+
/
+
bone marrow transplantation. In addition, CCR7 antibody treatment blocked all the protective effects observed in A
2A
R-cKO mice, including attenuation of aortic remodeling and decreased macrophage retention. In in vitro studies, A
2A
R activation induced by Ang II suppressed macrophage migration to CCL19 (CC-chemokine ligand) 19 through downregulation and internalization of CCR7. In summary, A
2A
R activation contributes to Ang II–induced macrophage retention and subsequent aortic remodeling by inhibiting migration of macrophages to the draining lymph node through regulating CCR7 expression and internalization.
Collapse
Affiliation(s)
- Zuowen He
- From the Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiological Disorders, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
| | - Xizhen Xu
- From the Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiological Disorders, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
| | - Chen Chen
- From the Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiological Disorders, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
| | - Huaping Li
- From the Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiological Disorders, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
| | - Dao Wen Wang
- From the Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiological Disorders, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
| |
Collapse
|
29
|
Das A, Sudhahar V, Ushio-Fukai M, Fukai T. Novel interaction of antioxidant-1 with TRAF4: role in inflammatory responses in endothelial cells. Am J Physiol Cell Physiol 2019; 317:C1161-C1171. [PMID: 31553645 DOI: 10.1152/ajpcell.00264.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
NADPH oxidase (NOX)-derived reactive oxygen species (ROS) and copper (Cu), an essential micronutrient, have been implicated in vascular inflammatory diseases. We reported that in proinflammatory cytokine TNF-α-stimulated endothelial cells (ECs), cytosolic Cu chaperone antioxidant-1 (Atox1) functions as a Cu-dependent transcription factor for the NOX organizer p47phox, thereby increasing ROS-dependent inflammatory gene expression. However, the role and mechanism of Atox1 nuclear translocation in inflamed ECs remain unclear. Using enface staining and nuclear fractionation, here we show that Atox1 was localized in the nucleus in inflamed aortas from ApoE-/- mice with angiotensin II infusion on a high-fat diet, while it was found in cytosol in those from control mice. In cultured human ECs, TNF-α stimulation promoted Atox1 nuclear translocation within 15 min, which was associated with Atox1 binding to TNF-α receptor-associated factor 4 (TRAF4) in a Cu-dependent manner. TRAF4 depletion by siRNA significantly inhibited Atox1 nuclear translocation, p47phox expression, and ROS production as well as its downstream VCAM1/ICAM1 expression and monocyte adhesion to inflamed ECs, which were rescued by overexpression of nuclear targeted Atox1. Furthermore, Atox1 colocalized with TRAF4 at the nucleus in TNF-α-stimulated inflamed ECs and vessels. In summary, Cu-dependent Atox1 binding to TRAF4 plays an important role in Atox1 nuclear translocation and ROS-dependent inflammatory responses in TNF-α-stimulated ECs. Thus the Atox1-TRAF4 axis is a novel therapeutic target for vascular inflammatory disease such as atherosclerosis.
Collapse
Affiliation(s)
- Archita Das
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia.,Departments of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Varadarajan Sudhahar
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia.,Departments of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia.,Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | - Masuko Ushio-Fukai
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia.,Department of Medicine (Cardiology), Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Tohru Fukai
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia.,Departments of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia.,Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| |
Collapse
|
30
|
Tissier R, Hocini H, Tchitchek N, Deye N, Legriel S, Pichon N, Daubin C, Hermine O, Carli P, Vivien B, Tréluyer JM, Lefebvre C, Tisserand P, Dubois-Randé JL, Berdeaux A, Ghaleh B, Lelièvre JD, Levy Y, Cariou A. Early blood transcriptomic signature predicts patients' outcome after out-of-hospital cardiac arrest. Resuscitation 2019; 138:222-232. [PMID: 30885824 DOI: 10.1016/j.resuscitation.2019.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/27/2019] [Accepted: 03/10/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Early prognostication is a major challenge after out-of-hospital cardiac arrest (OHCA). AIMS We hypothesized that a genome-wide analysis of blood gene expression could offer new prognostic tools and lines of research. METHODS Sixty-nine patients were enrolled from an ancillary study of the clinical trial NCT00999583 that tested the effect of erythropoietin (EPO) after OHCA. Blood samples were collected in comatose survivors of OHCA at hospital admission and 1 and 3 days after resuscitation. Gene expression profiles were analyzed (Illumina HumanHT-12 V4 BeadChip; >34,000 genes). Patients were classified into two categories representing neurological favorable outcome (cerebral performance category [CPC] = 1-2) vs unfavorable outcome (CPC > 2) at Day 60 after OHCA. Differential and functional enrichment analyses were performed to compare transcriptomic profiles between these two categories. RESULTS Among the 69 enrolled patients, 33 and 36 patients were treated or not by EPO, respectively. Among them, 42% had a favorable neurological outcome in both groups. EPO did not affect the transcriptomic response at Day-0 and 1 after OHCA. In contrast, 76 transcripts differed at Day-0 between patients with unfavorable vs favorable neurological outcome. This signature persisted at Day-1 after OHCA. Functional enrichment analysis revealed a down-regulation of adaptive immunity with concomitant up-regulation of innate immunity and inflammation in patients with unfavorable vs favorable neurological outcome. The transcription of many genes of the HLA family was decreased in patients with unfavorable vs favorable neurological outcome. Concomitantly, neutrophil activation and inflammation were observed. Up-stream regulators analysis showed the implication of numerous factors involved in cell cycle and damages. A logistic regression including a set of genes allowed a reliable prediction of the clinical outcomes (specificity = 88%; Hit Rate = 83%). CONCLUSIONS A transcriptomic signature involving a counterbalance between adaptive and innate immune responses is able to predict neurological outcome very early after hospital admission after OHCA. This deserves confirmation in a larger population.
Collapse
Affiliation(s)
- Renaud Tissier
- Inserm, U955, F94000, Créteil, France; Université Paris Est, UMR_S955, UPEC, Ecole Nationale Vétérinaire d'Alfort, F-94000, Créteil, France.
| | - Hakim Hocini
- Inserm, U955, F94000, Créteil, France; Vaccine Research Institute, Université Paris Est-Créteil, F-94000, Créteil, France
| | - Nicolas Tchitchek
- Vaccine Research Institute, Université Paris Est-Créteil, F-94000, Créteil, France; CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Infrastructure, F-92265 Fontenay-aux-Roses, France
| | - Nicolas Deye
- Medical ICU, Inserm U942, Lariboisiere Hospital, APHP, F-75010, Paris, France
| | - Stéphane Legriel
- Intensive Care Unit, Versailles Hospital, Le Chesnay, F-78150, France
| | - Nicolas Pichon
- Intensive Care Unit, University Hospital Dupuytren, Limoges, F-87042, France
| | - Cédric Daubin
- CHU de Caen, Department of Medical Intensive Care, Caen, F-14000, France
| | - Olivier Hermine
- Department of Hematology and INSERM U1163 CNRS ERL 8654, Imagine Institute and Necker Hospital, Paris, F-75015, France
| | - Pierre Carli
- SAMU de Paris, Service d'Anesthésie-Réanimation, Hôpital Universitaire Necker- Enfants Malades, Université Paris Descartes, F-75015, Paris, France
| | - Benoît Vivien
- SAMU de Paris, Service d'Anesthésie-Réanimation, Hôpital Universitaire Necker- Enfants Malades, Université Paris Descartes, F-75015, Paris, France
| | - Jean-Marc Tréluyer
- Clinical Research Unit, Paris Centre and Paris Descartes University, Paris, France
| | - Cécile Lefebvre
- Inserm, U955, F94000, Créteil, France; Vaccine Research Institute, Université Paris Est-Créteil, F-94000, Créteil, France
| | - Pascaline Tisserand
- Inserm, U955, F94000, Créteil, France; Vaccine Research Institute, Université Paris Est-Créteil, F-94000, Créteil, France
| | - Jean-Luc Dubois-Randé
- Inserm, U955, F94000, Créteil, France; Université Paris Est, UMR_S955, UPEC, Ecole Nationale Vétérinaire d'Alfort, F-94000, Créteil, France
| | - Alain Berdeaux
- Inserm, U955, F94000, Créteil, France; Université Paris Est, UMR_S955, UPEC, Ecole Nationale Vétérinaire d'Alfort, F-94000, Créteil, France
| | - Bijan Ghaleh
- Inserm, U955, F94000, Créteil, France; Université Paris Est, UMR_S955, UPEC, Ecole Nationale Vétérinaire d'Alfort, F-94000, Créteil, France; APHP, Hôpitaux Universitaires Henri Mondor, Plateforme de Ressources Biologiques, F-94000, Créteil, France
| | - Jean-Daniel Lelièvre
- Inserm, U955, F94000, Créteil, France; Vaccine Research Institute, Université Paris Est-Créteil, F-94000, Créteil, France
| | - Yves Levy
- Inserm, U955, F94000, Créteil, France; Vaccine Research Institute, Université Paris Est-Créteil, F-94000, Créteil, France.
| | - Alain Cariou
- Service de Réanimation Médicale, Hôpitaux Universitaires Paris Centre, Hôpital Cochin, Paris, France
| |
Collapse
|
31
|
Zou ZY, Liu J, Chang C, Li JJ, Luo J, Jin Y, Ma Z, Wang TH, Shao JL. Biliverdin administration regulates the microRNA-mRNA expressional network associated with neuroprotection in cerebral ischemia reperfusion injury in rats. Int J Mol Med 2019; 43:1356-1372. [PMID: 30664169 PMCID: PMC6365090 DOI: 10.3892/ijmm.2019.4064] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 12/18/2018] [Indexed: 12/21/2022] Open
Abstract
Inflammatory response has an important role in the outcome of cerebral ischemia reperfusion injury (CIR). Biliverdin (BV) administration can relieve CIR in rats, but the mechanism remains unknown. The aim of the present study was to explore the expressional network of microRNA (miRNA)-mRNA in CIR rats following BV administration. A rat middle cerebral artery occlusion model with BV treatment was established. After neurobehavior was evaluated by neurological severity scores (NSS), miRNA and mRNA expressional profiles were analyzed by microarray technology from the cerebral cortex subjected to ischemia and BV administration. Then, bioinformatics prediction was used to screen the correlation between miRNA and mRNA, and 20 candidate miRNAs and 33 candidate mRNAs were verified by reverse transcription-quantitative polymerase chain reaction. Furthermore, the regulation relationship between ETS proto-oncogene 1 (Ets1) and miRNA204-5p was examined by luciferase assay. A total of 86 miRNAs were differentially expressed in the BV group compared with the other groups. A total of 10 miRNAs and 26 candidate genes were identified as a core 'microRNA-mRNA' regulatory network that was linked with the functional improvement of BV administration in CIR rats. Lastly, the luciferase assay results confirmed that miRNA204-5p directly targeted Ets1. The present findings suggest that BV administration may regulate multiple miRNAs and mRNAs to improve neurobehavior in CIR rats, by influencing cell proliferation, apoptosis, maintaining ATP homeostasis, and angiogenesis.
Collapse
Affiliation(s)
- Zhi-Yao Zou
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Jia Liu
- Experimental Animal Center, Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Cheng Chang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Jun-Jie Li
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Jing Luo
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Yuan Jin
- Experimental Animal Center, Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Zheng Ma
- Experimental Animal Center, Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Ting-Hua Wang
- Experimental Animal Center, Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Jian-Lin Shao
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| |
Collapse
|
32
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol Rev 2018; 98:1627-1738. [PMID: 29873596 DOI: 10.1152/physrev.00038.2017] [Citation(s) in RCA: 663] [Impact Index Per Article: 110.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - George W Booz
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Curt D Sigmund
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Thomas M Coffman
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
33
|
Squires KE, Montañez-Miranda C, Pandya RR, Torres MP, Hepler JR. Genetic Analysis of Rare Human Variants of Regulators of G Protein Signaling Proteins and Their Role in Human Physiology and Disease. Pharmacol Rev 2018; 70:446-474. [PMID: 29871944 DOI: 10.1124/pr.117.015354] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Regulators of G protein signaling (RGS) proteins modulate the physiologic actions of many neurotransmitters, hormones, and other signaling molecules. Human RGS proteins comprise a family of 20 canonical proteins that bind directly to G protein-coupled receptors/G protein complexes to limit the lifetime of their signaling events, which regulate all aspects of cell and organ physiology. Genetic variations account for diverse human traits and individual predispositions to disease. RGS proteins contribute to many complex polygenic human traits and pathologies such as hypertension, atherosclerosis, schizophrenia, depression, addiction, cancers, and many others. Recent analysis indicates that most human diseases are due to extremely rare genetic variants. In this study, we summarize physiologic roles for RGS proteins and links to human diseases/traits and report rare variants found within each human RGS protein exome sequence derived from global population studies. Each RGS sequence is analyzed using recently described bioinformatics and proteomic tools for measures of missense tolerance ratio paired with combined annotation-dependent depletion scores, and protein post-translational modification (PTM) alignment cluster analysis. We highlight selected variants within the well-studied RGS domain that likely disrupt RGS protein functions and provide comprehensive variant and PTM data for each RGS protein for future study. We propose that rare variants in functionally sensitive regions of RGS proteins confer profound change-of-function phenotypes that may contribute, in newly appreciated ways, to complex human diseases and/or traits. This information provides investigators with a valuable database to explore variation in RGS protein function, and for targeting RGS proteins as future therapeutic targets.
Collapse
Affiliation(s)
- Katherine E Squires
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Carolina Montañez-Miranda
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Rushika R Pandya
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Matthew P Torres
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - John R Hepler
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| |
Collapse
|
34
|
Perschbacher KJ, Deng G, Fisher RA, Gibson-Corley KN, Santillan MK, Grobe JL. Regulators of G protein signaling in cardiovascular function during pregnancy. Physiol Genomics 2018; 50:590-604. [PMID: 29702036 PMCID: PMC6139632 DOI: 10.1152/physiolgenomics.00037.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptor signaling mechanisms are implicated in many aspects of cardiovascular control, and dysfunction of such signaling mechanisms is commonly associated with disease states. Investigators have identified a large number of regulator of G protein signaling (RGS) proteins that variously contribute to the modulation of intracellular second-messenger signaling kinetics. These many RGS proteins each interact with a specific set of second-messenger cascades and receptor types and exhibit tissue-specific expression patterns. Increasing evidence supports the contribution of RGS proteins, or their loss, in the pathogenesis of cardiovascular dysfunctions. This review summarizes the current understanding of the functional contributions of RGS proteins, particularly within the B/R4 family, in cardiovascular disorders of pregnancy including gestational hypertension, uterine artery dysfunction, and preeclampsia.
Collapse
Affiliation(s)
| | - Guorui Deng
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
| | - Rory A Fisher
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
| | - Katherine N Gibson-Corley
- Department of Pathology, University of Iowa , Iowa City, Iowa
- UIHC Center for Hypertension Research, University of Iowa , Iowa City, Iowa
| | - Mark K Santillan
- Department of Obstetrics & Gynecology, University of Iowa , Iowa City, Iowa
- UIHC Center for Hypertension Research, University of Iowa , Iowa City, Iowa
- Abboud Cardiovascular Research Center, University of Iowa , Iowa City, Iowa
| | - Justin L Grobe
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
- UIHC Center for Hypertension Research, University of Iowa , Iowa City, Iowa
- Abboud Cardiovascular Research Center, University of Iowa , Iowa City, Iowa
- Fraternal Order of Eagles' Diabetes Research Center, University of Iowa , Iowa City, Iowa
- Obesity Education & Research Initiative, University of Iowa , Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa , Iowa City, Iowa
| |
Collapse
|
35
|
Batugedara HM, Li J, Chen G, Lu D, Patel JJ, Jang JC, Radecki KC, Burr AC, Lo DD, Dillman AR, Nair MG. Hematopoietic cell-derived RELMα regulates hookworm immunity through effects on macrophages. J Leukoc Biol 2018; 104:855-869. [PMID: 29992625 DOI: 10.1002/jlb.4a0917-369rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 06/06/2018] [Accepted: 06/14/2018] [Indexed: 12/29/2022] Open
Abstract
Resistin-like molecule α (RELMα) is a highly secreted protein in type 2 (Th2) cytokine-induced inflammation including helminth infection and allergy. In infection with Nippostrongylus brasiliensis (Nb), RELMα dampens Th2 inflammatory responses. RELMα is expressed by immune cells, and by epithelial cells (EC); however, the functional impact of immune versus EC-derived RELMα is unknown. We generated bone marrow (BM) chimeras that were RELMα deficient (RELMα-/ - ) in BM or non BM cells and infected them with Nb. Non BM RELMα-/- chimeras had comparable inflammatory responses and parasite burdens to RELMα+/+ mice. In contrast, both RELMα-/- and BM RELMα-/- mice exhibited increased Nb-induced lung and intestinal inflammation, correlated with elevated Th2 cytokines and Nb killing. CD11c+ lung macrophages were the dominant BM-derived source of RELMα and can mediate Nb killing. Therefore, we employed a macrophage-worm co-culture system to investigate whether RELMα regulates macrophage-mediated Nb killing. Compared to RELMα+ /+ macrophages, RELMα-/- macrophages exhibited increased binding to Nb and functionally impaired Nb development. Supplementation with recombinant RELMα partially reversed this phenotype. Gene expression analysis revealed that RELMα decreased cell adhesion and Fc receptor signaling pathways, which are associated with macrophage-mediated helminth killing. Collectively, these studies demonstrate that BM-derived RELMα is necessary and sufficient to dampen Nb immune responses, and identify that one mechanism of action of RELMα is through inhibiting macrophage recruitment and interaction with Nb. Our findings suggest that RELMα acts as an immune brake that provides mutually beneficial effects for the host and parasite by limiting tissue damage and delaying parasite expulsion.
Collapse
Affiliation(s)
- Hashini M Batugedara
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Jiang Li
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Gang Chen
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Dihong Lu
- Department of Nematology, University of California Riverside, Riverside, California, USA
| | - Jay J Patel
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Jessica C Jang
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Kelly C Radecki
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Abigail C Burr
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - David D Lo
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Adler R Dillman
- Department of Nematology, University of California Riverside, Riverside, California, USA
| | - Meera G Nair
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| |
Collapse
|
36
|
Chan EC, Ren C, Xie Z, Jude J, Barker T, Koziol-White CA, Ma M, Panettieri RA, Wu D, Rosenberg HF, Druey KM. Regulator of G protein signaling 5 restricts neutrophil chemotaxis and trafficking. J Biol Chem 2018; 293:12690-12702. [PMID: 29929985 DOI: 10.1074/jbc.ra118.002404] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/14/2018] [Indexed: 01/09/2023] Open
Abstract
Neutrophils are white blood cells that are mobilized to damaged tissues and to sites of pathogen invasion, providing the first line of host defense. Chemokines displayed on the surface of blood vessels promote migration of neutrophils to these sites, and tissue- and pathogen-derived chemoattractant signals, including N-formylmethionylleucylphenylalanine (fMLP), elicit further migration to sites of infection. Although nearly all chemoattractant receptors use heterotrimeric G proteins to transmit signals, many of the mechanisms lying downstream of chemoattractant receptors that either promote or limit neutrophil motility are incompletely defined. Here, we show that regulator of G protein signaling 5 (RGS5), a protein that modulates G protein activity, is expressed in both human and murine neutrophils. We detected significantly more neutrophils in the airways of Rgs5-/- mice than WT counterparts following acute respiratory virus infection and in the peritoneum in response to injection of thioglycollate, a biochemical proinflammatory stimulus. RGS5-deficient neutrophils responded with increased chemotaxis elicited by the chemokines CXC motif chemokine ligand 1 (CXCL1), CXCL2, and CXCL12 but not fMLP. Moreover, adhesion of these cells was increased in the presence of both CXCL2 and fMLP. In summary, our results indicate that RGS5 deficiency increases chemotaxis and adhesion, leading to more efficient neutrophil mobilization to inflamed tissues in mice. These findings suggest that RGS5 expression and activity in neutrophils determine their migrational patterns in the complex microenvironments characteristic of inflamed tissues.
Collapse
Affiliation(s)
- Eunice C Chan
- Molecular Signal Transduction Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Chunguang Ren
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Zhihui Xie
- Molecular Signal Transduction Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Joseph Jude
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers New Jersey School of Medicine, Rutgers, New Jersey 07103
| | - Tolga Barker
- Molecular Signal Transduction Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Cynthia A Koziol-White
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers New Jersey School of Medicine, Rutgers, New Jersey 07103
| | - Michelle Ma
- Inflammation Immunobiology Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers New Jersey School of Medicine, Rutgers, New Jersey 07103
| | - Dianqing Wu
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Helene F Rosenberg
- Inflammation Immunobiology Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Kirk M Druey
- Molecular Signal Transduction Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892.
| |
Collapse
|
37
|
Patel J, Chuaiphichai S, Douglas G, Gorvin CM, Channon KM. Vascular wall regulator of G-protein signalling-1 (RGS-1) is required for angiotensin II-mediated blood pressure control. Vascul Pharmacol 2018; 108:15-22. [PMID: 29654907 PMCID: PMC6073721 DOI: 10.1016/j.vph.2018.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 03/18/2018] [Accepted: 04/05/2018] [Indexed: 02/07/2023]
Abstract
G-Protein coupled receptors (GPCRs) activate intracellular signalling pathways by coupling to heterotrimeric G-proteins that control many physiological processes including blood pressure homeostasis. The Regulator of G-Protein Signalling-1 (RGS1) controls the magnitude and duration of downstream GPCR signalling by acting as a GTPase-activating protein for specific Gα-proteins. RGS1 has contrasting roles in haematopoietic and non-haematopoietic cells. Rgs1−/−ApoE−/− mice are protected from Angiotensin II (Ang II)-induced aortic aneurysm rupture. Conversely, Ang II treatment increases systolic blood pressure to a greater extent in Rgs1−/−ApoE−/− mice than ApoE−/− mice, independent of its role in myeloid cells. However the precise role of RGS1 in hypertension and vascular-derived cells remains unknown. We determined the effects of Rgs1 deletion on vascular function in ApoE−/− mice. Rgs1 deletion led to enhanced vasoconstriction in aortas and mesenteric arteries from ApoE−/− mice in response to phenylephrine (PE) and U46619 respectively. Rgs1 was shown to have a role in the vasculature, with endothelium-dependent vasodilation being impaired, and endothelium-independent dilatation to SNP being enhanced in Rgs1−/−ApoE−/− mesenteric arteries. To address the downstream signalling pathways in vascular smooth muscle cells (VSMCs) in response to Ang II-stimulation, we assessed pErk1/2, pJNK and pp38 MAPK activation in VSMCs transiently transfected with Rgs1. pErk1/2 signalling but not pJNK and pp38 signalling was impaired in the presence of Rgs1. Furthermore, we demonstrated that the enhanced contractile response to PE in Rgs1−/−ApoE−/− aortas was reduced by a MAPK/Erk (MEK) inhibitor and an L-type voltage gated calcium channel antagonist, suggesting that Erk1/2 signalling and calcium influx are major effectors of Rgs1-mediated vascular contractile responses, respectively. These findings indicate RGS1 is a novel regulator of blood pressure homeostasis and highlight RGS1-controlled signalling pathways in the vasculature that may be new drug development targets for hypertension.
Collapse
MESH Headings
- Angiotensin II
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/physiopathology
- Blood Pressure/genetics
- Calcium Signaling
- Cell Line
- Disease Models, Animal
- Hypertension/chemically induced
- Hypertension/genetics
- Hypertension/metabolism
- Hypertension/physiopathology
- Male
- Mesenteric Arteries/metabolism
- Mesenteric Arteries/physiopathology
- Mice, Knockout, ApoE
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Phosphorylation
- RGS Proteins/deficiency
- RGS Proteins/genetics
- RGS Proteins/metabolism
- Receptor, Angiotensin, Type 1/metabolism
- Vasoconstriction
- Vasodilation
Collapse
Affiliation(s)
- Jyoti Patel
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK.
| | - Surawee Chuaiphichai
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Gillian Douglas
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Caroline M Gorvin
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
| | - Keith M Channon
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| |
Collapse
|
38
|
Connaughton EP, Naicker S, Hanley SA, Slevin SM, Eykelenboom JK, Lowndes NF, O'Brien T, Ceredig R, Griffin MD, Dennedy MC. Phenotypic and functional heterogeneity of human intermediate monocytes based on HLA-DR expression. Immunol Cell Biol 2018; 96:742-758. [PMID: 29505094 DOI: 10.1111/imcb.12032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 01/07/2018] [Accepted: 02/27/2018] [Indexed: 12/24/2022]
Abstract
Human blood monocytes are subclassified as classical, intermediate and nonclassical. In this study, it was shown that conventionally defined human intermediate monocytes can be divided into two distinct subpopulations with mid- and high-level surface expression of HLA-DR (referred to as DRmid and DRhi intermediate monocytes). These IM subpopulations were phenotypically and functionally characterized in healthy adult blood by flow cytometry, migration assays and lipoprotein uptake assays. Their absolute numbers and proportions were then compared in blood samples from obese and nonobese adults. DRmid and DRhi intermediate monocytes differentially expressed several proteins including CD62L, CD11a, CX3CR1 and CCR2. Overall, the DRmid intermediate monocytes surface profile more closely resembled that of classical monocytes while DRhi intermediate monocytes were more similar to nonclassical. However, in contrast to classical monocytes, DRmid intermediate monocytes migrated weakly to CCL2, had reduced intracellular calcium flux following CCR2 ligation and favored adherence to TNFα-activated endothelium over transmigration. In lipid uptake assays, DRmid intermediate monocytes demonstrated greater internalization of oxidized and acetylated low-density lipoprotein than DRhi intermediate monocytes. In obese compared to nonobese adults, proportions and absolute numbers of DRmid , but not DRhi intermediate monocytes, were increased in blood. The results are consistent with phenotypic and functional heterogeneity within the intermediate monocytes subset that may be of specific relevance to lipoprotein scavenging and metabolic health.
Collapse
Affiliation(s)
- Eanna P Connaughton
- School of Medicine, College of Medicine, Nursing and Health Sciences, Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Serika Naicker
- School of Medicine, College of Medicine, Nursing and Health Sciences, Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Shirley A Hanley
- School of Medicine, College of Medicine, Nursing and Health Sciences, Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Stephanie M Slevin
- School of Medicine, College of Medicine, Nursing and Health Sciences, Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - John K Eykelenboom
- Centre for Chromosomal Biology, Department of Biochemistry, School of Natural Sciences, College of Science, National University of Ireland, Galway, Ireland
| | - Noel F Lowndes
- Centre for Chromosomal Biology, Department of Biochemistry, School of Natural Sciences, College of Science, National University of Ireland, Galway, Ireland
| | - Timothy O'Brien
- School of Medicine, College of Medicine, Nursing and Health Sciences, Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Rhodri Ceredig
- School of Medicine, College of Medicine, Nursing and Health Sciences, Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Matthew D Griffin
- School of Medicine, College of Medicine, Nursing and Health Sciences, Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Michael C Dennedy
- School of Medicine, College of Medicine, Nursing and Health Sciences, Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
- Discipline of Pharmacology and Therapeutics, School of Medicine, Lambe Institute for Translational Medicine, National University of Ireland, Galway, Ireland
| |
Collapse
|
39
|
Moncrieffe H, Bennett MF, Tsoras M, Luyrink LK, Johnson AL, Xu H, Dare J, Becker ML, Prahalad S, Rosenkranz M, O'Neil KM, Nigrovic PA, Griffin TA, Lovell DJ, Grom AA, Medvedovic M, Thompson SD. Transcriptional profiles of JIA patient blood with subsequent poor response to methotrexate. Rheumatology (Oxford) 2017; 56:1542-1551. [PMID: 28582527 DOI: 10.1093/rheumatology/kex206] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Indexed: 11/13/2022] Open
Abstract
Objective The mechanisms that determine the efficacy or inefficacy of MTX in JIA are ill-defined. The objective of this study was to identify a gene expression transcriptional signature associated with poor response to MTX in patients with JIA. Methods RNA sequencing was used to measure gene expression in peripheral blood mononuclear cells collected from 47 patients with JIA prior to MTX treatment and 14 age-matched controls. Differentially expressed baseline genes between responders and non-responders were evaluated. Biological differences between all JIA patients and controls were explored by constructing a signature of differentially expressed genes. Unsupervised clustering and pathway analysis was performed. Results A signature of 99 differentially expressed genes (Bonferroni-corrected P < 0.05) capturing the biological differences between all JIA patients and controls was identified. Unsupervised clustering of samples based on this list of 99 genes produced subgroups enriched for MTX response status. Comparing this gene signature with reference signatures from sorted cell populations revealed high concordance between the expression signatures of monocytes and of MTX non-responders. CXCL8 (IL-8) was the most significantly differentially expressed gene transcript comparing all JIA patients with controls (Bonferroni-corrected P = 4.12 × 10-10). Conclusion Variability in clinical response to MTX in JIA patients is associated with differences in gene transcripts modulated in monocytes. These gene expression profiles may provide a basis for biomarkers predictive of treatment response.
Collapse
Affiliation(s)
- Halima Moncrieffe
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center.,Department of Pediatrics
| | - Mark F Bennett
- Department of Environmental Health, University of Cincinnati
| | - Monica Tsoras
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center
| | - Lorie K Luyrink
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center
| | - Anne L Johnson
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Huan Xu
- Department of Environmental Health, University of Cincinnati
| | - Jason Dare
- Pediatrics/Rheumatology, UAMS, Little Rock, AR
| | - Mara L Becker
- Pediatrics, Section of Rheumatology, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | - Sampath Prahalad
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | | | | | - Peter A Nigrovic
- Division of Immunology, Boston Children's Hospital.,Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA
| | | | - Daniel J Lovell
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Alexei A Grom
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | | | - Susan D Thompson
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center.,Department of Pediatrics
| |
Collapse
|
40
|
Abstract
As the largest receptor gene family in the human genome, with >800 members, the signal-transducing G protein-coupled receptors (GPCRs) play critical roles in nearly all conceivable physiological processes, ranging from the sensing of photons and odorants to metabolic homeostasis and migration of leukocytes. Unfortunately, an exhaustive review of the several hundred GPCRs expressed by myeloid cells/macrophages (P.J. Groot-Kormelink, L .Fawcett, P.D. Wright, M. Gosling, and T.C. Kent, BMC Immunol 12:57, 2012, doi:10.1186/1471-2172-13-57) is beyond the scope of this chapter; however, we will endeavor to cover the GPCRs that contribute to the major facets of macrophage biology, i.e., those whose expression is restricted to macrophages and the GPCRs involved in macrophage differentiation/polarization, microbial elimination, inflammation and resolution, and macrophage-mediated pathology. The chemokine receptors, a major group of myeloid GPCRs, will not be extensively covered as they are comprehensively reviewed elsewhere.
Collapse
|
41
|
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening disease associated with high morbidity, and high mortality in the event of aortic rupture. Major advances in open surgical and endovascular repair of AAA have been achieved during the past 2 decades. However, drug-based therapies are still lacking, highlighting a real need for better understanding of the molecular and cellular mechanisms involved in AAA formation and progression. The main pathological features of AAA include extracellular matrix remodelling associated with degeneration and loss of vascular smooth muscle cells and accumulation and activation of inflammatory cells. The inflammatory process has a crucial role in AAA and substantially influences many determinants of aortic wall remodelling. In this Review, we focus specifically on the involvement of monocytes and macrophages, summarizing current knowledge on the roles, origin, and functions of these cells in AAA development and its complications. Furthermore, we show and propose that distinct monocyte and macrophage subsets have critical and differential roles in initiation, progression, and healing of the aneurysmal process. On the basis of experimental and clinical studies, we review potential translational applications to detect, assess, and image macrophage subsets in AAA, and discuss the relevance of these applications for clinical practice.
Collapse
|
42
|
Druey KM. Emerging Roles of Regulators of G Protein Signaling (RGS) Proteins in the Immune System. Adv Immunol 2017; 136:315-351. [PMID: 28950950 DOI: 10.1016/bs.ai.2017.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Kirk M Druey
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, NIAID/NIH, Bethesda, MD, United States.
| |
Collapse
|
43
|
Kehrl JH. The impact of RGS and other G-protein regulatory proteins on Gαi-mediated signaling in immunity. Biochem Pharmacol 2016; 114:40-52. [PMID: 27071343 DOI: 10.1016/j.bcp.2016.04.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 04/08/2016] [Indexed: 01/30/2023]
Abstract
Leukocyte chemoattractant receptors are members of the G-protein coupled receptor (GPCR) family. Signaling downstream of these receptors directs the localization, positioning and homeostatic trafficking of leukocytes; as well as their recruitment to, and their retention at, inflammatory sites. Ligand induced changes in the molecular conformation of chemoattractant receptors results in the engagement of heterotrimeric G-proteins, which promotes α subunits to undergo GTP/GDP exchange. This results in the functional release of βγ subunits from the heterotrimers, thereby activating downstream effector molecules, which initiate leukocyte polarization, gradient sensing, and directional migration. Pertussis toxin ADP ribosylates Gαi subunits and prevents chemoattractant receptors from triggering Gαi nucleotide exchange. The use of pertussis toxin revealed the essential importance of Gαi subunit nucleotide exchange for chemoattractant receptor signaling. More recent studies have identified a range of regulatory mechanisms that target these receptors and their associated heterotrimeric G-proteins, thereby helping to control the magnitude, kinetics, and duration of signaling. A failure in these regulatory pathways can lead to impaired receptor signaling and immunopathology. The analysis of mice with targeted deletions of Gαi isoforms as well as some of these G-protein regulatory proteins is providing insights into their roles in chemoattractant receptor signaling.
Collapse
Affiliation(s)
- John H Kehrl
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 2089, United States.
| |
Collapse
|
44
|
Xie Z, Chan EC, Druey KM. R4 Regulator of G Protein Signaling (RGS) Proteins in Inflammation and Immunity. AAPS JOURNAL 2015; 18:294-304. [PMID: 26597290 DOI: 10.1208/s12248-015-9847-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/11/2015] [Indexed: 11/30/2022]
Abstract
G protein-coupled receptors (GPCRs) have important functions in both innate and adaptive immunity, with the capacity to bridge interactions between the two arms of the host responses to pathogens through direct recognition of secreted microbial products or the by-products of host cells damaged by pathogen exposure. In the mid-1990s, a large group of intracellular proteins was discovered, the regulator of G protein signaling (RGS) family, whose main, but not exclusive, function appears to be to constrain the intensity and duration of GPCR signaling. The R4/B subfamily--the focus of this review--includes RGS1-5, 8, 13, 16, 18, and 21, which are the smallest RGS proteins in size, with the exception of RGS3. Prominent roles in the trafficking of B and T lymphocytes and macrophages have been described for RGS1, RGS13, and RGS16, while RGS18 appears to control platelet and osteoclast functions. Additional G protein independent functions of RGS13 have been uncovered in gene expression in B lymphocytes and mast cell-mediated allergic reactions. In this review, we discuss potential physiological roles of this RGS protein subfamily, primarily in leukocytes having central roles in immune and inflammatory responses. We also discuss approaches to target RGS proteins therapeutically, which represents a virtually untapped strategy to combat exaggerated immune responses leading to inflammation.
Collapse
Affiliation(s)
- Zhihui Xie
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, NIAID/NIH, 50 South Drive Room 4154, Bethesda, Maryland, 20892, USA
| | - Eunice C Chan
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, NIAID/NIH, 50 South Drive Room 4154, Bethesda, Maryland, 20892, USA
| | - Kirk M Druey
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, NIAID/NIH, 50 South Drive Room 4154, Bethesda, Maryland, 20892, USA.
| |
Collapse
|