1
|
Ye H, Li ZQ, Yang JM, Long Y, Zhong YB, Wu Y, Wang MY. A network pharmacology-based study to investigate the mechanism of curcumin-regulated regenerative repair of quadriceps femoris muscle in KOA rats. Eur J Pharmacol 2024; 982:176910. [PMID: 39154821 DOI: 10.1016/j.ejphar.2024.176910] [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: 05/05/2024] [Revised: 07/29/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Knee osteoarthritis (KOA) is a very common musculoskeletal disorder, and patients with KOA often exhibit significant quadriceps femoris muscle atrophy. It is well established that curcumin (CUR) exerts protective effects on skeletal muscle. However, the efficacy of CUR in treating KOA-induced quadriceps femoris muscle atrophy and its underlying mechanisms remain uncertain. In this study, we employed network pharmacology to investigate the mechanism by which CUR promotes regenerative repair of the quadriceps femoris muscle in rats with KOA. METHODS The potential targets of CUR were obtained from Swiss Target Prediction. The targets of skeletal muscle regeneration were identified from GeneCard and OMIM. A Venn diagram was generated to visualize the intersection of CUR targets and skeletal muscle regeneration targets, and the core targets were identified using STRING. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were conducted using DAVID. Finally, the network pharmacology results were further validated by establishing a KOA rat model using the Hulth method. RESULTS Network pharmacology analysis and molecular docking results revealed that CUR affects skeletal muscle regeneration through multiple targets and pathways. In vivo experimental results were validated by demonstrating that KOA causes atrophy and induces apoptosis in the quadriceps femoris muscle. Furthermore, CUR was shown to inhibit apoptosis in the quadriceps femoris muscle by regulating STAT3 and FOS, as well as the PI3K/AKT signaling pathway. CONCLUSIONS Our study revealed the apoptosis-inhibiting effects of CUR and its underlying mechanisms. Consequently, CUR has the potential to improve quadriceps femoris muscle atrophy caused by KOA.
Collapse
Affiliation(s)
- Hua Ye
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi, China; Gannan Medical University, Ganzhou City, Jiangxi, China
| | - Ze-Qin Li
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi, China; Gannan Medical University, Ganzhou City, Jiangxi, China
| | - Jia-Ming Yang
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi, China
| | - Yi Long
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi, China; Gannan Medical University, Ganzhou City, Jiangxi, China
| | - Yan-Biao Zhong
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi, China; Ganzhou Intelligent Rehabilitation Technology Innovation Center, Ganzhou City, Jiangxi, China
| | - Yi Wu
- Gannan Medical University, Ganzhou City, Jiangxi, China; Jiangxi Provincal Key Laboratory of Tissue Engineering (2024SSY06291), School of Pharmacy, Gannan Medical University, Gouzhou, Jiangxi, China.
| | - Mao-Yuan Wang
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi, China; Ganzhou Key Laboratory of Rehabilitation Medicine, GanZhou City, Jiangxi, China.
| |
Collapse
|
2
|
Zhang J, Zhang S, Qiao J, Qiu M, Li X. Risk factors analysis and risk assessment model construction of systemic lupus erythematosus patients with infection. Lupus 2023; 32:119-128. [PMID: 36433710 DOI: 10.1177/09612033221141255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To analyze the characteristics of peripheral blood lymphocyte subsets in systemic lupus erythematosus (SLE) patients with infection and non-infection group. Explore the risk factors of infection in SLE patients and establish a risk matrix model to predict the occurrence of co-infection. METHODS total of 333 SLE patients without infection, 163 patients suffering from infection, and 132 healthy controls (HCs) were recruited. General clinical data and disease activity indicators were collected. The levels of total T, B, CD4+T, CD8+T, NK, Th1, Th2, Th17, and Treg cells in peripheral blood of HCs, SLE patients (including infected and non-infected group) were analyzed by flow cytometry. The risk assessment model was constructed, and the receiver operating characteristic curve was drawn. 39 SLE patients with infection and 20 patients without infection were randomly selected to evaluate the predictive power of the regression model. RESULTS The levels of T, B, CD4+T, CD8+T, and NK cells in the infected patients were significantly decreased when compared with that of both non-infected patients and HCs (p < .05). The non-infected patients had a higher level of Th17 than that of HCs (p < . 05), but the absolute numbers of Th17 in infected patients was the lowest among the three groups (p < .001). The number of Treg cells in SLE patients was significantly lower than that of HCs (p < .01), and the infected patients had the fewest Treg cells among all these groups (p < . 05). A risk assessment model for SLE with infection was established, p = 1/(1-e-y), Y = 1.763-0.004 × Absolute number of CD4 + T cells-0.005 × Absolute number of NK cells -0.005 × Platelet count(×1012/L) + 1.033 × Absolute number of lymphocytes (×109/L) + 0.023 × C-reactive protein (mg/dL), whose predictive sensitivity is 77.5%, and specificity is 78.3%. CONCLUSION The new risk assessment model exhibits good predictive ability to assess co-infection risk in SLE patients. T cells, NK cells, and CD4 + T cells along with other parameters help in differentiating Lupus with infection from Lupus alone.
Collapse
Affiliation(s)
- Jiaqian Zhang
- Department of Rheumatology, 74761The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Shengxiao Zhang
- Department of Rheumatology, 74761The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jun Qiao
- Department of Rheumatology, 74761The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Mengting Qiu
- Department of Rheumatology, 74761The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaofeng Li
- Department of Rheumatology, 74761The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
3
|
Leal AS, Reich LA, Moerland JA, Zhang D, Liby KT. Potential therapeutic uses of rexinoids. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 91:141-183. [PMID: 34099107 DOI: 10.1016/bs.apha.2021.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The discovery of nuclear receptors, particularly retinoid X receptors (RXR), and their involvement in numerous pathways related to development sparked interest in their immunomodulatory properties. Genetic models using deletion or overexpression of RXR and the subsequent development of several small molecules that are agonists or antagonists of this receptor support a promising therapeutic role for these receptors in immunology. Bexarotene was approved in 1999 for the treatment of cutaneous T cell lymphoma. Several other small molecule RXR agonists have since been synthesized with limited preclinical development, but none have yet achieved FDA approval. Cancer treatment has recently been revolutionized with the introduction of immune checkpoint inhibitors, but their success has been restricted to a minority of patients. This review showcases the emerging immunomodulatory effects of RXR and the potential of small molecules that target this receptor as therapies for cancer and other diseases. Here we describe the essential roles that RXR and partner receptors play in T cells, dendritic cells, macrophages and epithelial cells, especially within the tumor microenvironment. Most of these effects are site and cancer type dependent but skew immune cells toward an anti-inflammatory and anti-tumor effect. This beneficial effect on immune cells supports the promise of combining rexinoids with approved checkpoint blockade therapies in order to enhance efficacy of the latter and to delay or potentially eliminate drug resistance. The data compiled in this review strongly suggest that targeting RXR nuclear receptors is a promising new avenue in immunomodulation for cancer and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- Ana S Leal
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, United States
| | - Lyndsey A Reich
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, United States
| | - Jessica A Moerland
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, United States
| | - Di Zhang
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, United States
| | - Karen T Liby
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, United States.
| |
Collapse
|
4
|
Qin L, Qiu Z, Hsieh E, Geng T, Zhao J, Zeng X, Wan L, Xie J, Ramendra R, Routy JP, Li T. Association between lymphocyte subsets and cytomegalovirus infection status among patients with systemic lupus erythematosus: A pilot study. Medicine (Baltimore) 2019; 98:e16997. [PMID: 31574798 PMCID: PMC6775378 DOI: 10.1097/md.0000000000016997] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/26/2019] [Accepted: 08/06/2019] [Indexed: 11/04/2022] Open
Abstract
This study aimed to determine the association between different lymphocyte subsets and cytomegalovirus (CMV) infection status in patients with systemic lupus erythematosus (SLE). We performed a retrospective study among SLE patients with CMV infection and collected patient socio-demographic and clinical characteristics, as well as their recorded circulating lymphocyte subsets. Univariate and multivariable logistic regression analyses examined the relationship between CMV infection status and lymphocyte subset counts. We included 125 hospitalized patients with SLE, consisting of 88 with documented CMV infection and 37 without any evidence of CMV or other infections. Among the 88 CMV-infected patients, 65 (73.8%) patients developed CMV disease and 23 (26.2%) presented as CMV viremia. Compared to uninfected patients (1520 ± 101 cells/μL), lymphocytes remained stable among those with CMV viremia (1305 ± 272 cells/μL, P = .995). However, compared to their uninfected counterparts, there was a marked decrease in lymphocytes among patients with CMV disease (680 ± 513 cells/μL, P < .001). Analysis of lymphocyte subsets via flow cytometry showed that CD4+ T cell, CD8+ T cell, and natural killer cell counts were lower among those with CMV disease compared to those with CMV viremia and those without infection. Further, multivariable analysis showed that total lymphocyte (odds ratio [OR] 0.999, 95% confidence interval [CI] 0.998-1.000, P = .007) and CD4+ T cell counts (OR 0.99, 95% CI 0.992-0.998, P = .003) were negatively associated with CMV disease. Our findings support a potential inverse relationship between lymphopenia, specifically CD4+ T-cell lymphopenia, and CMV disease among hospitalized SLE patients.
Collapse
Affiliation(s)
- Ling Qin
- Department of Infectious Diseases, Peking Union Medical College Hospital
- Center for AIDS Research
| | - Zhifeng Qiu
- Department of Infectious Diseases, Peking Union Medical College Hospital
- Center for AIDS Research
- Clinical Immunology Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Evelyn Hsieh
- Department of Infectious Diseases, Peking Union Medical College Hospital
- Section of Rheumatology, Allergy & Immunology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Taoran Geng
- Department of Infectious Diseases, Peking Union Medical College Hospital
- Center for AIDS Research
| | | | | | - Lu Wan
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jing Xie
- Department of Infectious Diseases, Peking Union Medical College Hospital
- Center for AIDS Research
- Clinical Immunology Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Rayoun Ramendra
- Department of Microbiology and Immunology, McGill University
| | - Jean Pierre Routy
- Chronic Viral Illnesses Service and Division of Hematology, McGill University Health Centre, Montreal, QC, Canada
| | - Taisheng Li
- Department of Infectious Diseases, Peking Union Medical College Hospital
- Center for AIDS Research
- Clinical Immunology Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
5
|
Bing SJ, Shemesh I, Chong WP, Horai R, Jittayasothorn Y, Silver PB, Sredni B, Caspi RR. AS101 ameliorates experimental autoimmune uveitis by regulating Th1 and Th17 responses and inducing Treg cells. J Autoimmun 2019; 100:52-61. [PMID: 30853312 DOI: 10.1016/j.jaut.2019.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/24/2019] [Accepted: 02/25/2019] [Indexed: 10/27/2022]
Abstract
AS101 is an organotellurium compound with multifaceted immunoregulatory properties that is remarkable for its lack of toxicity. We tested the therapeutic effect of AS101 in experimental autoimmune uveitis (EAU), a model for human autoimmune uveitis. Unexpectedly, treatment with AS101 elicited Treg generation in vivo in otherwise unmanipulated mice. Mice immunized for EAU with the retinal antigen IRBP and treated with AS101 developed attenuated disease, as did AS101-treated recipients of retina-specific T cells activated in vitro. In both settings, eye-infiltrating effector T cells were decreased, whereas regulatory T (Treg) cells in the spleen were increased. Mechanistic studies in vitro revealed that AS101 restricted polarization of retina-specific T cells towards Th1 or Th17 lineage by repressing activation of their respective lineage-specific transcription factors and downstream signals. Retina-specific T cells polarized in vitro towards Th1 or Th17 in the presence of AS101 had impaired ability to induce EAU in naïve recipients. Finally, AS101 promoted differentiation of retina-specific T cells to Tregs in vitro independently of TGF-β. We conclude that AS101 modulates autoimmune T cells by inhibiting acquisition and expression of effector function and by promoting Treg generation, and suggest that AS101 could be useful as a therapeutic approach for autoimmune uveitis.
Collapse
Affiliation(s)
- So Jin Bing
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Itay Shemesh
- C.A.I.R. Institute, Safdié AIDS and Immunology Research Center, Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | - Wai Po Chong
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Reiko Horai
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yingyos Jittayasothorn
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Phyllis B Silver
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Benjamin Sredni
- C.A.I.R. Institute, Safdié AIDS and Immunology Research Center, Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel.
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
6
|
Yang Q, Cai X, Huang M, Jia L, Wang S. Immunomodulatory effects of Pseudostellaria heterophylla peptide on spleen lymphocytes via a Ca2+/CaN/NFATc1/IFN-γ pathway. Food Funct 2019; 10:3466-3476. [DOI: 10.1039/c9fo00577c] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Screening and isolation of Pseudostellaria heterophylla peptide with immunomodulatory activity via a Ca2+/CaN/NFATc1/IFN-γ pathway.
Collapse
Affiliation(s)
- Qian Yang
- College of Chemistry
- Fuzhou University
- Fuzhou
- People's Republic of China
- The Key Lab of Analysis and Detection Technology for Food Safety of the MOE
| | - Xixi Cai
- The Key Lab of Analysis and Detection Technology for Food Safety of the MOE
- College of Biological Science and Technology
- Fuzhou University
- Fuzhou
- People's Republic of China
| | - Muchen Huang
- The Key Lab of Analysis and Detection Technology for Food Safety of the MOE
- College of Biological Science and Technology
- Fuzhou University
- Fuzhou
- People's Republic of China
| | - Lee Jia
- College of Chemistry
- Fuzhou University
- Fuzhou
- People's Republic of China
| | - Shaoyun Wang
- The Key Lab of Analysis and Detection Technology for Food Safety of the MOE
- College of Biological Science and Technology
- Fuzhou University
- Fuzhou
- People's Republic of China
| |
Collapse
|
7
|
Shiraishi H, Fujino M, Shirakawa N, Ishida N, Funato H, Hirata A, Abe N, Iizuka M, Jobu K, Yokota J, Miyamura M. Effect of Minerals on Intestinal IgA Production Using Deep Sea Water Drinks. Biol Pharm Bull 2018; 40:1700-1705. [PMID: 28966241 DOI: 10.1248/bpb.b17-00258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Minerals are essential for life, as they are a vital part of protein constituents, enzyme cofactors, and other components in living organisms. Deep sea water is characterized by its cleanliness and stable low temperature, and its possible health- and medical benefits are being studied. However, no study has yet evaluated the physical properties of the numerous commercially available deep sea water products, which have varying water sources and production methods. We analyzed these products' mineral content and investigated their effect on living organism, focusing on immune functions, and investigated the relation between physiological immunoactivities and mineral intake. We qualitatively analyzed the mineral compositions of the deep sea water drinks and evaluated the drinks' physical properties using principal component analysis, a type of multivariate analysis, of their mineral content. We create an iron and copper-deficient rat model and administered deep sea water drinks for 8 weeks. We then measured their fecal immunoglobulin A (IgA) to evaluate immune function. Principal component analysis suggested that physical properties of deep sea water drinks could be determined by their sources. Administration of deep sea water drinks increased fecal IgA, thus tending to stimulate immune function, but the extent of this effect varied by drink. Of the minerals contained in deep sea water, iron showed positive correlations with the fecal IgA. The principal component analysis used in this study is suitable for evaluating deep sea water containing many minerals, and our results form a useful basis for comparative evaluations of deep sea water's bioactivity.
Collapse
Affiliation(s)
- Hisashi Shiraishi
- Kochi Medical Graduate School.,Department of Pharmacy, Kochi Medical School Hospital
| | - Maho Fujino
- Department of Pharmacy, Kochi Medical School Hospital
| | | | - Nanao Ishida
- Kochi Medical Graduate School.,Department of Pharmacy, Kochi Medical School Hospital
| | - Hiroki Funato
- Department of Pharmacy, Kochi Medical School Hospital
| | - Ayumu Hirata
- Department of Pharmacy, Kochi Medical School Hospital
| | - Noriaki Abe
- Kochi Medical Graduate School.,Department of Pharmacy, Kochi Medical School Hospital
| | | | - Kohei Jobu
- Department of Pharmacy, Kochi Medical School Hospital
| | - Junko Yokota
- Department of Pharmacy, Kochi Medical School Hospital
| | - Mitsuhiko Miyamura
- Kochi Medical Graduate School.,Department of Pharmacy, Kochi Medical School Hospital
| |
Collapse
|
8
|
Jo E, Elvitigala DAS, Wan Q, Oh M, Oh C, Lee J. Identification and molecular profiling of DC-SIGN-like from big belly seahorse (Hippocampus abdominalis) inferring its potential relevancy in host immunity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 77:270-279. [PMID: 28867209 DOI: 10.1016/j.dci.2017.08.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/30/2017] [Accepted: 08/30/2017] [Indexed: 06/07/2023]
Abstract
Dendritic-cell-specific ICAM-3-grabbing non-integrin (DC-SIGN) is a C-type lectin that functions as a pattern recognition receptor by recognizing pathogen-associated molecular patterns (PAMPs). It is also involved in various events of the dendritic cell (DC) life cycle, such as DC migration, antigen capture and presentation, and T cell priming. In this study, a DC-SIGN-like gene from the big belly seahorse Hippocampus abdominalis (designated as ShDCS-like) was identified and molecularly characterized. The putative, complete ORF was found to be 1368 bp in length, encoding a protein of 462 amino acids with a molecular mass of 52.6 kDa and a theoretical isoelectric point of 8.26. The deduced amino acid sequence contains a single carbohydrate recognition domain (CRD), in which six conserved cysteine residues and two Ca2+-binding site motifs (QPN, WND) were identified. Based on pairwise sequence analysis, ShDCS-like exhibits the highest amino acid identity (94.6%) and similarity (97.4%) with DC-SIGN-like counterpart from tiger tail seahorse Hippocampus comes. Quantitative real-time PCR revealed that ShDCS-like mRNA is transcribed universally in all tissues examined, but with abundance in kidney and gill tissues. The basal mRNA expression of ShDCS-like was modulated in blood cell, kidney, gill and liver tissues in response to the stimulation of healthy fish with lipopolysaccharides (LPS), Edwardsiella tarda, or Streptococcus iniae. Moreover, recombinant ShDCS-like-CRD domain exhibited detectable agglutination activity against different bacteria. Collectively, these results suggest that ShDCS-like may potentially involve in immune function in big belly seahorses.
Collapse
Affiliation(s)
- Eunyoung Jo
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Jeju International Marine Science Research & Education Center, Korea Institute of Ocean Science & Technology (KIOST), Jeju Special Self-Governing Province, 63349, Republic of Korea
| | - Don Anushka Sandaruwan Elvitigala
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Department of Chemistry, University of Colombo, Colombo 03, 00300, Sri Lanka
| | - Qiang Wan
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - Minyoung Oh
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - Chulhong Oh
- Jeju International Marine Science Research & Education Center, Korea Institute of Ocean Science & Technology (KIOST), Jeju Special Self-Governing Province, 63349, Republic of Korea
| | - Jehee Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea.
| |
Collapse
|
9
|
Otowa T, Maher BS, Aggen SH, McClay JL, van den Oord EJ, Hettema JM. Genome-wide and gene-based association studies of anxiety disorders in European and African American samples. PLoS One 2014; 9:e112559. [PMID: 25390645 PMCID: PMC4229211 DOI: 10.1371/journal.pone.0112559] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/07/2014] [Indexed: 01/02/2023] Open
Abstract
Anxiety disorders (ADs) are common mental disorders caused by a combination of genetic and environmental factors. Since ADs are highly comorbid with each other, partially due to shared genetic basis, studying AD phenotypes in a coordinated manner may be a powerful strategy for identifying potential genetic loci for ADs. To detect these loci, we performed genome-wide association studies (GWAS) of ADs. In addition, as a complementary approach to single-locus analysis, we also conducted gene- and pathway-based analyses. GWAS data were derived from the control sample of the Molecular Genetics of Schizophrenia (MGS) project (2,540 European American and 849 African American subjects) genotyped on the Affymetrix GeneChip 6.0 array. We applied two phenotypic approaches: (1) categorical case-control comparisons (CC) based upon psychiatric diagnoses, and (2) quantitative phenotypic factor scores (FS) derived from a multivariate analysis combining information across the clinical phenotypes. Linear and logistic models were used to analyse the association with ADs using FS and CC traits, respectively. At the single locus level, no genome-wide significant association was found. A trans-population gene-based meta-analysis across both ethnic subsamples using FS identified three genes (MFAP3L on 4q32.3, NDUFAB1 and PALB2 on 16p12) with genome-wide significance (false discovery rate (FDR] <5%). At the pathway level, several terms such as transcription regulation, cytokine binding, and developmental process were significantly enriched in ADs (FDR <5%). Our approaches studying ADs as quantitative traits and utilizing the full GWAS data may be useful in identifying susceptibility genes and pathways for ADs.
Collapse
Affiliation(s)
- Takeshi Otowa
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Neuropsychiatry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Brion S. Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Steven H. Aggen
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Joseph L. McClay
- Department of Pharmacy, Center for Biomarker Research and Personalized Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Edwin J. van den Oord
- Department of Pharmacy, Center for Biomarker Research and Personalized Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - John M. Hettema
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- * E-mail:
| |
Collapse
|
10
|
Advanced glycation end products promote differentiation of CD4+ T helper cells toward pro-inflammatory response. ACTA ACUST UNITED AC 2014; 34:10-17. [DOI: 10.1007/s11596-014-1224-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 01/20/2014] [Indexed: 12/16/2022]
|
11
|
Characterization of T cell phenotype and function in a double transgenic (collagen-specific TCR/HLA-DR1) humanized model of arthritis. Arthritis Res Ther 2014; 16:R7. [PMID: 24405551 PMCID: PMC3978884 DOI: 10.1186/ar4433] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 12/27/2013] [Indexed: 12/22/2022] Open
Abstract
Introduction T cells orchestrate joint inflammation in rheumatoid arthritis (RA), yet they are difficult to study due to the small numbers of antigen-specific cells. The goal of this study was to characterize a new humanized model of autoimmune arthritis and to describe the phenotypic and functional changes that occur in autoimmune T cells following the induction of pathological events. Methods We developed a double transgenic mouse containing both the HLA-DR1 transgene and an HLA-DR1-restricted collagen-specific TCR in order to obtain large numbers of antigen-specific T cells that can be used for immunologic studies. Results In vitro, CII-specific T cells from this mouse proliferated vigorously in response to the CII immunodominant peptide A2 and the cells altered their phenotype to become predominately CD62Llow and CD44high “activated” T cells. The response was accompanied by the production of Th1, Th2, and Th17-type cytokines. Following immunization with bovine CII/CFA, these mice develop an accelerated arthritis compared to single transgenic HLA-DR1 mice. On the other hand, when the mice were treated orally with the analog peptide A12, (a suppressive analog of collagen we have previously described), arthritis was significantly suppressed, despite the fact that >90% of the CD4+ T cells express the TCR Tg. In GALT tissues taken from the A12-treated mice, IL-2, IFN-γ, and IL-17 production to the autoimmune collagen determinant dropped while high levels of IL-10 and IL-4 were produced. Conclusions We have developed a humanized model of autoimmune arthritis that will be useful for the study of T cell directed therapies as well as T cell mediated mechanisms of autoimmune diseases.
Collapse
|