1
|
Bodhale N, Nair A, Saha B. Isoform-specific functions of Ras in T-cell development and differentiation. Eur J Immunol 2023; 53:e2350430. [PMID: 37173132 DOI: 10.1002/eji.202350430] [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: 02/13/2023] [Revised: 05/02/2023] [Accepted: 05/11/2023] [Indexed: 05/15/2023]
Abstract
Ras GTPases, well characterized for their role in oncogenesis, are the cells' molecular switches that signal to maintain immune homeostasis through cellular development, proliferation, differentiation, survival, and apoptosis. In the immune system, T cells are the central players that cause autoimmunity if dysregulated. Antigen-specific T-cell receptor (TCR) stimulation activates Ras-isoforms, which exhibit isoform-specific activator and effector requirements, functional specificities, and a selective role in T-cell development and differentiation. Recent studies show the role of Ras in T-cell-mediated autoimmune diseases; however, there is a scarcity of knowledge about the role of Ras in T-cell development and differentiation. To date, limited studies have demonstrated Ras activation in response to positive and negative selection signals and Ras isoform-specific signaling, including subcellular signaling, in immune cells. The knowledge of isoform-specific functions of Ras in T cells is essential, but still inadequate to develop the T-cell-targeted Ras isoform-specific treatment strategies for the diseases caused by altered Ras-isoform expression and activation in T cells. In this review, we discuss the role of Ras in T-cell development and differentiation, critically analyzing the isoform-specific functions.
Collapse
Affiliation(s)
| | - Arathi Nair
- National Centre for Cell Science, Pune, India
| | | |
Collapse
|
2
|
Sadeghi Shaker M, Rokni M, Mahmoudi M, Farhadi E. Ras family signaling pathway in immunopathogenesis of inflammatory rheumatic diseases. Front Immunol 2023; 14:1151246. [PMID: 37256120 PMCID: PMC10225558 DOI: 10.3389/fimmu.2023.1151246] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/26/2023] [Indexed: 06/01/2023] Open
Abstract
The Ras (rat sarcoma virus) is a GTP-binding protein that is considered one of the important members of the Ras-GTPase superfamily. The Ras involves several pathways in the cell that include proliferation, migration, survival, differentiation, and fibrosis. Abnormalities in the expression level and activation of the Ras family signaling pathway and its downstream kinases such as Raf/MEK/ERK1-2 contribute to the pathogenic mechanisms of rheumatic diseases including immune system dysregulation, inflammation, and fibrosis in systemic sclerosis (SSc); destruction and inflammation of synovial tissue in rheumatoid arthritis (RA); and autoantibody production and immune complexes formation in systemic lupus erythematosus (SLE); and enhance osteoblast differentiation and ossification during skeletal formation in ankylosing spondylitis (AS). In this review, the basic biology, signaling of Ras, and abnormalities in this pathway in rheumatic diseases including SSc, RA, AS, and SLE will be discussed.
Collapse
Affiliation(s)
- Mina Sadeghi Shaker
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Rokni
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Farhadi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Expression of Non-T Cell Activation Linker (NTAL) in Jurkat Cells Negatively Regulates TCR Signaling: Potential Role in Rheumatoid Arthritis. Int J Mol Sci 2023; 24:ijms24054574. [PMID: 36902005 PMCID: PMC10003381 DOI: 10.3390/ijms24054574] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
T lymphocytes are key players in adaptive immune responses through the recognition of peptide antigens through the T Cell Receptor (TCR). After TCR engagement, a signaling cascade is activated, leading to T cell activation, proliferation, and differentiation into effector cells. Delicate control of activation signals coupled to the TCR is needed to avoid uncontrolled immune responses involving T cells. It has been previously shown that mice deficient in the expression of the adaptor NTAL (Non-T cell activation linker), a molecule structurally and evolutionarily related to the transmembrane adaptor LAT (Linker for the Activation of T cells), develop an autoimmune syndrome characterized by the presence of autoantibodies and enlarged spleens. In the present work we intended to deepen investigation into the negative regulatory functions of the NTAL adaptor in T cells and its potential relationship with autoimmune disorders. For this purpose, in this work we used Jurkat cells as a T cell model, and we lentivirally transfected them to express the NTAL adaptor in order to analyze the effect on intracellular signals associated with the TCR. In addition, we analyzed the expression of NTAL in primary CD4+ T cells from healthy donors and Rheumatoid Arthritis (RA) patients. Our results showed that NTAL expression in Jurkat cells decreased calcium fluxes and PLC-γ1 activation upon stimulation through the TCR complex. Moreover, we showed that NTAL was also expressed in activated human CD4+ T cells, and that the increase of its expression was reduced in CD4+ T cells from RA patients. Our results, together with previous reports, suggest a relevant role for the NTAL adaptor as a negative regulator of early intracellular TCR signaling, with a potential implication in RA.
Collapse
|
4
|
Yang H, Qi G, Dong H, Liu Z, Ma M, Liu P. Identification of Potential Serum Protein Biomarkers in Thymoma with Myasthenia Gravis After Docetaxel Treatment. Neurol Ther 2023; 12:559-570. [PMID: 36786935 PMCID: PMC10043105 DOI: 10.1007/s40120-023-00442-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/23/2023] [Indexed: 02/15/2023] Open
Abstract
INTRODUCTION Myasthenia gravis (MG) is a devastating acquired autoimmune disease that can seriously affect the patient's quality of life. It is also a common complication of thymoma. Previous studies have shown that docetaxel alleviates myasthenic symptoms in thymoma with MG (TMG). However, little is known about the protein expression profiles and biomarkers for efficacy after docetaxel treatment. METHODS We recruited 9 healthy controls and 30 patients with TMG for the serum proteomics study with data-independent acquisition (DIA) technology. We further recruited additional 30 patients for the key protein validation by enzyme-linked immunosorbent assay (ELISA). RESULTS We identified 43 proteins by trend analysis and analyzed the interaction between these proteins and MG pathogenic proteins from the DisGNET database and the correlation analysis with clinical data of patients with TMG. Among these, KRAS and SELP were screened out and validated. KRAS and SELP increased in patients with TMG and decreased significantly after docetaxel treatment. CONCLUSIONS Our study revealed that the serum proteins were differentially expressed after docetaxel treatment, suggesting their important role in patients with TMG, as well as the critical role of KRAS and SELP as biomarkers in evaluating the efficacy of docetaxel treatment.
Collapse
Affiliation(s)
- Hongxia Yang
- Center of Treatment of Myasthenia Gravis, People's Hospital of Shijiazhuang Affiliated to Hebei Medical University, 365 Jianhua Nan Street, Shijiazhuang, Hebei, China.,Hebei Provincial Clinical Research Center for Myasthenia Gravis, Shijiazhuang, China
| | - Guoyan Qi
- Center of Treatment of Myasthenia Gravis, People's Hospital of Shijiazhuang Affiliated to Hebei Medical University, 365 Jianhua Nan Street, Shijiazhuang, Hebei, China. .,Hebei Provincial Key Laboratory of Myasthenia Gravis, Shijiazhuang, China. .,Hebei Provincial Clinical Research Center for Myasthenia Gravis, Shijiazhuang, China.
| | - Huimin Dong
- Center of Treatment of Myasthenia Gravis, People's Hospital of Shijiazhuang Affiliated to Hebei Medical University, 365 Jianhua Nan Street, Shijiazhuang, Hebei, China
| | - Ze Liu
- Center of Treatment of Myasthenia Gravis, People's Hospital of Shijiazhuang Affiliated to Hebei Medical University, 365 Jianhua Nan Street, Shijiazhuang, Hebei, China.,Hebei Provincial Key Laboratory of Myasthenia Gravis, Shijiazhuang, China
| | - Mei Ma
- Center of Treatment of Myasthenia Gravis, People's Hospital of Shijiazhuang Affiliated to Hebei Medical University, 365 Jianhua Nan Street, Shijiazhuang, Hebei, China
| | - Peng Liu
- Center of Treatment of Myasthenia Gravis, People's Hospital of Shijiazhuang Affiliated to Hebei Medical University, 365 Jianhua Nan Street, Shijiazhuang, Hebei, China.,Hebei Provincial Key Laboratory of Myasthenia Gravis, Shijiazhuang, China
| |
Collapse
|
5
|
Yu JH, Moon EY, Kim J, Koo JH. Identification of Small GTPases That Phosphorylate IRF3 through TBK1 Activation Using an Active Mutant Library Screen. Biomol Ther (Seoul) 2023; 31:48-58. [PMID: 36579460 PMCID: PMC9810446 DOI: 10.4062/biomolther.2022.119] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 12/30/2022] Open
Abstract
Interferon regulatory factor 3 (IRF3) integrates both immunological and non-immunological inputs to control cell survival and death. Small GTPases are versatile functional switches that lie on the very upstream in signal transduction pathways, of which duration of activation is very transient. The large number of homologous proteins and the requirement for site-directed mutagenesis have hindered attempts to investigate the link between small GTPases and IRF3. Here, we constructed a constitutively active mutant expression library for small GTPase expression using Gibson assembly cloning. Small-scale screening identified multiple GTPases capable of promoting IRF3 phosphorylation. Intriguingly, 27 of 152 GTPases, including ARF1, RHEB, RHEBL1, and RAN, were found to increase IRF3 phosphorylation. Unbiased screening enabled us to investigate the sequence-activity relationship between the GTPases and IRF3. We found that the regulation of IRF3 by small GTPases was dependent on TBK1. Our work reveals the significant contribution of GTPases in IRF3 signaling and the potential role of IRF3 in GTPase function, providing a novel therapeutic approach against diseases with GTPase overexpression or active mutations, such as cancer.
Collapse
Affiliation(s)
- Jae-Hyun Yu
- Department of Pharmacology and Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea,College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun-Yi Moon
- Department of Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Jiyoon Kim
- Department of Pharmacology and Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea,Corresponding Authors E-mail: (Kim J), (Koo JH), Tel: +82-2-3147-8358 (Kim J), +82-2-880-7839 (Koo JH), Fax: +82-2-536-2485 (Kim J), +82-2-888-9122 (Koo JH)
| | - Ja Hyun Koo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea,Corresponding Authors E-mail: (Kim J), (Koo JH), Tel: +82-2-3147-8358 (Kim J), +82-2-880-7839 (Koo JH), Fax: +82-2-536-2485 (Kim J), +82-2-888-9122 (Koo JH)
| |
Collapse
|
6
|
Zhao J, Wei K, Chang C, Xu L, Jiang P, Guo S, Schrodi SJ, He D. DNA Methylation of T Lymphocytes as a Therapeutic Target: Implications for Rheumatoid Arthritis Etiology. Front Immunol 2022; 13:863703. [PMID: 35309322 PMCID: PMC8927780 DOI: 10.3389/fimmu.2022.863703] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 02/14/2022] [Indexed: 11/28/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that can cause joint damage and disability. Epigenetic variation, especially DNA methylation, has been shown to be involved in almost all the stages of the pathology of RA, from autoantibody production to various self-effector T cells and the defects of protective T cells that can lead to chronic inflammation and erosion of bones and joints. Given the critical role of T cells in the pathology of RA, the regulatory functions of DNA methylation in T cell biology remain unclear. In this review, we elaborate on the relationship between RA pathogenesis and DNA methylation in the context of different T cell populations. We summarize the relevant methylation events in T cell development, differentiation, and T cell-related genes in disease prediction and drug efficacy. Understanding the epigenetic regulation of T cells has the potential to profoundly translate preclinical results into clinical practice and provide a framework for the development of novel, individualized RA therapeutics.
Collapse
Affiliation(s)
- Jianan Zhao
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Kai Wei
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Cen Chang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Lingxia Xu
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Ping Jiang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Shicheng Guo
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Steven J Schrodi
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Dongyi He
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China.,Arthritis Institute of Integrated Traditional and Western medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| |
Collapse
|
7
|
Zuo J, Tang J, Lu M, Zhou Z, Li Y, Tian H, Liu E, Gao B, Liu T, Shao P. Glycolysis Rate-Limiting Enzymes: Novel Potential Regulators of Rheumatoid Arthritis Pathogenesis. Front Immunol 2021; 12:779787. [PMID: 34899740 PMCID: PMC8651870 DOI: 10.3389/fimmu.2021.779787] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 11/02/2021] [Indexed: 01/10/2023] Open
Abstract
Rheumatoid arthritis (RA) is a classic autoimmune disease characterized by uncontrolled synovial proliferation, pannus formation, cartilage injury, and bone destruction. The specific pathogenesis of RA, a chronic inflammatory disease, remains unclear. However, both key glycolysis rate-limiting enzymes, hexokinase-II (HK-II), phosphofructokinase-1 (PFK-1), and pyruvate kinase M2 (PKM2), as well as indirect rate-limiting enzymes, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), are thought to participate in the pathogenesis of RA. In here, we review the latest literature on the pathogenesis of RA, introduce the pathophysiological characteristics of HK-II, PFK-1/PFKFB3, and PKM2 and their expression characteristics in this autoimmune disease, and systematically assess the association between the glycolytic rate-limiting enzymes and RA from a molecular level. Moreover, we highlight HK-II, PFK-1/PFKFB3, and PKM2 as potential targets for the clinical treatment of RA. There is great potential to develop new anti-rheumatic therapies through safe inhibition or overexpression of glycolysis rate-limiting enzymes.
Collapse
Affiliation(s)
- Jianlin Zuo
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinshuo Tang
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Meng Lu
- Department of Nursing, The First Bethune Hospital of Jilin University, Changchun, China
| | - Zhongsheng Zhou
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yang Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hao Tian
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Enbo Liu
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Baoying Gao
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Te Liu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Pu Shao
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
8
|
PD-L1 signaling on human memory CD4+ T cells induces a regulatory phenotype. PLoS Biol 2021; 19:e3001199. [PMID: 33901179 PMCID: PMC8101994 DOI: 10.1371/journal.pbio.3001199] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 05/06/2021] [Accepted: 03/22/2021] [Indexed: 12/17/2022] Open
Abstract
Programmed cell death protein 1 (PD-1) is expressed on T cells upon T cell receptor (TCR) stimulation. PD-1 ligand 1 (PD-L1) is expressed in most tumor environments, and its binding to PD-1 on T cells drives them to apoptosis or into a regulatory phenotype. The fact that PD-L1 itself is also expressed on T cells upon activation has been largely neglected. Here, we demonstrate that PD-L1 ligation on human CD25-depleted CD4+ T cells, combined with CD3/TCR stimulation, induces their conversion into highly suppressive T cells. Furthermore, this effect was most prominent in memory (CD45RA−CD45RO+) T cells. PD-L1 engagement on T cells resulted in reduced ERK phosphorylation and decreased AKT/mTOR/S6 signaling. Importantly, T cells from rheumatoid arthritis patients exhibited high basal levels of phosphorylated ERK and following PD-L1 cross-linking both ERK signaling and the AKT/mTOR/S6 pathway failed to be down modulated, making them refractory to the acquisition of a regulatory phenotype. Altogether, our results suggest that PD-L1 signaling on memory T cells could play an important role in resolving inflammatory responses; maintaining a tolerogenic environment and its failure could contribute to ongoing autoimmunity. This study shows that programmed death cell receptor ligand 1 (PD-L1) signaling in memory CD4+ T cells from healthy individuals induces a regulatory phenotype; this mechanism seems to be defective in equivalent T cells from rheumatoid arthritis patients and could be in part responsible for the pathology.
Collapse
|
9
|
Zayoud M, Vax E, Elad-Sfadia G, Barshack I, Pinkas-Kramarski R, Goldstein I. Inhibition of Ras GTPases prevents Collagen-Induced Arthritis by Reducing the Generation of Pathogenic CD4 + T Cells and the Hyposialylation of Autoantibodies. ACR Open Rheumatol 2020; 2:512-524. [PMID: 32869536 PMCID: PMC7504479 DOI: 10.1002/acr2.11169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/06/2020] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE RasGTPases are master regulators of multiple intracellular signaling cascades. Perturbation of this pathway has been implicated in the pathogenesis of rheumatoid arthritis (RA). In this study we aimed to define the therapeutic potential of a novel RasGTPases inhibitor, farnesylthiosalicylate (FTS), in the preclinical mouse model of collagen-induced arthritis (CIA) and better delineate its immunomodulatory effects both ex vivo and in the mouse. METHODS We analyzed in vitro the immunomodulatory effects of FTS on various CD4+ T-cell functions such as activation, proliferation, T-helper polarization, and production of proinflammatory cytokines. Using the CIA model, we further determined the efficacy of FTS to inhibit clinical, histopathologic, and diverse immunological outcomes of arthritis. RESULTS FTS treatment of CD4+ T cells in vitro effectively targeted distinct kinases (extracellular signal-regulated kinase 1/2, p38, protein kinase B/AKT, and mammalian target of rapamycin), the production of interleukin (IL)-17A, IL-22, and granulocyte-macrophage colony-stimulating factor, and Th17 polarization. FTS therapy in the mouse CIA model significantly reduced clinical disease severity and joint inflammation/damage by histology. Importantly, FTS suppressed the in vivo induction of splenic IL-17+ IL-22+ Th17 cells and the secretion of proinflammatory cytokines. The production of pathogenic autoantibodies and their abnormal hyposialylation was significantly attenuated by FTS therapy. Importantly, in vivo generation of collagen type-II specific effector CD4+ T cells was likewise repressed by FTS therapy. CONCLUSION The RasGTPases inhibitor FTS attenuates the production of proinflammatory cytokines by in vitro-activated T cells and is a potent immunomodulatory compound in the CIA model, primarily targeting the generation of autoreactive Th17 cells and the production of autoantibodies and their subsequent pathogenic hyposialylation.
Collapse
Affiliation(s)
- Morad Zayoud
- Tel-Aviv University, Tel-Aviv, Israel.,Rambam Health Care Campus, Haifa, Israel
| | - Einav Vax
- Tel-Aviv University, Tel-Aviv, Israel.,Chaim Sheba Academic Medical Center, Ramat Gan, Israel
| | | | - Iris Barshack
- Tel-Aviv University, Tel-Aviv, Israel.,Chaim Sheba Academic Medical Center, Ramat Gan, Israel
| | | | - Itamar Goldstein
- Tel-Aviv University, Tel-Aviv, Israel.,Chaim Sheba Academic Medical Center, Ramat Gan, Israel
| |
Collapse
|
10
|
Ormseth MJ, Solus JF, Sheng Q, Ye F, Wu Q, Guo Y, Oeser AM, Allen RM, Vickers KC, Stein CM. Development and Validation of a MicroRNA Panel to Differentiate Between Patients with Rheumatoid Arthritis or Systemic Lupus Erythematosus and Controls. J Rheumatol 2019; 47:188-196. [PMID: 31092710 DOI: 10.3899/jrheum.181029] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE MicroRNA (miRNA) are short noncoding RNA that regulate genes and are both biomarkers and mediators of disease. We used small RNA (sRNA) sequencing and machine learning methodology to develop an miRNA panel to reliably differentiate between rheumatoid arthritis (RA) or systemic lupus erythematosus (SLE) and control subjects. METHODS Plasma samples from 167 RA and 91 control subjects who frequency-matched for age, race, and sex were used for sRNA sequencing. TIGER was used to analyze miRNA. DESeq2 and random forest analyses were used to identify a prioritized list of miRNA differentially expressed in patients with RA. Prioritized miRNA were validated by quantitative PCR, and lasso and logistic regression were used to select the final panel of 6 miRNA that best differentiated RA from controls. The panel was validated in a separate cohort of 12 SLE, 32 RA, and 32 control subjects. Panel efficacy was assessed by area under the receiver operative characteristic curve (AUC) analyses. RESULTS The final panel included miR-22-3p, miR-24-3p, miR-96-5p, miR-134-5p, miR-140-3p, and miR-627-5p. The panel differentiated RA from control subjects in discovery (AUC = 0.81) and validation cohorts (AUC = 0.71), seronegative RA (AUC = 0.84), RA remission (AUC = 0.85), and patients with SLE (AUC = 0.80) versus controls. Pathway analysis showed upstream regulators and targets of panel miRNA are associated with pathways implicated in RA pathogenesis. CONCLUSION An miRNA panel identified by a bioinformatic approach differentiated between RA or SLE patients and control subjects. The panel may represent an autoimmunity signature, perhaps related to inflammatory arthritis, which is not dependent on active disease or seropositivity.
Collapse
Affiliation(s)
- Michelle J Ormseth
- From the Tennessee Valley Healthcare System, US Department of Veterans Affairs; Department of Medicine, Vanderbilt University Medical Center; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Bioinformatics, University of New Mexico, Albuquerque, New Mexico, USA. .,M.J. Ormseth, MD, Tennessee Valley Healthcare System, US Department of Veterans Affairs, and Department of Medicine, Vanderbilt University Medical Center; J.F. Solus, PhD, Department of Medicine, Vanderbilt University Medical Center; Q. Sheng, PhD, Department of Biostatistics, Vanderbilt University Medical Center; F. Ye, PhD, Department of Biostatistics, Vanderbilt University Medical Center; Q. Wu, PhD, Department of Medicine, Vanderbilt University Medical Center; Y. Guo, PhD, Department of Bioinformatics, University of New Mexico; A.M. Oeser, BA, MLAS, CCRP, Department of Medicine, Vanderbilt University Medical Center; R.M. Allen, PhD, Department of Medicine, Vanderbilt University Medical Center; K.C. Vickers, PhD, Department of Medicine, Vanderbilt University Medical Center; C.M. Stein, MBChB, Department of Medicine, Vanderbilt University Medical Center.
| | - Joseph F Solus
- From the Tennessee Valley Healthcare System, US Department of Veterans Affairs; Department of Medicine, Vanderbilt University Medical Center; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Bioinformatics, University of New Mexico, Albuquerque, New Mexico, USA.,M.J. Ormseth, MD, Tennessee Valley Healthcare System, US Department of Veterans Affairs, and Department of Medicine, Vanderbilt University Medical Center; J.F. Solus, PhD, Department of Medicine, Vanderbilt University Medical Center; Q. Sheng, PhD, Department of Biostatistics, Vanderbilt University Medical Center; F. Ye, PhD, Department of Biostatistics, Vanderbilt University Medical Center; Q. Wu, PhD, Department of Medicine, Vanderbilt University Medical Center; Y. Guo, PhD, Department of Bioinformatics, University of New Mexico; A.M. Oeser, BA, MLAS, CCRP, Department of Medicine, Vanderbilt University Medical Center; R.M. Allen, PhD, Department of Medicine, Vanderbilt University Medical Center; K.C. Vickers, PhD, Department of Medicine, Vanderbilt University Medical Center; C.M. Stein, MBChB, Department of Medicine, Vanderbilt University Medical Center
| | - Quanhu Sheng
- From the Tennessee Valley Healthcare System, US Department of Veterans Affairs; Department of Medicine, Vanderbilt University Medical Center; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Bioinformatics, University of New Mexico, Albuquerque, New Mexico, USA.,M.J. Ormseth, MD, Tennessee Valley Healthcare System, US Department of Veterans Affairs, and Department of Medicine, Vanderbilt University Medical Center; J.F. Solus, PhD, Department of Medicine, Vanderbilt University Medical Center; Q. Sheng, PhD, Department of Biostatistics, Vanderbilt University Medical Center; F. Ye, PhD, Department of Biostatistics, Vanderbilt University Medical Center; Q. Wu, PhD, Department of Medicine, Vanderbilt University Medical Center; Y. Guo, PhD, Department of Bioinformatics, University of New Mexico; A.M. Oeser, BA, MLAS, CCRP, Department of Medicine, Vanderbilt University Medical Center; R.M. Allen, PhD, Department of Medicine, Vanderbilt University Medical Center; K.C. Vickers, PhD, Department of Medicine, Vanderbilt University Medical Center; C.M. Stein, MBChB, Department of Medicine, Vanderbilt University Medical Center
| | - Fei Ye
- From the Tennessee Valley Healthcare System, US Department of Veterans Affairs; Department of Medicine, Vanderbilt University Medical Center; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Bioinformatics, University of New Mexico, Albuquerque, New Mexico, USA.,M.J. Ormseth, MD, Tennessee Valley Healthcare System, US Department of Veterans Affairs, and Department of Medicine, Vanderbilt University Medical Center; J.F. Solus, PhD, Department of Medicine, Vanderbilt University Medical Center; Q. Sheng, PhD, Department of Biostatistics, Vanderbilt University Medical Center; F. Ye, PhD, Department of Biostatistics, Vanderbilt University Medical Center; Q. Wu, PhD, Department of Medicine, Vanderbilt University Medical Center; Y. Guo, PhD, Department of Bioinformatics, University of New Mexico; A.M. Oeser, BA, MLAS, CCRP, Department of Medicine, Vanderbilt University Medical Center; R.M. Allen, PhD, Department of Medicine, Vanderbilt University Medical Center; K.C. Vickers, PhD, Department of Medicine, Vanderbilt University Medical Center; C.M. Stein, MBChB, Department of Medicine, Vanderbilt University Medical Center
| | - Qiong Wu
- From the Tennessee Valley Healthcare System, US Department of Veterans Affairs; Department of Medicine, Vanderbilt University Medical Center; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Bioinformatics, University of New Mexico, Albuquerque, New Mexico, USA.,M.J. Ormseth, MD, Tennessee Valley Healthcare System, US Department of Veterans Affairs, and Department of Medicine, Vanderbilt University Medical Center; J.F. Solus, PhD, Department of Medicine, Vanderbilt University Medical Center; Q. Sheng, PhD, Department of Biostatistics, Vanderbilt University Medical Center; F. Ye, PhD, Department of Biostatistics, Vanderbilt University Medical Center; Q. Wu, PhD, Department of Medicine, Vanderbilt University Medical Center; Y. Guo, PhD, Department of Bioinformatics, University of New Mexico; A.M. Oeser, BA, MLAS, CCRP, Department of Medicine, Vanderbilt University Medical Center; R.M. Allen, PhD, Department of Medicine, Vanderbilt University Medical Center; K.C. Vickers, PhD, Department of Medicine, Vanderbilt University Medical Center; C.M. Stein, MBChB, Department of Medicine, Vanderbilt University Medical Center
| | - Yan Guo
- From the Tennessee Valley Healthcare System, US Department of Veterans Affairs; Department of Medicine, Vanderbilt University Medical Center; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Bioinformatics, University of New Mexico, Albuquerque, New Mexico, USA.,M.J. Ormseth, MD, Tennessee Valley Healthcare System, US Department of Veterans Affairs, and Department of Medicine, Vanderbilt University Medical Center; J.F. Solus, PhD, Department of Medicine, Vanderbilt University Medical Center; Q. Sheng, PhD, Department of Biostatistics, Vanderbilt University Medical Center; F. Ye, PhD, Department of Biostatistics, Vanderbilt University Medical Center; Q. Wu, PhD, Department of Medicine, Vanderbilt University Medical Center; Y. Guo, PhD, Department of Bioinformatics, University of New Mexico; A.M. Oeser, BA, MLAS, CCRP, Department of Medicine, Vanderbilt University Medical Center; R.M. Allen, PhD, Department of Medicine, Vanderbilt University Medical Center; K.C. Vickers, PhD, Department of Medicine, Vanderbilt University Medical Center; C.M. Stein, MBChB, Department of Medicine, Vanderbilt University Medical Center
| | - Annette M Oeser
- From the Tennessee Valley Healthcare System, US Department of Veterans Affairs; Department of Medicine, Vanderbilt University Medical Center; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Bioinformatics, University of New Mexico, Albuquerque, New Mexico, USA.,M.J. Ormseth, MD, Tennessee Valley Healthcare System, US Department of Veterans Affairs, and Department of Medicine, Vanderbilt University Medical Center; J.F. Solus, PhD, Department of Medicine, Vanderbilt University Medical Center; Q. Sheng, PhD, Department of Biostatistics, Vanderbilt University Medical Center; F. Ye, PhD, Department of Biostatistics, Vanderbilt University Medical Center; Q. Wu, PhD, Department of Medicine, Vanderbilt University Medical Center; Y. Guo, PhD, Department of Bioinformatics, University of New Mexico; A.M. Oeser, BA, MLAS, CCRP, Department of Medicine, Vanderbilt University Medical Center; R.M. Allen, PhD, Department of Medicine, Vanderbilt University Medical Center; K.C. Vickers, PhD, Department of Medicine, Vanderbilt University Medical Center; C.M. Stein, MBChB, Department of Medicine, Vanderbilt University Medical Center
| | - Ryan M Allen
- From the Tennessee Valley Healthcare System, US Department of Veterans Affairs; Department of Medicine, Vanderbilt University Medical Center; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Bioinformatics, University of New Mexico, Albuquerque, New Mexico, USA.,M.J. Ormseth, MD, Tennessee Valley Healthcare System, US Department of Veterans Affairs, and Department of Medicine, Vanderbilt University Medical Center; J.F. Solus, PhD, Department of Medicine, Vanderbilt University Medical Center; Q. Sheng, PhD, Department of Biostatistics, Vanderbilt University Medical Center; F. Ye, PhD, Department of Biostatistics, Vanderbilt University Medical Center; Q. Wu, PhD, Department of Medicine, Vanderbilt University Medical Center; Y. Guo, PhD, Department of Bioinformatics, University of New Mexico; A.M. Oeser, BA, MLAS, CCRP, Department of Medicine, Vanderbilt University Medical Center; R.M. Allen, PhD, Department of Medicine, Vanderbilt University Medical Center; K.C. Vickers, PhD, Department of Medicine, Vanderbilt University Medical Center; C.M. Stein, MBChB, Department of Medicine, Vanderbilt University Medical Center
| | - Kasey C Vickers
- From the Tennessee Valley Healthcare System, US Department of Veterans Affairs; Department of Medicine, Vanderbilt University Medical Center; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Bioinformatics, University of New Mexico, Albuquerque, New Mexico, USA.,M.J. Ormseth, MD, Tennessee Valley Healthcare System, US Department of Veterans Affairs, and Department of Medicine, Vanderbilt University Medical Center; J.F. Solus, PhD, Department of Medicine, Vanderbilt University Medical Center; Q. Sheng, PhD, Department of Biostatistics, Vanderbilt University Medical Center; F. Ye, PhD, Department of Biostatistics, Vanderbilt University Medical Center; Q. Wu, PhD, Department of Medicine, Vanderbilt University Medical Center; Y. Guo, PhD, Department of Bioinformatics, University of New Mexico; A.M. Oeser, BA, MLAS, CCRP, Department of Medicine, Vanderbilt University Medical Center; R.M. Allen, PhD, Department of Medicine, Vanderbilt University Medical Center; K.C. Vickers, PhD, Department of Medicine, Vanderbilt University Medical Center; C.M. Stein, MBChB, Department of Medicine, Vanderbilt University Medical Center
| | - C Michael Stein
- From the Tennessee Valley Healthcare System, US Department of Veterans Affairs; Department of Medicine, Vanderbilt University Medical Center; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Bioinformatics, University of New Mexico, Albuquerque, New Mexico, USA.,M.J. Ormseth, MD, Tennessee Valley Healthcare System, US Department of Veterans Affairs, and Department of Medicine, Vanderbilt University Medical Center; J.F. Solus, PhD, Department of Medicine, Vanderbilt University Medical Center; Q. Sheng, PhD, Department of Biostatistics, Vanderbilt University Medical Center; F. Ye, PhD, Department of Biostatistics, Vanderbilt University Medical Center; Q. Wu, PhD, Department of Medicine, Vanderbilt University Medical Center; Y. Guo, PhD, Department of Bioinformatics, University of New Mexico; A.M. Oeser, BA, MLAS, CCRP, Department of Medicine, Vanderbilt University Medical Center; R.M. Allen, PhD, Department of Medicine, Vanderbilt University Medical Center; K.C. Vickers, PhD, Department of Medicine, Vanderbilt University Medical Center; C.M. Stein, MBChB, Department of Medicine, Vanderbilt University Medical Center
| |
Collapse
|
11
|
Inhibition of glucose metabolism selectively targets autoreactive follicular helper T cells. Nat Commun 2018; 9:4369. [PMID: 30348969 PMCID: PMC6197193 DOI: 10.1038/s41467-018-06686-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 09/14/2018] [Indexed: 01/19/2023] Open
Abstract
Follicular helper T (TFH) cells are expanded in systemic lupus erythematosus, where they are required to produce high affinity autoantibodies. Eliminating TFH cells would, however compromise the production of protective antibodies against viral and bacterial pathogens. Here we show that inhibiting glucose metabolism results in a drastic reduction of the frequency and number of TFH cells in lupus-prone mice. However, this inhibition has little effect on the production of T-cell-dependent antibodies following immunization with an exogenous antigen or on the frequency of virus-specific TFH cells induced by infection with influenza. In contrast, glutaminolysis inhibition reduces both immunization-induced and autoimmune TFH cells and humoral responses. Solute transporter gene signature suggests different glucose and amino acid fluxes between autoimmune TFH cells and exogenous antigen-specific TFH cells. Thus, blocking glucose metabolism may provide an effective therapeutic approach to treat systemic autoimmunity by eliminating autoreactive TFH cells while preserving protective immunity against pathogens. T cell functions depend on distinct metabolic fluxes. Here the authors show different metabolic requirements of humoral responses to self versus microbial antigens: while glucose is dispensable for antiviral Tfh and antibody responses, it is essential to mount these responses against autoantigens.
Collapse
|
12
|
Small GTPase RAS in multiple sclerosis - exploring the role of RAS GTPase in the etiology of multiple sclerosis. Small GTPases 2018; 11:312-319. [PMID: 30043672 DOI: 10.1080/21541248.2018.1502591] [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: 10/28/2022] Open
Abstract
RAS signaling is involved in the development of autoimmunity in general. Multiple sclerosis (MS) is a T cell-mediated autoimmune disease of the central nervous system. It is widely recognized that a reduction of Foxp3+ regulatory T (Treg) cells is an immunological hallmark of MS, but the underlying mechanisms are unclear. In experimental autoimmune models, N-Ras and K-Ras inhibition triggers an anti-inflammatory effect up-regulating, via foxp3 elevation, the numbers and the functional suppressive properties of Tregs. Similarly, an increase in natural Tregs number during Experimental Autoimmune Encephalomyelitis (EAE) in R-RAS -/- mice results in attenuated disease. In humans, only KRAS GTPase isoform is involved in mechanism causing tolerance defects in rheumatoid arthritis (RA). T cells from these patients have increased transcription of KRAS (but not NRAS). RAS genes are major drivers in human cancers. Consequently, there has been considerable interest in developing anti-RAS inhibitors for cancer treatment. Despite efforts, no anti-RAS therapy has succeeded in the clinic. The major strategy that has so far reached the clinic aimed to inhibit activated Ras indirectly through blocking its post-translational modification and inducing its mis-localization. The disappointing clinical outcome of Farnesyl Transferase Inhibitors (FTIs) in cancers has decreased interest in these drugs. However, FTIs suppress EAE by downregulation of myelin-reactive activated T-lymphocytes and statins are currently studied in clinical trials for MS. However, no pharmacologic approaches to targeting Ras proteins directly have yet succeeded. The therapeutic strategy to recover immune function through the restoration of impaired Tregs function with the mounting evidences regarding KRAS in autoimmune mediated disorder (MS, SLE, RA, T1D) suggest as working hypothesis the direct targeting KRAS activation using cancer-derived small molecules may be clinically relevant. ABBREVIATIONS FTIs: Farnesyl Transferase Inhibitors; MS: Multiple Sclerosis; RRMS: Relapsing Remitting Multiple Sclerosis; PPMS: Primary Progressive Multiple Sclerosis; Tregs: regulatory T-cells; Foxp3: Forkhead box P3; EAE: Experimental Autoimmune Encephalomyelitis; T1D: Type 1 Diabete; SLE: Systemic Lupus Erythematosus; RA: Rheumatoid Arthritis; CNS: Central Nervous System; TMEV: Theiler's murine encephalomyelitis virus; FTS: farnesyl thiosalicylic acid; TCR: T-Cell Receptor; AIA: Adjuvant-induced Arthritis; EAN: experimental autoimmune neuritis; HVR: hypervariable region; HMG-CoA: 3-hydroxy-3-methylglutaryl coenzyme A reductase; PBMC: Peripheral Blood Mononuclear Cells.
Collapse
|
13
|
Inhibition of KRAS-dependent lung cancer cell growth by deltarasin: blockage of autophagy increases its cytotoxicity. Cell Death Dis 2018; 9:216. [PMID: 29440631 PMCID: PMC5833846 DOI: 10.1038/s41419-017-0065-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 10/09/2017] [Accepted: 10/12/2017] [Indexed: 12/18/2022]
Abstract
Deltarasin is a recently identified small molecule that can inhibit KRAS–PDEδ interactions by binding to a hydrophobic pocket on PDEδ, resulting in the impairment of cell growth, KRAS activity, and RAS/RAF signaling in human pancreatic ductal adenocarcinoma cell lines. Since KRAS mutations are the most common oncogene mutations in lung adenocarcinomas, implicated in over 30% of all lung cancer cases, we examined the ability of deltarasin to inhibit KRAS-dependent lung cancer cell growth. Here, for the first time, we document that deltarasin produces both apoptosis and autophagy in KRAS-dependent lung cancer cells in vitro and inhibits lung tumor growth in vivo. Deltarasin induces apoptosis by inhibiting the interaction of with PDEδ and its downstream signaling pathways, while it induces autophagy through the AMPK-mTOR signaling pathway. Importantly, the autophagy inhibitor, 3-methyl adenine (3-MA) markedly enhances deltarasin-induced apoptosis via elevation of reactive oxygen species (ROS). In contrast, inhibition of ROS by N-acetylcysteine (NAC) significantly attenuated deltarasin-induced cell death. Collectively, these observations suggest that the anti-cancer cell activity of deltarasin can be enhanced by simultaneously blocking “tumor protective” autophagy, but inhibited if combined with an anti-oxidant.
Collapse
|
14
|
Zayoud M, Marcu-Malina V, Vax E, Jacob-Hirsch J, Elad-Sfadia G, Barshack I, Kloog Y, Goldstein I. Ras Signaling Inhibitors Attenuate Disease in Adjuvant-Induced Arthritis via Targeting Pathogenic Antigen-Specific Th17-Type Cells. Front Immunol 2017; 8:799. [PMID: 28736556 PMCID: PMC5500629 DOI: 10.3389/fimmu.2017.00799] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/23/2017] [Indexed: 11/29/2022] Open
Abstract
The Ras family of GTPases plays an important role in signaling nodes downstream to T cell receptor and CD28 activation, potentially lowering the threshold for T-cell receptor activation by autoantigens. Somatic mutation in NRAS or KRAS may cause a rare autoimmune disorder coupled with abnormal expansion of lymphocytes. T cells from rheumatoid arthritis (RA) patients show excessive activation of Ras/MEK/ERK pathway. The small molecule farnesylthiosalicylic acid (FTS) interferes with the interaction between Ras GTPases and their prenyl-binding chaperones to inhibit proper plasma membrane localization. In the present study, we tested the therapeutic and immunomodulatory effects of FTS and its derivative 5-fluoro-FTS (F-FTS) in the rat adjuvant-induced arthritis model (AIA). We show that AIA severity was significantly reduced by oral FTS and F-FTS treatment compared to vehicle control treatment. FTS was as effective as the mainstay anti-rheumatic drug methotrexate, and combining the two drugs significantly increased efficacy compared to each drug alone. We also discovered that FTS therapy inhibited both the CFA-driven in vivo induction of Th17 and IL-17/IFN-γ producing “double positive” as well as the upregulation of serum levels of the Th17-associated cytokines IL-17A and IL-22. By gene microarray analysis of effector CD4+ T cells from CFA-immunized rats, re-stimulated in vitro with the mycobacterium tuberculosis heat-shock protein 65 (Bhsp65), we determined that FTS abrogated the Bhsp65-induced transcription of a large list of genes (e.g., Il17a/f, Il22, Ifng, Csf2, Lta, and Il1a). The functional enrichment bioinformatics analysis showed significant overlap with predefined gene sets related to inflammation, immune system processes and autoimmunity. In conclusion, FTS and F-FTS display broad immunomodulatory effects in AIA with inhibition of the Th17-type response to a dominant arthritogenic antigen. Hence, targeting Ras signal-transduction cascade is a potential novel therapeutic approach for RA.
Collapse
Affiliation(s)
- Morad Zayoud
- Sheba Cancer Research Center, Chaim Sheba Academic Medical Center, Ramat Gan, Israel.,Rheumatology Unit, Chaim Sheba Academic Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Victoria Marcu-Malina
- Sheba Cancer Research Center, Chaim Sheba Academic Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Einav Vax
- Sheba Cancer Research Center, Chaim Sheba Academic Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Jasmine Jacob-Hirsch
- Sheba Cancer Research Center, Chaim Sheba Academic Medical Center, Ramat Gan, Israel
| | - Galit Elad-Sfadia
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences & Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - Iris Barshack
- Institute of Pathology, Chaim Sheba Academic Medical Center, Ramat Gan, Israel
| | - Yoel Kloog
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences & Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - Itamar Goldstein
- Sheba Cancer Research Center, Chaim Sheba Academic Medical Center, Ramat Gan, Israel.,Rheumatology Unit, Chaim Sheba Academic Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
15
|
Mitchell CJ, Getnet D, Kim MS, Manda SS, Kumar P, Huang TC, Pinto SM, Nirujogi RS, Iwasaki M, Shaw PG, Wu X, Zhong J, Chaerkady R, Marimuthu A, Muthusamy B, Sahasrabuddhe NA, Raju R, Bowman C, Danilova L, Cutler J, Kelkar DS, Drake CG, Prasad TSK, Marchionni L, Murakami PN, Scott AF, Shi L, Thierry-Mieg J, Thierry-Mieg D, Irizarry R, Cope L, Ishihama Y, Wang C, Gowda H, Pandey A. A multi-omic analysis of human naïve CD4+ T cells. BMC SYSTEMS BIOLOGY 2015; 9:75. [PMID: 26542228 PMCID: PMC4636073 DOI: 10.1186/s12918-015-0225-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 10/28/2015] [Indexed: 12/21/2022]
Abstract
Background Cellular function and diversity are orchestrated by complex interactions of fundamental biomolecules including DNA, RNA and proteins. Technological advances in genomics, epigenomics, transcriptomics and proteomics have enabled massively parallel and unbiased measurements. Such high-throughput technologies have been extensively used to carry out broad, unbiased studies, particularly in the context of human diseases. Nevertheless, a unified analysis of the genome, epigenome, transcriptome and proteome of a single human cell type to obtain a coherent view of the complex interplay between various biomolecules has not yet been undertaken. Here, we report the first multi-omic analysis of human primary naïve CD4+ T cells isolated from a single individual. Results Integrating multi-omics datasets allowed us to investigate genome-wide methylation and its effect on mRNA/protein expression patterns, extent of RNA editing under normal physiological conditions and allele specific expression in naïve CD4+ T cells. In addition, we carried out a multi-omic comparative analysis of naïve with primary resting memory CD4+ T cells to identify molecular changes underlying T cell differentiation. This analysis provided mechanistic insights into how several molecules involved in T cell receptor signaling are regulated at the DNA, RNA and protein levels. Phosphoproteomics revealed downstream signaling events that regulate these two cellular states. Availability of multi-omics data from an identical genetic background also allowed us to employ novel proteogenomics approaches to identify individual-specific variants and putative novel protein coding regions in the human genome. Conclusions We utilized multiple high-throughput technologies to derive a comprehensive profile of two primary human cell types, naïve CD4+ T cells and memory CD4+ T cells, from a single donor. Through vertical as well as horizontal integration of whole genome sequencing, methylation arrays, RNA-Seq, miRNA-Seq, proteomics, and phosphoproteomics, we derived an integrated and comparative map of these two closely related immune cells and identified potential molecular effectors of immune cell differentiation following antigen encounter. Electronic supplementary material The online version of this article (doi:10.1186/s12918-015-0225-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christopher J Mitchell
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Derese Getnet
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Min-Sik Kim
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Srikanth S Manda
- Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, India.
| | - Praveen Kumar
- Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, India.
| | - Tai-Chung Huang
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Sneha M Pinto
- Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, India.
| | - Raja Sekhar Nirujogi
- Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, India.
| | - Mio Iwasaki
- Department of Molecular & Cellular BioAnalysis, Kyoto University, Kyoto, Japan.
| | - Patrick G Shaw
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Xinyan Wu
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Jun Zhong
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Raghothama Chaerkady
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Arivusudar Marimuthu
- Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, India.
| | | | | | - Rajesh Raju
- Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, India.
| | - Caitlyn Bowman
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Ludmila Danilova
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Jevon Cutler
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Dhanashree S Kelkar
- Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, India.
| | - Charles G Drake
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - T S Keshava Prasad
- Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, India.
| | - Luigi Marchionni
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Peter N Murakami
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| | - Alan F Scott
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Leming Shi
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, USA.
| | - Jean Thierry-Mieg
- National Center for Biotechnology Information, National Institutes of Health, Bethesda, MD, USA.
| | - Danielle Thierry-Mieg
- National Center for Biotechnology Information, National Institutes of Health, Bethesda, MD, USA.
| | - Rafael Irizarry
- Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, Boston, MA, USA.
| | - Leslie Cope
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Yasushi Ishihama
- Department of Molecular & Cellular BioAnalysis, Kyoto University, Kyoto, Japan.
| | - Charles Wang
- Center for Genomics and Division of Microbiology & Molecular Genetics, Loma Linda University, Loma Linda, CA, USA.
| | - Harsha Gowda
- Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, India.
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, India. .,Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Department of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
16
|
Hawtree S, Muthana M, Wilkinson JM, Akil M, Wilson AG. Histone deacetylase 1 regulates tissue destruction in rheumatoid arthritis. Hum Mol Genet 2015; 24:5367-77. [DOI: 10.1093/hmg/ddv258] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 07/03/2015] [Indexed: 01/03/2023] Open
|
17
|
Callahan MK, Masters G, Pratilas CA, Ariyan C, Katz J, Kitano S, Russell V, Gordon RA, Vyas S, Yuan J, Gupta A, Wigginton JM, Rosen N, Merghoub T, Jure-Kunkel M, Wolchok JD. Paradoxical activation of T cells via augmented ERK signaling mediated by a RAF inhibitor. Cancer Immunol Res 2014; 2:70-9. [PMID: 24416731 DOI: 10.1158/2326-6066.cir-13-0160] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
RAF inhibitors selectively block ERK signaling in BRAF-mutant melanomas and have defined a genotype-guided approach to care for this disease. RAF inhibitors have the opposite effect in BRAF wild-type tumor cells, where they cause hyperactivation of ERK signaling. Here, we predict that RAF inhibitors can enhance T cell activation, based upon the observation that these agents paradoxically activate ERK signaling in BRAF wild-type cells. To test this hypothesis, we have evaluated the effects of the RAF inhibitor BMS908662 on T cell activation and signaling in vitro and in vivo. We observe that T cell activation is enhanced in a concentration-dependent manner and that this effect corresponds with increased ERK signaling, consistent with paradoxical activation of the pathway. Furthermore, we find that the combination of BMS908662 with CTLA-4 blockade in vivo potentiates T cell expansion, corresponding with hyperactivation of ERK signaling in T cells detectable ex vivo. Lastly, this combination demonstrates superior anti-tumor activity, compared to either agent alone, in two transplantable tumor models. This study provides clear evidence that RAF inhibitors can modulate T cell function by potentiating T cell activation in vitro and in vivo. Paradoxical activation of ERK signaling in T cells offers one mechanism to explain the enhanced antitumor activity seen when RAF inhibitors are combined with CTLA-4 blockade in preclinical models.
Collapse
Affiliation(s)
- Margaret K Callahan
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065.,Weill-Cornell Medical College, New York, NY 10065
| | | | - Christine A Pratilas
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY 10065.,Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065.,Weill-Cornell Medical College, New York, NY 10065
| | - Charlotte Ariyan
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10065.,Weill-Cornell Medical College, New York, NY 10065
| | - Jessica Katz
- Bristol-Myers Squibb Company, Princeton, NJ 08543
| | - Shigehisa Kitano
- Ludwig Center for Cancer Immunotherapy at Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Valerie Russell
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Ruth Ann Gordon
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Shachi Vyas
- Ludwig Center for Cancer Immunotherapy at Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Jianda Yuan
- Ludwig Center for Cancer Immunotherapy at Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Ashok Gupta
- Bristol-Myers Squibb Company, Princeton, NJ 08543
| | | | - Neal Rosen
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065.,Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065.,Weill-Cornell Medical College, New York, NY 10065
| | - Taha Merghoub
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | | | - Jedd D Wolchok
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065.,Ludwig Institute for Cancer Research, New York Branch, New York, NY 10065.,Ludwig Center for Cancer Immunotherapy at Memorial Sloan-Kettering Cancer Center, New York, NY 10065.,Weill-Cornell Medical College, New York, NY 10065
| |
Collapse
|
18
|
Lleo A, Zhang W, McDonald WH, Seeley EH, Leung PS, Coppel RL, Ansari AA, Adams DH, Afford S, Invernizzi P, Gershwin ME. Shotgun proteomics: identification of unique protein profiles of apoptotic bodies from biliary epithelial cells. Hepatology 2014; 60:1314-23. [PMID: 24841946 PMCID: PMC4175017 DOI: 10.1002/hep.27230] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 05/16/2014] [Indexed: 01/06/2023]
Abstract
UNLABELLED Shotgun proteomics is a powerful analytic method to characterize complex protein mixtures in combination with multidimensional liquid chromatography-tandem mass spectrometry (LC-MS/MS). We used this platform for proteomic characterization of apoptotic bodies in an effort to define the complex protein mixtures found in primary cultures of human intrahepatic biliary epithelial cells (HiBEC), human renal proximal tubular epithelial cells, human bronchial epithelial cells, isolated intrahepatic biliary epithelial cells from explanted primary biliary cirrhosis (PBC), and control liver using a total of 24 individual samples. Further, as additional controls and for purposes of comparison, proteomic signatures were also obtained from intact cells and apoptotic bodies. The data obtained from LC-MS/MS, combined with database searches and protein assembly algorithms, allowed us to address significant differences in protein spectral counts and identify unique pathways that may be a component of the induction of the signature inflammatory cytokine response against BECs, including the Notch signaling pathway, interleukin (IL)8, IL6, CXCR2, and integrin signaling. Indeed, there are 11 proteins that localize specifically to apoptotic bodies of HiBEC and eight proteins that were specifically absent in HiBEC apoptotic bodies. CONCLUSION Proteomic analysis of BECs from PBC liver compared to normal liver are significantly different, suggesting that an immunological attack affects the repertoire of proteins expressed and that such cells should be thought of as living in an environment undergoing continuous selection secondary to an innate and adaptive immune response, reflecting an almost "Darwinian" bias.
Collapse
Affiliation(s)
- Ana Lleo
- Liver Unit and Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano (MI), Italy
| | - Weici Zhang
- Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, CA, USA
| | - W. Hayes McDonald
- Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Erin H. Seeley
- Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Patrick S.C. Leung
- Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, CA, USA
| | - Ross L. Coppel
- Department of Medical Microbiology, Monash University, Melbourne, Australia
| | - Aftab A. Ansari
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - David H. Adams
- Centre for Liver Research and NIHR Biomedical Research Unit in Liver Disease, Institute of Biomedical Research, University of Birmingham, Birmingham, UK
| | - Simon Afford
- Centre for Liver Research and NIHR Biomedical Research Unit in Liver Disease, Institute of Biomedical Research, University of Birmingham, Birmingham, UK
| | - Pietro Invernizzi
- Liver Unit and Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano (MI), Italy,Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, CA, USA
| | - M. Eric Gershwin
- Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, CA, USA
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW With progressive age, the immune system and the propensity for abnormal immunity change fundamentally. Individuals greater than 50 years of age are not only more susceptible to infection and cancer, but also at higher risk for chronic inflammation and immune-mediated tissue damage. The process of immunosenescence is accelerated in rheumatoid arthritis (RA). RECENT FINDINGS Premature T-cell senescence occurs not only in RA, but also has been involved in morbidity and mortality of chronic HIV infection. Senescent cells acquire the 'senescence-associated secretory phenotype', which promotes and sustains tissue inflammation. Molecular mechanisms underlying T-cell aging are beginning to be understood. In addition to the contraction of T-cell diversity because of uneven clonal expansion, senescent T cells have defects in balancing cytoplasmic kinase and phosphatase activities, changing their activation thresholds. Also, leakiness in repairing DNA lesions and uncapped telomeres imposes genomic stress. Age-induced changes in the tissue microenvironment may alter the T-cell responses. SUMMARY Gain-of-function and loss-of-function in senescent T cells undermine protective immunity and create the conditions for chronic tissue inflammation, a combination typically encountered in RA. Genetic programs involved in T-cell signaling and DNA repair are of high interest in the search for underlying molecular defects.
Collapse
|
20
|
Hohensinner PJ, Goronzy JJ, Weyand CM. Targets of immune regeneration in rheumatoid arthritis. Mayo Clin Proc 2014; 89:563-75. [PMID: 24684878 PMCID: PMC4605139 DOI: 10.1016/j.mayocp.2014.01.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/27/2014] [Accepted: 01/29/2014] [Indexed: 10/25/2022]
Abstract
Many of the aging-related morbidities, including cancer, cardiovascular disease, neurodegenerative disease, and infectious susceptibility, are linked to a decline in immune competence with a concomitant rise in proinflammatory immunity, placing the process of immune aging at the center of aging biology. Immune aging affects individuals older than 50 years and is accelerated in patients with the autoimmune disease rheumatoid arthritis. Immune aging results in a marked decline in protective immune responses and a parallel increase in tissue inflammatory responses. By studying immune cells in patients with rheumatoid arthritis, several of the molecular underpinnings of the immune aging process have been delineated, such as the loss of telomeres and inefficiencies in the repair of damaged DNA. Aging T cells display a series of abnormalities, including the unopposed up-regulation of cytoplasmic phosphatases and the loss of glycolytic competence, that alter their response to stimulating signals and undermine their longevity. Understanding the connection between accelerated immune aging and autoimmunity remains an area of active research. With increasing knowledge of the molecular pathways that cause immunosenescence, therapeutic interventions can be designed to slow or halt the seemingly inevitable deterioration of protective immunity with aging.
Collapse
Affiliation(s)
- Philipp J Hohensinner
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA
| | - Jörg J Goronzy
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA
| | - Cornelia M Weyand
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA.
| |
Collapse
|
21
|
Moon BS, Jeong WJ, Park J, Kim TI, Min DS, Choi KY. Role of oncogenic K-Ras in cancer stem cell activation by aberrant Wnt/β-catenin signaling. J Natl Cancer Inst 2014; 106:djt373. [PMID: 24491301 DOI: 10.1093/jnci/djt373] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Adenomatous polyposis coli (APC) loss-of-function mutations and K-Ras gain-of-function mutations are common abnormalities that occur during the initiation and intermediate adenoma stages of colorectal tumorigenesis, respectively. However, little is known about the role these mutations play in cancer stem cells (CSCs) associated with colorectal cancer (CRC) tumorigenesis. METHODS We analyzed tissue from CRC patients (n = 49) to determine whether K-Ras mutations contributed to CSC activation during colorectal tumorigenesis. DLD-1-K-Ras-WT and DLD-1-K-Ras-MT cells were cultured and evaluated for their ability to differentiate, form spheroids in vitro, and form tumors in vivo. Interaction between APC and K-Ras mutations in colorectal tumorigenesis was evaluated using APC (Min/+)/K-Ras (LA2) mice and DLD-1-K-Ras-WT and DLD-1-K-Ras-MT cell xenografts. (n = 4) Group differences were determined by Student t test. All statistical tests were two-sided. RESULTS The sphere-forming capability of DLD-1-K-Ras-MT cells was statistically significantly higher than that of DLD-1-K-Ras-WT cells (DLD-1-K-Ras-MT mean = 86.661 pixel, 95% confidence interval [CI] = 81.701 to 91.621 pixel; DLD-1-K-Ras-WT mean = 42.367 pixel, 95% CI = 36.467 to 48.267 pixel; P = .003). Moreover, both the size and weight of tumors from DLD-1-K-Ras-MT xenografts were markedly increased compared with tumors from DLD-1-K-Ras-WT cells. Expression of the CSC markers CD44, CD133, and CD166 was induced in intestinal tumors from APC (Min/+)/K-Ras (LA2)mice, but not K-Ras (LA2) mice, indicating that APC mutation is required for CSC activation by oncogenic K-Ras mutation. CONCLUSIONS K-Ras mutation activates CSCs, contributing to colorectal tumorigenesis and metastasis in CRC cells harboring APC mutations. Initial activation of β-catenin by APC loss and further enhancement through K-Ras mutation induces CD44, CD133, and CD166 expression.
Collapse
Affiliation(s)
- Byoung-San Moon
- Affiliations of authors: Department of Biotechnology (B-SM, W-JJ, JP, K-YC), Translational Research Center for Protein Function Control (B-SM, W-JJ, JP, DSM, K-YC), and Department of Internal Medicine and Institute of Gastroenterology, College of Medicine (TIK), Yonsei University, Seoul, Korea; Department of Molecular Biology, College of Natural Science, Pusan National University, Pusan, Korea (DSM)
| | | | | | | | | | | |
Collapse
|
22
|
Yang Z, Fujii H, Mohan SV, Goronzy JJ, Weyand CM. Phosphofructokinase deficiency impairs ATP generation, autophagy, and redox balance in rheumatoid arthritis T cells. ACTA ACUST UNITED AC 2013; 210:2119-34. [PMID: 24043759 PMCID: PMC3782046 DOI: 10.1084/jem.20130252] [Citation(s) in RCA: 248] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
In the HLA class II-associated autoimmune syndrome rheumatoid arthritis (RA), CD4 T cells are critical drivers of pathogenic immunity. We have explored the metabolic activity of RA T cells and its impact on cellular function and fate. Naive CD4 T cells from RA patients failed to metabolize equal amounts of glucose as age-matched control cells, generated less intracellular ATP, and were apoptosis-susceptible. The defect was attributed to insufficient induction of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), a regulatory and rate-limiting glycolytic enzyme known to cause the Warburg effect. Forced overexpression of PFKFB3 in RA T cells restored glycolytic flux and protected cells from excessive apoptosis. Hypoglycolytic RA T cells diverted glucose toward the pentose phosphate pathway, generated more NADPH, and consumed intracellular reactive oxygen species (ROS). PFKFB3 deficiency also constrained the ability of RA T cells to resort to autophagy as an alternative means to provide energy and biosynthetic precursor molecules. PFKFB3 silencing and overexpression identified a novel extraglycolytic role of the enzyme in autophagy regulation. In essence, T cells in RA patients, even those in a naive state, are metabolically reprogrammed with insufficient up-regulation of the glycolytic activator PFKFB3, rendering them energy-deprived, ROS- and autophagy-deficient, apoptosis-sensitive, and prone to undergo senescence.
Collapse
Affiliation(s)
- Zhen Yang
- Division of Immunology & Rheumatology, Stanford University School of Medicine, Stanford, CA 94305
| | | | | | | | | |
Collapse
|
23
|
Miao CG, Yang YY, He X, Li XF, Huang C, Huang Y, Zhang L, Lv XW, Jin Y, Li J. Wnt signaling pathway in rheumatoid arthritis, with special emphasis on the different roles in synovial inflammation and bone remodeling. Cell Signal 2013; 25:2069-78. [PMID: 23602936 DOI: 10.1016/j.cellsig.2013.04.002] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 03/30/2013] [Accepted: 04/02/2013] [Indexed: 12/17/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic symmetrical autoimmune disease of unknown etiology that affects primarily the diarthrodial joints. Characteristic features of RA pathogenesis are synovial inflammation and proliferation accompanied by cartilage erosion and bone loss. Fibroblast-like synoviocytes (FLS) display an important role in the pathogenesis of RA. Several lines of evidence show that the Wnt signaling pathway significantly participates in the RA pathogenesis. The Wnt proteins are glycoproteins that bind to the Fz receptors on the cell surface, which leads to several important biological functions, such as cell differentiation, embryonic development, limb development and joint formation. Accumulated evidence has suggested that this signaling pathway plays a key role in the FLS activation, bone resorption and joint destruction during RA development. Greater knowledge of the role of the Wnt signaling pathway in RA could improve understanding of the RA pathogenesis and the differences in RA clinical presentation and prognosis. In this review, new advances of the Wnt signaling pathway in RA pathogenesis are discussed, with special emphasis on its different roles in synovial inflammation and bone remodeling. Further studies are needed to reveal the important role of the members of the Wnt signaling pathway in the RA pathogenesis and treatment.
Collapse
Affiliation(s)
- Cheng-gui Miao
- School of Pharmacy, Institute for Liver Diseases of Anhui Medical University, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Deswal S, Meyer A, Fiala GJ, Eisenhardt AE, Schmitt LC, Salek M, Brummer T, Acuto O, Schamel WWA. Kidins220/ARMS Associates with B-Raf and the TCR, Promoting Sustained Erk Signaling in T Cells. THE JOURNAL OF IMMUNOLOGY 2013; 190:1927-35. [DOI: 10.4049/jimmunol.1200653] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
25
|
Deshpande P, Cavanagh MM, Le Saux S, Singh K, Weyand CM, Goronzy JJ. IL-7- and IL-15-mediated TCR sensitization enables T cell responses to self-antigens. THE JOURNAL OF IMMUNOLOGY 2013; 190:1416-23. [PMID: 23325887 DOI: 10.4049/jimmunol.1201620] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Regulation of the ERK pathway is intimately involved in determining whether TCR stimulation is productive or induces anergy. T cells from patients with rheumatoid arthritis (RA) have increased ERK responsiveness, which may be relevant for disease pathogenesis. Inflammatory cytokines such as TNF-α did not reproduce the TCR hypersensitivity typical for RA in T cells from healthy individuals. In contrast, priming with the homeostatic cytokines (HCs) IL-7 and IL-15 amplified ERK phosphorylation to TCR stimulation 2- to 3-fold. The underlying mechanism involved a priming of the SOS-dependent amplification loop of RAS activation. The sensitization of the TCR signaling pathway has downstream consequences, such as increased proliferation and preferential Th1 differentiation. Importantly, priming with IL-7 or IL-15 enabled T cell responses to autoantigens associated with RA. Production of HCs is induced in lymphopenic conditions, which have been shown to predispose for autoimmunity and which appear to be present in the preclinical stages of RA. We propose that HCs, possibly induced by lymphopenia, decrease the signaling threshold for TCR activation and are thereby partly responsible for autoimmunity in RA.
Collapse
Affiliation(s)
- Pratima Deshpande
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|