1
|
Andrzejczak A, Partyka A, Wiśniewski A, Porębska I, Pawełczyk K, Ptaszkowski K, Kuśnierczyk P, Jasek M, Karabon L. The association of BTLA gene polymorphisms with non-small lung cancer risk in smokers and never-smokers. Front Immunol 2023; 13:1006639. [PMID: 36741370 PMCID: PMC9893504 DOI: 10.3389/fimmu.2022.1006639] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023] Open
Abstract
Introduction Lung cancer is the predominant cause of death among cancer patients and non-small cell lung cancer (NSCLC) is the most common type. Cigarette smoking is the prevailing risk factor for NSCLC, nevertheless, this cancer is also diagnosed in never-smokers. B and T lymphocyte attenuator (BTLA) belongs to immunological checkpoints which are key regulatory molecules of the immune response. A growing body of evidence highlights the important role of BTLA in cancer. In our previous studies, we showed a significant association between BTLA gene variants and susceptibility to chronic lymphoblastic leukemia and renal cell carcinoma in the Polish population. The present study aimed to analyze the impact of BTLA polymorphic variants on the susceptibility to NSCLC and NSCLC patients' overall survival (OS). Methods Using TaqMan probes we genotyped seven BTLA single-nucleotide polymorphisms (SNPs): rs2705511, rs1982809, rs9288952, rs9288953, rs1844089, rs11921669 and rs2633582 with the use of ViiA 7 Real-Time PCR System. Results We found that rs1982809 within BTLA is associated with NSCLC risk, where carriers of rs1982809G allele (AG+GG genotypes) were more frequent in patients compared to controls. In subgroup analyses, we also noticed that rs1982809G carriers are significantly overrepresented in never-smokers, but not in smokers compared to controls. Additionally, the global distribution of the haplotypes differed between the never-smokers and smokers, where haplotypes A G G C A, C G A C G, and C G A T G were more frequent in never-smoking patients. Furthermore, the presence rs1982809G (AG+GG genotypes) allele as well as the presence of rs9288953T allele (CT+TT genotypes) increased NSCLC risk in females' patients. After stratification by histological type, we noticed that rs1982809G and rs2705511C carriers were more frequent among adenocarcinoma patients. Moreover, rs1982809G and rs2705511C correlated with the more advanced stages of NSCLC (stage II and III), but not with stage IV. Furthermore, we showed that rs2705511 and rs1982809 significantly modified OS, while rs9288952 tend to be associated with patients' survival. Conclusion Our results indicate that BTLA polymorphic variants may be considered low penetrating risk factors for NSCLC especially in never-smokers, and in females, and are associated with OS of NSCLC patients.
Collapse
Affiliation(s)
- Anna Andrzejczak
- Laboratory of Genetics and Epigenetics of Human Diseases, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland,*Correspondence: Anna Andrzejczak, ; Lidia Karabon,
| | - Anna Partyka
- Laboratory of Immunopathology, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Andrzej Wiśniewski
- Laboratory of Immunogenetics and Tissue Immunology, Department of Clinical Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Irena Porębska
- Department of Pulmonology and Lung Oncology, Wrocław Medical University, Wrocław, Poland
| | - Konrad Pawełczyk
- Departament of Thoracic Surgery, Lower Silesian Centre of Oncology, Pulmonology and Haematology, Wrocław, Poland
| | - Kuba Ptaszkowski
- Department of Clinical Biomechanics and Physiotherapy in Motor System Disorders, Wrocław Medical University, Wrocław, Poland
| | - Piotr Kuśnierczyk
- Laboratory of Immunogenetics and Tissue Immunology, Department of Clinical Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Monika Jasek
- Laboratory of Genetics and Epigenetics of Human Diseases, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Lidia Karabon
- Laboratory of Genetics and Epigenetics of Human Diseases, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland,*Correspondence: Anna Andrzejczak, ; Lidia Karabon,
| |
Collapse
|
2
|
Basile MS, Bramanti P, Mazzon E. The Role of Cytotoxic T-Lymphocyte Antigen 4 in the Pathogenesis of Multiple Sclerosis. Genes (Basel) 2022; 13:genes13081319. [PMID: 35893056 PMCID: PMC9394409 DOI: 10.3390/genes13081319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 02/05/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune neurodegenerative disorder of the central nervous system that presents heterogeneous clinical manifestations and course. It has been shown that different immune checkpoints, including Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4), can be involved in the pathogenesis of MS. CTLA-4 is a critical regulator of T-cell homeostasis and self-tolerance and represents a key inhibitor of autoimmunity. In this scopingreview, we resume the current preclinical and clinical studies investigating the role of CTLA-4 in MS with different approaches. While some of these studies assessed the expression levels of CTLA-4 on T cells by comparing MS patients with healthy controls, others focused on the evaluation of the effects of common MS therapies on CTLA-4 modulation or on the study of the CTLA-4 blockade or deficiency in experimental autoimmune encephalomyelitis models. Moreover, other studies in this field aimed to discover if the CTLA-4 gene might be involved in the predisposition to MS, whereas others evaluated the effects of treatment with CTLA4-Ig in MS. Although these results are of great interest, they are often conflicting. Therefore, further studies are needed to reveal the exact mechanisms underlying the action of a crucial immune checkpoint such as CTLA-4 in MS to identify novel immunotherapeutic strategies for MS patients.
Collapse
|
3
|
Wojciechowicz K, Spodzieja M, Lisowska KA, Wardowska A. The role of the BTLA-HVEM complex in the pathogenesis of autoimmune diseases. Cell Immunol 2022; 376:104532. [PMID: 35537322 DOI: 10.1016/j.cellimm.2022.104532] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/11/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022]
|
4
|
Alanazi FE, As Sobeai HM, Alhazzani K, Al-Dhfyan A, Alshammari MA, Alotaibi M, Al-hosaini K, Korashy HM, Alhoshani A. Metformin attenuates V-domain Ig suppressor of T-cell activation through the aryl hydrocarbon receptor pathway in Melanoma: In Vivo and In Vitro Studies. Saudi Pharm J 2022; 30:138-149. [PMID: 35528855 PMCID: PMC9072704 DOI: 10.1016/j.jsps.2021.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/27/2021] [Indexed: 11/06/2022] Open
Abstract
Melanoma is an aggressive skin cancer with a high rate of metastasis to other organs. Recent studies specified the overexpression of V-domain Ig suppressor of T-cell activation (VISTA) and Aryl Hydrocarbon Receptor (AHR) in melanoma. Metformin shows anti-tumor activities in several cancer types. However, the mechanism is unclear. This study aims to investigate the inhibitory effect of metformin on VISTA via AHR in melanoma cells (CHL-1, B16) and animal models. VISTA and AHR levels were assessed by qPCR, Western blot, immunofluorescence microscope, flow cytometry, and immunohistochemistry. Here, metformin significantly decreased VISTA and AHR levels in vitro and in vivo. Furthermore, metformin inhibited all AHR-regulated genes. VISTA levels were dramatically inhibited by AHR modulations using shRNA and αNF, confirming the central role of AHR in VISTA. Finally, melanoma cells were xenografted in C57BL/6 and nude mice. Metformin significantly reduced the tumor volume and growth rate. Likewise, VISTA and AHR-regulated protein levels were suppressed in both models. These findings demonstrate for the first time that VISTA is suppressed by metformin and identified a new regulatory mechanism through AHR. The data suggest that metformin could be a new potential therapeutic strategy to treat melanoma patients combined with targeted immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Fawaz E. Alanazi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
- Pharmacy Services Department, Security Forces Hospital Program, P.O. Box 3643, Riyadh 11481, Saudi Arabia
| | - Homood M. As Sobeai
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Khalid Alhazzani
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Abdullah Al-Dhfyan
- Stem Cell & Tissue Re-Engineering, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - Musaad A Alshammari
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Moureq Alotaibi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Khaled Al-hosaini
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Hesham M. Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Ali Alhoshani
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
5
|
Dolatkhah K, Alizadeh N, Mohajjel-Shoja H, Abdoli Shadbad M, Hajiasgharzadeh K, Aghebati-Maleki L, Baghbanzadeh A, Hosseinkhani N, Karim Ahangar N, Baradaran B. B7 immune checkpoint family members as putative therapeutics in autoimmune disease: An updated overview. Int J Rheum Dis 2022; 25:259-271. [PMID: 34994525 DOI: 10.1111/1756-185x.14273] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 12/11/2021] [Accepted: 12/18/2021] [Indexed: 12/23/2022]
Abstract
Autoimmune diseases, especially among young people in the US, are one of the leading causes of morbidity and death. The immune responses are the fundamental pathogenicity of autoimmune disorders. The equilibrium between stimulatory and inhibitory signals is critical for the stimulation, migration, survival, and T cell-related immune responses. The B7 family can substantially regulate T cell-mediated immune responses. Nevertheless, recent breakthroughs in immune checkpoint blockade in cancer immunotherapy have facilitated autoimmune diseases, especially among the prone populations. In the current study, we tried to concisely review the role of the B7 family in regulating immune reactions and the influence of immune checkpoint inhibitors on autoimmunity development.
Collapse
Affiliation(s)
- Katayoun Dolatkhah
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hanieh Mohajjel-Shoja
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | | | | | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negar Hosseinkhani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Noora Karim Ahangar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Deepak HB, Prince SE, Deshpande P. Effect of baricitinib in regulating programmed death 1 and ligand programmed cell death ligand 1 through JAK/STAT pathway in psoriasis. Indian J Pharmacol 2022; 54:183-193. [PMID: 35848689 PMCID: PMC9396682 DOI: 10.4103/ijp.ijp_1089_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVES Psoriasis is a chronic infectious skin disease triggered by an autoimmune process involving T-cell-mediated hyper-proliferation of keratinocytes. The objective of this study is to assess the modulation of programmed death 1 (PD-1) and its ligand programmed cell death ligand 1 (PD-L1) through JAK/STAT pathway during the development of a psoriasis-like disease by both in vitro and in vivo model. Baricitinib, a known inhibitor of JAK1 and JAK2, was used to study the impact on PD-1 and PD-L1. MATERIALS AND METHODS Human peripheral blood mononuclear cells (PBMC) were stimulated with either anti-CD3/CD28 or PMA/Ionomycin, to modulate level of PD-1 and PD-L1 under psoriasis-like condition. Interferon-gamma (IFNγ) was used to treat HaCaT cells to mimic the diseased keratinocytes found in Psoriatic patients. Psoriasis was induced with Imiquimod (IMQ) in animal model to study the cross-talk between different cell types and pathways. RESULTS Expression levels of PD-1 and PD-L1 in PBMC, and secretion of cytokines, namely tumor necrosis factor-α (TNFα), IFNγ, interleukin (IL)-6, and IL-1 β, were down-regulated on treatment with baricitinib. Further, in IFNγ-treated HaCaT cells (keratinocytes) mRNA levels of KRT-17 and PD-L1 were up-regulated.). Interestingly, in IFNγ-treated HaCat cells baricitinib decreased the levels of inflammatory cytokines such as IL-1 β, IL-6, and TNFα along with KRT-17 and PD-L1. On IFNγ-treatment. Data from both PBMC and HaCaT suggest an anti-inflammatory role for this compound. Accordingly, baricitinib was able to alleviate disease symptom in IMQ induce mice model of psoriasis. As a consequence of baricitinib treatment down-regulation of p-STAT3, PD- and PD-L1 expression levels were observed. CONCLUSION This study demonstrates a crosstalk between JAK/STAT and PD-1/PD-L1 pathways. It also demonstrates that cytokines such as IFNγ and IL-17 are down-regulated by baricitinib. We believe decreased expressions of PD-1 and PD-L1 may be a consequence of baricitinib-induced down-regulation of IFNγ and IL-17. More importantly, our data from the acute model of psoriasis indicates that PD-L1 behaves as a T-cell-associated T-cell-associated surrogate activation marker rather than immunosuppressive marker in early phase of psoriasis. Therefore it does not exhibit a causal relationship to disease.
Collapse
Affiliation(s)
- H. B. Deepak
- Department of Biomedical Sciences, School of Biosciences and Technology, VIT, Vellore, Tamil Nadu, India,Department of Biology, Jubilant Biosys Ltd., Bengaluru, Karnataka, India
| | - Sabina Evan Prince
- Department of Biomedical Sciences, School of Biosciences and Technology, VIT, Vellore, Tamil Nadu, India,Address for correspondence: Dr. Sabina Evan Prince, Department of Biomedical Sciences, School of Biosciences and Technology, VIT, Vellore - 632 014, Tamil Nadu, India. E-mail:
| | - Pratima Deshpande
- Department of Biology, Jubilant Biosys Ltd., Bengaluru, Karnataka, India
| |
Collapse
|
7
|
Karabon L, Andrzejczak A, Ciszak L, Tomkiewicz A, Szteblich A, Bojarska-Junak A, Roliński J, Wołowiec D, Wróbel T, Kosmaczewska A. BTLA Expression in CLL: Epigenetic Regulation and Impact on CLL B Cell Proliferation and Ability to IL-4 Production. Cells 2021; 10:cells10113009. [PMID: 34831232 PMCID: PMC8616199 DOI: 10.3390/cells10113009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/22/2021] [Accepted: 10/31/2021] [Indexed: 12/13/2022] Open
Abstract
In our previous study, while chronic lymphocytic leukemia (CLL) cases showed higher levels of B and T lymphocyte attenuator (BTLA) mRNA compared to controls, lower BTLA protein expression was observed in cases compared to controls. Hence we hypothesize that micro RNA (miR) 155-5p regulates BTLA expression in CLL. In line with earlier data, expression of BTLA mRNA and miR-155-5p is elevated in CLL (p = 0.034 and p = 0.0006, respectively) as well as in MEC-1 cell line (p = 0.009 and 0.016, respectively). Inhibition of miR-155-5p partially restored BTLA protein expression in CLL patients (p = 0.01) and in MEC-1 cell lines (p = 0.058). Additionally, we aimed to evaluate the significance of BTLA deficiency in CLL cells on proliferation and IL-4 production of B cells. We found that secretion of IL-4 is not dependent on BTLA expression, since fractions of BTLA positive and BTLA negative B cells expressing intracellular IL-4 were similar in CLL patients and controls. We demonstrated that in controls the fraction of proliferating cells is lower in BTLA positive than in BTLA negative B cells (p = 0.059), which was not observed in CLL. However, the frequency of BTLA positive Ki67+ B cells in CLL was higher compared to corresponding cells from controls (p = 0.055) while there were no differences between the examined groups regarding frequency of BTLA negative Ki67+ B cells. Our studies suggest that miR-155-5p is involved in BTLA deficiency, affecting proliferation of CLL B cells, which may be one of the mechanisms responsible for CLL pathogenesis.
Collapse
MESH Headings
- Aged
- Base Sequence
- Cell Line, Tumor
- Cell Proliferation/genetics
- Epigenesis, Genetic
- Female
- Gene Expression Regulation, Leukemic
- Humans
- Interleukin-4/biosynthesis
- Ki-67 Antigen/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Male
- MicroRNAs/genetics
- MicroRNAs/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
Collapse
Affiliation(s)
- Lidia Karabon
- Laboratory of Genetics and Epigenetics of Human Diseases, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigl 12 Str., 53-114 Wroclaw, Poland; (A.A.); (A.T.)
- Department and Clinic of Urology and Oncologic Urology, Wroclaw Medical University, Borowska Str. 213, 50-556 Wroclaw, Poland
- Correspondence:
| | - Anna Andrzejczak
- Laboratory of Genetics and Epigenetics of Human Diseases, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigl 12 Str., 53-114 Wroclaw, Poland; (A.A.); (A.T.)
| | - Lidia Ciszak
- Laboratory of Immunopathology, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigl 12 Str., 53-114 Wroclaw, Poland; (L.C.); (A.S.); (A.K.)
| | - Anna Tomkiewicz
- Laboratory of Genetics and Epigenetics of Human Diseases, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigl 12 Str., 53-114 Wroclaw, Poland; (A.A.); (A.T.)
| | - Aleksandra Szteblich
- Laboratory of Immunopathology, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigl 12 Str., 53-114 Wroclaw, Poland; (L.C.); (A.S.); (A.K.)
| | - Agnieszka Bojarska-Junak
- Department of Clinical Immunology, Medical University of Lublin, ul. Chodźki 4a, 20-093 Lublin, Poland; (A.B.-J.); (J.R.)
| | - Jacek Roliński
- Department of Clinical Immunology, Medical University of Lublin, ul. Chodźki 4a, 20-093 Lublin, Poland; (A.B.-J.); (J.R.)
| | - Dariusz Wołowiec
- Department and Clinic of Hematology, Blood Neoplasms, and Bone Marrow Transplantation, Medical University, Wybrzeże Ludwika Pasteura 4, 50-367 Wroclaw, Poland; (D.W.); (T.W.)
| | - Tomasz Wróbel
- Department and Clinic of Hematology, Blood Neoplasms, and Bone Marrow Transplantation, Medical University, Wybrzeże Ludwika Pasteura 4, 50-367 Wroclaw, Poland; (D.W.); (T.W.)
| | - Agata Kosmaczewska
- Laboratory of Immunopathology, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigl 12 Str., 53-114 Wroclaw, Poland; (L.C.); (A.S.); (A.K.)
| |
Collapse
|
8
|
Harris KM, Smilek DE, Byron M, Lim N, Barry WT, McNamara J, Garcet S, Konrad RJ, Stengelin M, Bathala P, Korman NJ, Feldman SR, Boh EE, Barber K, Laumann AE, Helfrich YR, Krueger GG, Sofen H, Bissonnette R, Krueger JG. Effect of Costimulatory Blockade With Abatacept After Ustekinumab Withdrawal in Patients With Moderate to Severe Plaque Psoriasis: The PAUSE Randomized Clinical Trial. JAMA Dermatol 2021; 157:1306-1315. [PMID: 34643650 PMCID: PMC8515260 DOI: 10.1001/jamadermatol.2021.3492] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Question Does blockade of CD28/B7 costimulatory signaling with abatacept suppress the psoriasis molecular signature and prevent psoriasis relapse after ustekinumab withdrawal? Findings In this parallel-design, double-blind randomized clinical trial of 91 adults with moderate to severe plaque psoriasis, costimulatory blockade with abatacept did not prevent psoriasis relapse and did not maintain suppression of the pathogenic psoriasis molecular signature following ustekinumab withdrawal. Meaning In this study, abatacept did not prevent psoriasis relapse, which may rely on alternative, compensatory mechanisms of residual T-cell activation in skin. Importance Psoriasis relapse may involve compensatory T-cell activation pathways in the presence of CD28-CD80/CD86 blockade with abatacept. Objective To determine whether costimulatory signaling blockade with abatacept prevents psoriasis relapse after ustekinumab withdrawal. Design, Setting, and Participants Psoriasis Treatment with Abatacept and Ustekinumab: a Study of Efficacy (PAUSE), a parallel-design, double-blind, placebo-controlled randomized clinical trial, was conducted at 10 sites in the US and Canada. Participant enrollment opened on March 19, 2014, and concluded on April 11, 2016. Participants were adults with moderate to severe plaque psoriasis and received ustekinumab in a lead-in phase. Those who responded to ustekinumab at week 12 were randomized 1:1 to either the continued with ustekinumab group (ustekinumab group) or the switched to abatacept group (abatacept group). Treatment was discontinued at week 39, and participants were followed up for psoriasis relapse until week 88. Statistical analyses were performed in the intention-to-treat (ITT) and safety samples from May 3, 2018, to July 6, 2021. Interventions Participants received subcutaneous ustekinumab at weeks 0 and 4 (45 mg per dose for those ≤100 kg; 90 mg per dose for those >100 kg). Participants randomized to the abatacept group at week 12 received subcutaneous abatacept, 125 mg weekly, from weeks 12 to 39 and ustekinumab placebo at weeks 16 and 28. Participants randomized to the ustekinumab group received ustekinumab at weeks 16 and 28 and abatacept placebo weekly from weeks 12 to 39. Main Outcomes and Measures The primary end point was the proportion of participants with psoriasis relapse (loss of ≥50% of the initial Psoriasis Area and Severity Index improvement) between weeks 12 and 88. Secondary end points included time to psoriasis relapse, proportion of participants with psoriasis relapse between weeks 12 and 40, and adverse events. The psoriasis transcriptome and serum cytokines were evaluated. Results A total of 108 participants (mean [SD] age, 46.1 [12.1] years; 73 [67.6%] men) were treated with open-label ustekinumab; 91 were randomized to blinded treatment. Similar proportions of participants in the abatacept group and the ustekinumab group relapsed between weeks 12 and 88 (41 of 45 [91.1%] vs 40 of 46 [87.0%]; P = .41). Median time to relapse from the last dose of ustekinumab was similar between groups as well: 36 weeks (95% CI, 36-48 weeks) in the abatacept group vs 32 weeks (95% CI, 28-40 weeks) in the ustekinumab group. Similar numbers and rates of adverse events occurred. Abatacept did not maintain suppression of the pathogenic IL-23-mediated psoriasis molecular signature in lesions after ustekinumab withdrawal, and serum IL-19 levels increased. Conclusions and Relevance This parallel-design, double-blind randomized clinical trial found that abatacept did not prevent psoriasis relapse that occurred after ustekinumab withdrawal because it did not completely block the pathogenic psoriasis molecular pathways that led to relapse. Trial Registration ClinicalTrials.gov Identifier: NCT01999868
Collapse
Affiliation(s)
- Kristina M Harris
- Biomarker and Discovery Research, Immune Tolerance Network, University of California, San Francisco, San Francisco
| | - Dawn E Smilek
- Clinical Trials Group, Clinical and Translational Medicine, Immune Tolerance Network, University of California, San Francisco, San Francisco
| | | | - Noha Lim
- Biomarker and Discovery Research, Immune Tolerance Network, University of California, San Francisco, San Francisco
| | | | - James McNamara
- Autoimmunity and Mucosal Immunology Branch, Division of Allergy, Immunology, and Transplantation/National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | | | - Robert J Konrad
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | | | | | - Neil J Korman
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Steven R Feldman
- Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Erin E Boh
- Health Sciences Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Kirk Barber
- Department of Medicine (Dermatology), University of Calgary, Calgary, Alberta, Canada
| | - Anne E Laumann
- Department of Dermatology, Northwestern University, Colorado Springs, Colorado
| | | | - Gerald G Krueger
- Department of Dermatology, University of Utah School of Medicine, Salt Lake City
| | - Howard Sofen
- Dermatology, David Geffen UCLA (University of California, Los Angeles) School of Medicine, Los Angeles
| | | | | |
Collapse
|
9
|
Lower Functional and Proportional Characteristics of Cord Blood Treg of Male Newborns Compared with Female Newborns. Biomedicines 2021; 9:biomedicines9020170. [PMID: 33572097 PMCID: PMC7915235 DOI: 10.3390/biomedicines9020170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 11/16/2022] Open
Abstract
Understanding the early events involved in the induction of immune tolerance to harmless environmental antigens and microbiota compounds could reveal potential targets for allergic disease therapy or prevention. Regulatory T cells (Treg), particularly induced Treg (iTreg), are crucial for the induction and maintenance of tolerance against environmental antigens including allergens. A decrease in the number and/or function of Treg or iTreg could represent an early predictor of allergy development. We analyzed proportional and functional properties of Treg in the cord blood of children of allergic mothers (neonates at high risk of allergy development) and healthy mothers (neonates with relatively low risk of allergy development). We observed a higher number of induced Treg in the cord blood of females compared to males, suggesting an impaired capacity of male immunity to set up tolerance to allergens, which could contribute to the higher incidence of allergy observed in male infants. The decreased proportion of iTreg in cord blood compared with maternal peripheral blood documents the general immaturity of the neonatal immune system. We observed a positive correlation in the demethylation of the Treg-specific demethylated region (TSDR) and the proportion of Treg in cord blood. Our data suggest that immaturity of the neonatal immune system is more severe in males, predisposing them to increased risk of allergy development.
Collapse
|
10
|
Abnormal Expression of BTLA and CTLA-4 Immune Checkpoint Molecules in Chronic Lymphocytic Leukemia Patients. J Immunol Res 2020; 2020:6545921. [PMID: 32775467 PMCID: PMC7407019 DOI: 10.1155/2020/6545921] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/07/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by the peripheral accumulation of neoplastic B cells and is frequently complicated by the systemic immunosuppression associated with an impairment in B and T lymphocyte activation. We hypothesized that the expression of immune checkpoint suppressors B and T lymphocyte attenuator (BTLA) and cytotoxic T lymphocyte antigen (CTLA-4) is disturbed in both lymphocyte subpopulations in CLL. The expression of CTLA-4 and BTLA mRNA was determined by real-time PCR, while CTLA-4 protein expression (surface or intracellular) was estimated in BTLA+ lymphocytes by flow cytometry. In CLL patients, we observed a higher gene transcript level of BTLA and CTLA-4 than in healthy individuals in both freshly isolated and PMA stimulated B and T cells. Remarkably, lower amounts of both inhibitory proteins were found in peripheral blood (PB) CLL B cells, whereas normal BTLA and elevated CTLA-4 were found in T cells. Consistently, there was a prevalence of CTLA-4+ cells within circulating BTLA+ T cells cells of patients confronting PB healthy cells. After in vitro stimulation, the only change found in CLL patients was a decrease in BTLA expression in B and T lymphocytes. In contrast, healthy lymphocytes responded more vigorously as regards the BTLA and CTLA expression with substantially higher frequency of CD69+ cells under the stimulating condition compared to corresponding cells from the CLL group. Our results indicate that CLL development is associated with the affected expression of BTLA and CTLA-4 checkpoint receptors in PB and its impaired expression might be associated with lowering of the threshold for B cell activation and proliferation, while upregulated CTLA-4 expression in CLL peripheral BTLA+ T cells may contribute to suppressed T cell effector functions. This hypothesis needs to be validated in future studies, which would allow us to explain how the increased or decreased expression of these molecules affects the cell function.
Collapse
|
11
|
Zuo HX, Jin Y, Wang Z, Li MY, Zhang ZH, Wang JY, Xing Y, Ri MH, Jin CH, Xu GH, Piao LX, Ma J, Jin X. Curcumol inhibits the expression of programmed cell death-ligand 1 through crosstalk between hypoxia-inducible factor-1α and STAT3 (T705) signaling pathways in hepatic cancer. JOURNAL OF ETHNOPHARMACOLOGY 2020; 257:112835. [PMID: 32278762 DOI: 10.1016/j.jep.2020.112835] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/16/2020] [Accepted: 04/01/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Curcuma wenyujin is a Chinese traditional herbal medicine that is commonly used as an anti-oxidant, anti-proliferative, and anti-tumorigenic agent. Curcumol is a representative index component for the quality control of the essential oil of Curcuma wenyujin, which is currently used as an anti-cancer drug, and is included in the State Pharmacopoeia Commission of the People's Republic of China (2005). However, the mechanisms of action and molecular functions of curcumol are not yet fully elucidated. AIM OF THE STUDY This study aimed to identify new effects of curcumol from the perspective of cancer immunotherapy. MATERIALS AND METHODS The underlying mechanism of the inhibition of programmed cell death-ligand 1 (PD-L1) activation by curcumol was investigated in vitro via homology modeling, molecular docking experiments, luciferase reporter assays, MTT assays, RT-PCR, western blotting, and immunofluorescence assays. Changes in cellular proliferation, angiogenesis, and the tumor-killing activity of T-cells were analyzed via EdU labeling, colony formation, flow cytometry, wound-healing, Matrigel Transwell invasion, tube formation, and T-cell killing. The anti-tumor activity of curcumol was assessed in vivo in a murine xenograft model using Hep3B cells. RESULTS Curcumol reduced the expression of phosphorylated signal transducer and activator of transcription 3 (p-STAT3) via JAK1, JAK2, and Src pathways and inhibited hypoxia-inducible factor-1α (HIF-1α) protein synthesis via mTOR/p70S6K/eIF4E and MAPK pathways. Furthermore, we revealed crosstalk between STAT3 and HIF-1α pathways, which collaboratively regulated PD-L1 activation, and that curcumol played a role in this regulation. Curcumol inhibited cell proliferation, S-phase progression, tube formation, invasion, and metastasis by inhibiting PD-L1. In addition, curcumol restored the activity of cytotoxic T-cells and their capacity for tumor cell killing by inhibiting PD-L1. In vivo experiments confirmed that curcumol inhibited tumor growth in a xenograft model. CONCLUSIONS These results illustrated that curcumol inhibits the expression of PD-L1 through crosstalk between HIF-1α and p-STAT3 (T705) signaling pathways in hepatic cancer. Thus, curcumol might represent a promising lead compound for the development of new targeted anti-cancer therapeutics.
Collapse
Affiliation(s)
- Hong Xiang Zuo
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China; College of Pharmacy, Beihua University, Jilin, 132013, Jilin Province, China
| | - Yong Jin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Zhe Wang
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Ming Yue Li
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Zhi Hong Zhang
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Jing Ying Wang
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Yue Xing
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Myong Hak Ri
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Cheng Hua Jin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Guang Hua Xu
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Lian Xun Piao
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Juan Ma
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China.
| | - Xuejun Jin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China.
| |
Collapse
|
12
|
Agrawal B. New therapeutic targets for cancer: the interplay between immune and metabolic checkpoints and gut microbiota. Clin Transl Med 2019; 8:23. [PMID: 31468283 PMCID: PMC6715761 DOI: 10.1186/s40169-019-0241-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 08/16/2019] [Indexed: 02/07/2023] Open
Abstract
Transformation and growth of tumor cells are associated with profound alterations in neighbouring cells and their environment, together forming the tumor microenvironment (TME). The TME provides a conducive but complex milieu for the tumors to thrive while incapacitating the immune cells that home there as part of our natural immunosurveillance mechanism. The orchestration of this successful survival strategy by tumor cells is associated with exploitation of numerous metabolic and immune checkpoints, as well as metabolic reprogramming in the tumor cells. Together these form an intricate network of feedback mechanisms that favor the growing tumor. In addition, an ecosystem of microbiota, proximal or distal to tumors, influences the successful survival or elimination of tumor cells mediated by immune cells. Discovery and clinical application of immune checkpoint inhibitors (ICIs) i.e., monoclonal antibodies (mAbs) blocking specific immune checkpoints CTLA-4 and PD-1/PD-L1, have revolutionized therapy of various cancers. However, they are still associated with limited response rates, severe immune-related adverse events, development of resistance, and more serious exacerbation of cancer progression termed hyper-progressive disease. Checkpoint inhibitors only represent a milestone and not the finish-line in the quest for treating and curing cancer. Efforts are underway to investigate and develop inhibitors of other immune as well as metabolic checkpoint molecules. Future therapy for various cancers is projected to target immune and metabolic checkpoints and the microbiota together.
Collapse
Affiliation(s)
- Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
13
|
Roy S, Awasthi A. Emerging roles of noncoding RNAs in T cell differentiation and functions in autoimmune diseases. Int Rev Immunol 2019; 38:232-245. [PMID: 31411520 DOI: 10.1080/08830185.2019.1648454] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Noncoding RNA comprises of microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) that are abundantly present in mammalian transcriptome. These noncoding RNAs have been implicated in multiple biological processes through the regulation of gene expression. Each of these noncoding RNAs were found to have multiple genes targets. Emerging literature indicated the role of noncoding RNAs in shaping the immune responses which include immune cell development, helper T (Th) cell differentiation as well as maintenance of immune homeostasis by inducing the interplay between effector and regulatory T cells. Dysregulated expression and functions of noncoding RNAs in the immune system leads to aberrations in immune response that lead to the induction of tissue inflammation in autoimmune diseases. In this review, we summarize the current advances of post-transcriptional regulation, focusing on the functions of noncoding RNAs (miRNAs and lncRNAs) during differentiation of Th cells in tissue inflammation in autoimmune diseases.
Collapse
Affiliation(s)
- Suyasha Roy
- Immuno-Biology Lab, Translational Health Science and Technology Institute , Faridabad , India
| | - Amit Awasthi
- Immuno-Biology Lab, Translational Health Science and Technology Institute , Faridabad , India
| |
Collapse
|
14
|
Wang T, Jia Y, Chu B, Liu H, Dong X, Zhang Y. Nocardiosis in Kidney Disease Patients under Immunosuppressive Therapy: Case Report and Literature Review. Int J Med Sci 2019; 16:838-844. [PMID: 31337957 PMCID: PMC6643105 DOI: 10.7150/ijms.32440] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 05/05/2019] [Indexed: 12/17/2022] Open
Abstract
The increased use of novel and powerful immunosuppressive drugs in kidney diseases may concomitantly expose the patients to higher risk of opportunistic infections, some of which still remain underdiagnosed thus mishandled. As such, we recently had a less prepared encounter of pulmonary nocardial infection in an ANCA-associated renal vasculitis patient under steroid therapy. Despite the use of broad-spectrum antimicrobials including micafungin, the infection was still unbridled and eventually culminated in lethal brain abscess. We thus chose to renew the knowledge of the clinical features, imaging manifestations, differential diagnosis, specific laboratory tests and unique treatment about this rare infection in kidney diseases patients under immunosuppressive therapy. In addition, CT images of easily confused pulmonary lesions superimposed on kidney diseases were also retrieved from our depository. Moreover, impaired renal function as a risk factor for infection and pharmacological options for the treatment were also focused. By sharing our hard-learnt experience and reviewing the literatures, our report may contribute to the awareness among the clinicians in general and nephrologists in particular of this rare disease in susceptible patients and facilitate a swift thus life-saving treatment.
Collapse
Affiliation(s)
- Tao Wang
- Department of Science and Education, HeBei General Hospital, No.348 West HePing Boulevard, ShiJiaZhuang 050051, P.R. China
| | - Yun Jia
- Department of Clinical Immunology, Xijing Hospital, the Fourth Military Medical University, No.127 West Changle Road, Xi'an 710032, P.R. China
| | - Bao Chu
- Department of Neurology, No.348 West HePing Boulevard, ShiJiaZhuang 050051, P.R. China
| | - HongTao Liu
- Department of Pharmacology, No.348 West HePing Boulevard, ShiJiaZhuang 050051, P.R. China
| | - XiaoLi Dong
- Department of Neurology, No.348 West HePing Boulevard, ShiJiaZhuang 050051, P.R. China
| | - Yan Zhang
- Department of Dermatology, the 4th Affiliated Hospital of HeBei Medical University, No.12 JianKang Road, ShiJiaZhuang 050011, P.R. China
| |
Collapse
|
15
|
Černý V, Hrdý J, Novotná O, Petrásková P, Boráková K, Kolářová L, Prokešová L. Distinct characteristics of Tregs of newborns of healthy and allergic mothers. PLoS One 2018; 13:e0207998. [PMID: 30475891 PMCID: PMC6258229 DOI: 10.1371/journal.pone.0207998] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 11/11/2018] [Indexed: 12/19/2022] Open
Abstract
Allergic diseases represent a major issue in clinical and experimental immunology due to their high and increasing incidence worldwide. Allergy status of the mother remains the best predictor of an individual's increased risk of allergy development. Dysregulation of the balance between different branches of immune response, chiefly excessive polarization towards Th2, is the underlying cause of allergic diseases. Regulatory T cells (Tregs) play a pivotal role in the timely establishment of physiological immune polarization and are crucial for control of allergy. In our study we used flow cytometry to assess Tregs in cord blood of newborns of healthy (n = 121) and allergic (n = 108) mothers. We observed a higher percentage of Tregs (CD4+CD25+CD127lowFoxP3+) in cord blood of children of allergic mothers. However, the percentage of cells expressing extracellular (PD-1, CTLA-4, GITR) and intracellular (IL-10, TGF-β) markers of function was lower (significantly for PD-1 and IL-10) within Tregs of these children. Furthermore, Helios- induced Tregs in the cord blood of children of allergic mothers were decreased. These results were supported by a decrease in plasma levels of IL-10 and TGF-β in cord blood of newborns of allergic mothers, implying lower tolerogenic capacity on the systemic level. Taken together, these findings reflect deficient function of Tregs in the group with higher risk of allergy development. This may be caused by a lower maturation status of the immune system, specifically Tregs, at birth. Such immaturity may represent an important mechanism involved in the increased risk of allergy in children of allergic mothers.
Collapse
Affiliation(s)
- Viktor Černý
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Jiří Hrdý
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Olga Novotná
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Petra Petrásková
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | | | - Libuše Kolářová
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Ludmila Prokešová
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| |
Collapse
|
16
|
Agrawal B, Gupta N, Konowalchuk JD. MUC1 Mucin: A Putative Regulatory (Checkpoint) Molecule of T Cells. Front Immunol 2018; 9:2391. [PMID: 30405607 PMCID: PMC6204366 DOI: 10.3389/fimmu.2018.02391] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 09/26/2018] [Indexed: 01/30/2023] Open
Abstract
T lymphocytes are at the center of inducing an effective adaptive immune response and maintaining homeostasis. T cell responses are initiated through interactions between antigen presenting cells (APCs) and T cells. The type and strength of signals delivered through the T cell receptor (TCR) may modulate how the cells respond. The TCR-MHC (T cell receptor-major histocompatibility complex molecules) complex dictates the specificity, whereas co-stimulatory signals induced by interaction of various accessory cell surface molecules strengthen and optimize T cell responses. Multiple immune regulatory mechanisms brought about by co-inhibitory molecules expressed on T cells play a key role in orchestrating successful and non-damaging immunity. These co-inhibitory molecules are also referred to as initiators of immune check-points or co-inhibitory pathways. Knowledge of co-inhibitory pathways associated with activated T lymphocytes has allowed a better understanding of (a) the inflammatory and anti-inflammatory processes associated with infectious diseases and autoimmune diseases, and (b) mechanisms by which tumors evade immune attack. Many of these regulatory pathways are non-redundant and function in a highly concerted manner. Targeting them has provided effective approaches in treating cancer and autoimmune diseases. For this reason, it is valuable to identify any co-inhibitory molecules that affect these pathways. MUC1 mucin (CD227) has long been known to be expressed by epithelial cells and overexpressed by a multitude of adenocarcinomas. As long ago as 1998 we made a surprising discovery that MUC1 is also expressed by activated human T cells and we provided the first evidence of the role of MUC1 as a novel T cell regulator. Subsequent studies from different laboratories, as well as ours, supported an immuno-regulatory role of MUC1 in infections, inflammation, and autoimmunity that corroborated our original findings establishing MUC1 as a novel T cell regulatory molecule. In this article, we will discuss the experimental evidence supporting MUC1 as a putative regulatory molecule or a “checkpoint molecule” of T cells with implications as a novel biomarker and therapeutic target in chronic diseases such as autoimmunity, inflammation and cancer, and possibly infections.
Collapse
Affiliation(s)
- Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Nancy Gupta
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Jeffrey D Konowalchuk
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
17
|
Cha JH, Yang WH, Xia W, Wei Y, Chan LC, Lim SO, Li CW, Kim T, Chang SS, Lee HH, Hsu JL, Wang HL, Kuo CW, Chang WC, Hadad S, Purdie CA, McCoy AM, Cai S, Tu Y, Litton JK, Mittendorf EA, Moulder SL, Symmans WF, Thompson AM, Piwnica-Worms H, Chen CH, Khoo KH, Hung MC. Metformin Promotes Antitumor Immunity via Endoplasmic-Reticulum-Associated Degradation of PD-L1. Mol Cell 2018; 71:606-620.e7. [PMID: 30118680 PMCID: PMC6786495 DOI: 10.1016/j.molcel.2018.07.030] [Citation(s) in RCA: 560] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/26/2018] [Accepted: 07/23/2018] [Indexed: 11/17/2022]
Abstract
Metformin has been reported to possess antitumor activity and maintain high cytotoxic T lymphocyte (CTL) immune surveillance. However, the functions and detailed mechanisms of metformin's role in cancer immunity are not fully understood. Here, we show that metformin increases CTL activity by reducing the stability and membrane localization of programmed death ligand-1 (PD-L1). Furthermore, we discover that AMP-activated protein kinase (AMPK) activated by metformin directly phosphorylates S195 of PD-L1. S195 phosphorylation induces abnormal PD-L1 glycosylation, resulting in its ER accumulation and ER-associated protein degradation (ERAD). Consistently, tumor tissues from metformin-treated breast cancer patients exhibit reduced PD-L1 levels with AMPK activation. Blocking the inhibitory signal of PD-L1 by metformin enhances CTL activity against cancer cells. Our findings identify a new regulatory mechanism of PD-L1 expression through the ERAD pathway and suggest that the metformin-CTLA4 blockade combination has the potential to increase the efficacy of immunotherapy.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/genetics
- AMP-Activated Protein Kinases/immunology
- Animals
- Antineoplastic Agents/pharmacology
- B7-H1 Antigen/genetics
- B7-H1 Antigen/immunology
- CTLA-4 Antigen/genetics
- CTLA-4 Antigen/immunology
- Cell Line, Tumor
- Endoplasmic Reticulum/drug effects
- Endoplasmic Reticulum/genetics
- Endoplasmic Reticulum/metabolism
- Endoplasmic Reticulum-Associated Degradation
- Epithelial Cells/cytology
- Epithelial Cells/drug effects
- Epithelial Cells/immunology
- Female
- Gene Expression Regulation, Neoplastic
- Glycosylation
- Humans
- Hypoglycemic Agents/pharmacology
- Mammary Glands, Human/cytology
- Mammary Glands, Human/drug effects
- Mammary Glands, Human/immunology
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Metformin/pharmacology
- Mice
- Mice, Inbred NOD
- Phosphorylation
- Serine/metabolism
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Jong-Ho Cha
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 151-742, Korea
| | - Wen-Hao Yang
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Weiya Xia
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yongkun Wei
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Li-Chuan Chan
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Seung-Oe Lim
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chia-Wei Li
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Taewan Kim
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shih-Shin Chang
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Heng-Huan Lee
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer L Hsu
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan; Department of Biotechnology, Asia University, Taichung 413, Taiwan
| | - Hung-Ling Wang
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan
| | - Chu-Wei Kuo
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Wei-Chao Chang
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan; Genomics Research Center, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Sirwan Hadad
- Department of Surgery, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Colin A Purdie
- Department of Pathology, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
| | - Aaron M McCoy
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shirong Cai
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yizheng Tu
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer K Litton
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elizabeth A Mittendorf
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stacy L Moulder
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - William F Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alastair M Thompson
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Helen Piwnica-Worms
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Chung-Hsuan Chen
- Genomics Research Center, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, University of Texas Health Science Center, Houston, TX 77030, USA; Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan; Department of Biotechnology, Asia University, Taichung 413, Taiwan.
| |
Collapse
|
18
|
Sawaf M, Fauny JD, Felten R, Sagez F, Gottenberg JE, Dumortier H, Monneaux F. Defective BTLA functionality is rescued by restoring lipid metabolism in lupus CD4+ T cells. JCI Insight 2018; 3:99711. [PMID: 29997289 DOI: 10.1172/jci.insight.99711] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/29/2018] [Indexed: 12/11/2022] Open
Abstract
Coinhibitory receptors play an important role in the prevention of autoimmune diseases, such as systemic lupus erythematosus (SLE), by limiting T cell activation. B and T lymphocyte attenuator (BTLA) is an inhibitory receptor, similar to cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and programmed death 1 (PD1), that negatively regulates the immune response. The role of BTLA in the pathogenesis of autoimmune diseases in humans and, more specifically, in SLE is largely unknown. We investigated BTLA expression on various T cell subsets, and we did not observe significant variations of BTLA expression between lupus patients and healthy controls. However, the enhancement of BTLA expression after activation was significantly lower in SLE patients compared with that in healthy controls. Furthermore, we found an impaired capacity of BTLA to inhibit T cell activation in SLE due to a poor BTLA recruitment to the immunological synapse following T cell stimulation. Finally, we demonstrated that defective BTLA function can be corrected by restoring intracellular trafficking and by normalizing the lipid metabolism in lupus CD4+ T cells. Collectively, our results evidence that the BTLA signaling pathway is altered in SLE T cells and highlight the potential of targeting this pathway for the development of new therapeutic strategies in lupus.
Collapse
Affiliation(s)
- Matthieu Sawaf
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunologie, Immunopathologie et Chimie Thérapeutique, Strasbourg, France
| | - Jean-Daniel Fauny
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunologie, Immunopathologie et Chimie Thérapeutique, Strasbourg, France
| | - Renaud Felten
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunologie, Immunopathologie et Chimie Thérapeutique, Strasbourg, France.,Rheumatology Department, Strasbourg University Hospital, National Reference Center for Autoimmune Diseases, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Flora Sagez
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunologie, Immunopathologie et Chimie Thérapeutique, Strasbourg, France.,Rheumatology Department, Strasbourg University Hospital, National Reference Center for Autoimmune Diseases, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Jacques-Eric Gottenberg
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunologie, Immunopathologie et Chimie Thérapeutique, Strasbourg, France.,Rheumatology Department, Strasbourg University Hospital, National Reference Center for Autoimmune Diseases, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Hélène Dumortier
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunologie, Immunopathologie et Chimie Thérapeutique, Strasbourg, France
| | - Fanny Monneaux
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunologie, Immunopathologie et Chimie Thérapeutique, Strasbourg, France
| |
Collapse
|
19
|
Tavakolpour S. Current and future treatment options for pemphigus: Is it time to move towards more effective treatments? Int Immunopharmacol 2017; 53:133-142. [DOI: 10.1016/j.intimp.2017.10.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/18/2017] [Accepted: 10/20/2017] [Indexed: 02/07/2023]
|
20
|
Talepoor AG, Behnamfar N, Zibaeenezhad MJ, Doroudchi M. IL-17 producing CD4+CD45RO+ T-cells in atherosclerosis express GITR molecule. Artery Res 2017. [DOI: 10.1016/j.artres.2017.12.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
21
|
Xu Z, Ho S, Chang CC, Zhang QY, Vasilescu ER, Vlad G, Suciu-Foca N. Molecular and Cellular Characterization of Human CD8 T Suppressor Cells. Front Immunol 2016; 7:549. [PMID: 27965674 PMCID: PMC5127796 DOI: 10.3389/fimmu.2016.00549] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/16/2016] [Indexed: 12/29/2022] Open
Abstract
Bidirectional interactions between dendritic cells and Ag-experienced T cells initiate either a tolerogenic or immunogenic pathway. The outcome of these interactions is of crucial importance in malignancy, transplantation, and autoimmune diseases. Blockade of costimulation results in the induction of T helper cell anergy and subsequent differentiation of antigen-specific CD8+ T suppressor/regulatory cells (Ts). Ts, primed in the presence of inhibitory signals, exert their inhibitory function in an antigen-specific manner, a feature with tremendous clinical potential. In transplantation or autoimmunity, antigen-specific Ts can enforce tolerance to auto- or allo-antigens, while otherwise leaving the immune response to pathogens uninhibited. Alternatively, blockade of inhibitory receptors results in the generation of cytolytic CD8+ T cells, which is vital toward defense against tumors and viral diseases. Because CD8+ T cells are MHC Class I restricted, they are able to recognize HLA-bound antigenic peptides presented not only by APC but also on parenchymal cells, thus eliciting or suppressing auto- or allo-immune reactions.
Collapse
Affiliation(s)
- Zheng Xu
- Immunogenetics and Cellular Immunology, Department of Pathology and Cell Biology, Columbia University , New York, NY , USA
| | - Sophey Ho
- Immunogenetics and Cellular Immunology, Department of Pathology and Cell Biology, Columbia University , New York, NY , USA
| | - Chih-Chao Chang
- Immunogenetics and Cellular Immunology, Department of Pathology and Cell Biology, Columbia University , New York, NY , USA
| | - Qing-Yin Zhang
- Immunogenetics and Cellular Immunology, Department of Pathology and Cell Biology, Columbia University , New York, NY , USA
| | - Elena-Rodica Vasilescu
- Immunogenetics and Cellular Immunology, Department of Pathology and Cell Biology, Columbia University , New York, NY , USA
| | - George Vlad
- Immunogenetics and Cellular Immunology, Department of Pathology and Cell Biology, Columbia University , New York, NY , USA
| | - Nicole Suciu-Foca
- Immunogenetics and Cellular Immunology, Department of Pathology and Cell Biology, Columbia University , New York, NY , USA
| |
Collapse
|
22
|
Miller JFAP, Sadelain M. The journey from discoveries in fundamental immunology to cancer immunotherapy. Cancer Cell 2015; 27:439-49. [PMID: 25858803 DOI: 10.1016/j.ccell.2015.03.007] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/01/2015] [Accepted: 03/16/2015] [Indexed: 01/04/2023]
Abstract
Recent advances in cancer immunotherapy have directly built on 50 years of fundamental and technological advances that made checkpoint blockade and T cell engineering possible. In this review, we intend to show that research, not specifically designed to bring relief or cure to any particular disease, can, when creatively exploited, lead to spectacular results in the management of cancer. The discovery of thymus immune function, T cells, and immune surveillance bore the seeds for today's targeted immune interventions and chimeric antigen receptors.
Collapse
Affiliation(s)
- Jacques F A P Miller
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3050, Australia.
| | - Michel Sadelain
- The Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
23
|
Immunomodulatory effects of Lactobacillus rhamnosus GG on dendritic cells, macrophages and monocytes from healthy donors. J Funct Foods 2015. [DOI: 10.1016/j.jff.2014.12.040] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
24
|
Šedý J, Bekiaris V, Ware CF. Tumor necrosis factor superfamily in innate immunity and inflammation. Cold Spring Harb Perspect Biol 2014; 7:a016279. [PMID: 25524549 DOI: 10.1101/cshperspect.a016279] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The tumor necrosis factor superfamily (TNFSF) and its corresponding receptor superfamily (TNFRSF) form communication pathways required for developmental, homeostatic, and stimulus-responsive processes in vivo. Although this receptor-ligand system operates between many different cell types and organ systems, many of these proteins play specific roles in immune system function. The TNFSF and TNFRSF proteins lymphotoxins, LIGHT (homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for herpes virus entry mediator [HVEM], a receptor expressed by T lymphocytes), lymphotoxin-β receptor (LT-βR), and HVEM are used by embryonic and adult innate lymphocytes to promote the development and homeostasis of lymphoid organs. Lymphotoxin-expressing innate-acting B cells construct microenvironments in lymphoid organs that restrict pathogen spread and initiate interferon defenses. Recent results illustrate how the communication networks formed among these cytokines and the coreceptors B and T lymphocyte attenuator (BTLA) and CD160 both inhibit and activate innate lymphoid cells (ILCs), innate γδ T cells, and natural killer (NK) cells. Understanding the role of TNFSF/TNFRSF and interacting proteins in innate cells will likely reveal avenues for future therapeutics for human disease.
Collapse
Affiliation(s)
- John Šedý
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| | - Vasileios Bekiaris
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| | - Carl F Ware
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| |
Collapse
|
25
|
Smilek DE, Ehlers MR, Nepom GT. Restoring the balance: immunotherapeutic combinations for autoimmune disease. Dis Model Mech 2014; 7:503-13. [PMID: 24795433 PMCID: PMC4007402 DOI: 10.1242/dmm.015099] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Autoimmunity occurs when T cells, B cells or both are inappropriately activated, resulting in damage to one or more organ systems. Normally, high-affinity self-reactive T and B cells are eliminated in the thymus and bone marrow through a process known as central immune tolerance. However, low-affinity self-reactive T and B cells escape central tolerance and enter the blood and tissues, where they are kept in check by complex and non-redundant peripheral tolerance mechanisms. Dysfunction or imbalance of the immune system can lead to autoimmunity, and thus elucidation of normal tolerance mechanisms has led to identification of therapeutic targets for treating autoimmune disease. In the past 15 years, a number of disease-modifying monoclonal antibodies and genetically engineered biologic agents targeting the immune system have been approved, notably for the treatment of rheumatoid arthritis, inflammatory bowel disease and psoriasis. Although these agents represent a major advance, effective therapy for other autoimmune conditions, such as type 1 diabetes, remain elusive and will likely require intervention aimed at multiple components of the immune system. To this end, approaches that manipulate cells ex vivo and harness their complex behaviors are being tested in preclinical and clinical settings. In addition, approved biologic agents are being examined in combination with one another and with cell-based therapies. Substantial development and regulatory hurdles must be overcome in order to successfully combine immunotherapeutic biologic agents. Nevertheless, such combinations might ultimately be necessary to control autoimmune disease manifestations and restore the tolerant state.
Collapse
Affiliation(s)
- Dawn E Smilek
- The Immune Tolerance Network, 185 Berry Street #3515, San Francisco, CA 94107, USA
| | | | | |
Collapse
|
26
|
Myeloid-derived suppressor cells impair alveolar macrophages through PD-1 receptor ligation during Pneumocystis pneumonia. Infect Immun 2014; 83:572-82. [PMID: 25404033 DOI: 10.1128/iai.02686-14] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) were recently found to accumulate in the lungs during Pneumocystis pneumonia (PcP). Adoptive transfer of these cells caused lung damage in recipient mice, suggesting that MDSC accumulation is a mechanism of pathogenesis in PcP. In this study, the phagocytic activity of alveolar macrophages (AMs) was found to decrease by 40% when they were incubated with MDSCs from Pneumocystis-infected mice compared to those incubated with Gr-1(+) cells from the bone marrow of uninfected mice. The expression of the PU.1 gene in AMs incubated with MDSCs also was decreased. This PU.1 downregulation was due mainly to decreased histone 3 acetylation and increased DNA methylation caused by MDSCs. MDSCs were found to express high levels of PD-L1, and alveolar macrophages (AMs) were found to express high levels of PD-1 during PcP. Furthermore, PD-1 expression in AMs from uninfected mice was increased by 18-fold when they were incubated with MDSCs compared to those incubated with Gr-1(+) cells from the bone marrow of uninfected mice. The adverse effects of MDSCs on AMs were diminished when the MDSCs were pretreated with anti-PD-L1 antibody, suggesting that MDSCs disable AMs through PD-1/PD-L1 ligation during PcP.
Collapse
|
27
|
Autoimmunity versus autoinflammation--friend or foe? Wien Med Wochenschr 2014; 164:274-7. [PMID: 25004809 DOI: 10.1007/s10354-014-0290-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/17/2014] [Indexed: 01/08/2023]
Abstract
"Autoimmunity" is a designation dependent on the conventional immunological issue of self/non-self discrimination. Identification of novel target autoantigens is still an important issue ongoing in classical tissue-specific autoimmune bullous diseases and autoimmune connective tissue diseases. In contrast, synchronized with the paradigm shift of the fundamental aspect of immunity to danger sensing/signaling, distinct collagen-like diseases have been defined by the genetic mutations causing dysregulated innate immunity/inflammation and have been designated as “autoinflammatory”diseases. Due to the clinical and etiological similarities,the concept of autoinflammatory diseases has expanded to include non-hereditary collagen-like diseases, tissue-specific chronic idiopathic inflammatory diseases and metabolic diseases. On the other hand, various genetic causes of autoimmune diseases have been identified and the border of these two pathophysiologies is becoming obscure. Instead, a variable mixture of both autoimmunity and autoinflammation can cause each inflammatory phenotype with a variable level of antigen specificity
Collapse
|
28
|
Alexithymia, mood states and pain experience in systemic lupus erythematosus and rheumatoid arthritis. Clin Rheumatol 2014; 33:1443-50. [PMID: 24718486 DOI: 10.1007/s10067-014-2593-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 02/04/2014] [Accepted: 03/24/2014] [Indexed: 02/03/2023]
Abstract
This prospective study aims to examine alexithymia, mood states and pain experience in systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) patients. We enrolled 49 patients with SLE or RA. All patients were evaluated through a set of questionnaires: (1) the Toronto Alexithymia Scale-20 (TAS), (2) the Profile of Mood States (POMS) and (3) visual analogue scale (VAS) and Questionario Italiano sul Dolore, self-report measures to assess pain intensity. Alexithymia was more prevalent in RA (44 %) than in SLE (37.5 %). The mean values of VAS were significantly higher in RA than in SLE population (p < 0.05). A linear relation between TAS and VAS values has been found in SLE (R = 0.714, p < 0.0001). The mean values of POMS regarding all negative dimensions of mood were higher in SLE than in RA. There was a linear relationship between TAS and POMS values in SLE patients (R = 0.7, p < 0.001). We found a high prevalence of alexithymia in SLE and RA. The chronic pain is influenced by emotional status as documented by a linear relation between TAS and VAS values in SLE patients. The difficulty in reporting emotional responses in these patients seems to be mediated by negative mood states.
Collapse
|
29
|
Cytotoxic-T-lymphocyte antigen 4 receptor signaling for lymphocyte adhesion is mediated by C3G and Rap1. Mol Cell Biol 2014; 34:978-88. [PMID: 24396067 DOI: 10.1128/mcb.01024-13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
T-lymphocyte adhesion plays a critical role in both inflammatory and autoimmune responses. The small GTPase Rap1 is the key coordinator mediating T-cell adhesion to endothelial cells, antigen-presenting cells, and virus-infected cells. We describe a signaling pathway, downstream of the cytotoxic T-lymphocyte antigen 4 (CTLA-4) receptor, leading to Rap1-mediated adhesion. We identified a role for the Rap1 guanine nucleotide exchange factor C3G in the regulation of T-cell adhesion and showed that this factor is required for both T-cell receptor (TCR)-mediated and CTLA-4-mediated T-cell adhesion. Our data indicated that C3G translocates to the plasma membrane downstream of TCR signaling, where it regulates activation of Rap1. We also showed that CTLA-4 receptor signaling mediates tyrosine phosphorylation in the C3G protein, and that this is required for augmented activation of Rap1 and increased adhesion mediated by leukocyte function-associated antigen type 1 (LFA-1). Zap70 is required for C3G translocation to the plasma membrane, whereas the Src family member Hck facilitates C3G phosphorylation. These findings point to C3G and Hck as promising potential therapeutic targets for the treatment of T-cell-dependent autoimmune disorders.
Collapse
|
30
|
Expression regulation of co-inhibitory molecules on human natural killer cells in response to cytokine stimulations. Cytokine 2014; 65:33-41. [DOI: 10.1016/j.cyto.2013.09.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Revised: 09/02/2013] [Accepted: 09/23/2013] [Indexed: 01/22/2023]
|
31
|
TLR2 directing PD-L2 expression inhibit T cells response in Schistosoma japonicum infection. PLoS One 2013; 8:e82480. [PMID: 24376539 PMCID: PMC3869711 DOI: 10.1371/journal.pone.0082480] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 10/23/2013] [Indexed: 01/01/2023] Open
Abstract
Toll-like receptor 2 (TLR2) was shown to be an important immune receptor involved in the recognition of schistosome antigens, especially soluble egg antigen (SEA). In mice models with Schistosoma japonicum acute infection, we observed enhanced T cell-mediated immune responses in TLR2 knock out (TLR2(-/-)) mice compared with B6 mice. In Schistosoma japonicum chronic infection models, programmed death ligand 1 (PD-L1) and programmed death ligand 2 (PD-L2) expression as well as TLR2 expression gradually increased in B6 mice, while only PD-L2 expression significantly decreased in TLR2(-/-) mice. Meanwhile, Programmed Death 1(PD-1) expression on CD4(+)T cells was down-regulated in TLR2(-/-) mice after a large number of egg appeared. We also found that stimulation with schistosome antigens, especially SEA, could up-regulate PD-L2 expression on BMDCs in a TLR2-dependent manner in vitro. Schistosome antigens primed-BMDCs with impaired expression of TLR2 or PD-L2 could induce CD4(+)T cells to produce low level of IL-10 or high level of IFN-γ. Our results indicated that TLR2 signaling can direct PD-L2 expression on DCs, which binds to PD-1 mainly on CD4(+)T cells, to help inhibit T cells response in Schistosoma japonicum infection.
Collapse
|
32
|
Zhernakova A, Withoff S, Wijmenga C. Clinical implications of shared genetics and pathogenesis in autoimmune diseases. Nat Rev Endocrinol 2013; 9:646-59. [PMID: 23959365 DOI: 10.1038/nrendo.2013.161] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Many endocrine diseases, including type 1 diabetes mellitus, Graves disease, Addison disease and Hashimoto disease, originate as an autoimmune reaction that affects disease-specific target organs. These autoimmune diseases are characterized by the development of specific autoantibodies and by the presence of autoreactive T cells. They are caused by a complex genetic predisposition that is attributable to multiple genetic variants, each with a moderate-to-low effect size. Most of the genetic variants associated with a particular autoimmune endocrine disease are shared between other systemic and organ-specific autoimmune and inflammatory diseases, such as rheumatoid arthritis, coeliac disease, systemic lupus erythematosus and psoriasis. Here, we review the shared and specific genetic background of autoimmune diseases, summarize their treatment options and discuss how identifying the genetic and environmental factors that predispose patients to an autoimmune disease can help in the diagnosis and monitoring of patients, as well as the design of new treatments.
Collapse
Affiliation(s)
- Alexandra Zhernakova
- University of Groningen, University Medical Centre Groningen, Department of Genetics, PO Box 30001, 9700 RB Groningen, Netherlands
| | | | | |
Collapse
|
33
|
Gyorki DE, Callahan M, Wolchok JD, Ariyan CE. The delicate balance of melanoma immunotherapy. Clin Transl Immunology 2013; 2:e5. [PMID: 25505953 PMCID: PMC4232053 DOI: 10.1038/cti.2013.5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 06/21/2013] [Accepted: 06/22/2013] [Indexed: 12/31/2022] Open
Abstract
The strategy of immune modulation for the treatment of cancer is being refined with the introduction of multiple new therapeutic agents into the clinic. Melanoma is a disease where many of these agents have demonstrated efficacy. The mechanisms of action of these agents exploit the counter-regulatory mechanisms of the immune response. However, these agents are also associated with immune-related adverse events (IRAEs), which represent tissue-specific inflammatory responses. These IRAEs highlight the delicate balance of immunologic homeostasis and, with some interventions, may occur more frequently in patients who sustain a therapeutic response. This review will discuss melanoma immunogenicity and immunotherapy. Furthermore, the spectrum and distinction between a reversible immune adverse event and autoimmunity will be highlighted.
Collapse
Affiliation(s)
- David E Gyorki
- Memorial Sloan-Kettering Cancer Center , New York, NY, USA
| | - Margaret Callahan
- Memorial Sloan-Kettering Cancer Center , New York, NY, USA ; Ludwig Center, Memorial Sloan-Kettering Cancer Center , New York, NY, USA
| | - Jedd D Wolchok
- Memorial Sloan-Kettering Cancer Center , New York, NY, USA ; Ludwig Center, Memorial Sloan-Kettering Cancer Center , New York, NY, USA ; Weill Cornell Medical College , New York, NY, USA
| | - Charlotte E Ariyan
- Memorial Sloan-Kettering Cancer Center , New York, NY, USA ; Weill Cornell Medical College , New York, NY, USA
| |
Collapse
|
34
|
Romo-Tena J, Gómez-Martín D, Alcocer-Varela J. CTLA-4 and autoimmunity: new insights into the dual regulator of tolerance. Autoimmun Rev 2013; 12:1171-6. [PMID: 23851140 DOI: 10.1016/j.autrev.2013.07.002] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 07/07/2013] [Indexed: 12/23/2022]
Abstract
Cytotoxic T-Lymphocye Antigen 4 (CTLA-4) or CD152 is an inhibitory molecule that plays a critical role in maintenance of tolerance to self-antigens. CTLA-4 is structurally as well as functionally related to CD28, since it shares 31% of homology and binds the B7 family molecules CD80 and CD86 with higher affinity. Nevertheless, CTLA-4 has opposing effects on T cell activation and current evidence shows that its inhibitory role goes beyond the ligand-binding interaction. CTLA-4 competes with CD28 in binding to B7, interacts within the immunological synapsis elements and with clathrin adaptor proteins and tyrosine phosphatases through its cytoplasmic domain to regulate cell trafficking and to set the activation threshold within T cells. Moreover, we have learned from the knock out model that CTLA-4 plays a key role in regulatory T cells and in central tolerance. Because of its importance in maintenance of peripheral tolerance, CTLA-4 has been implicated in several autoimmune diseases, such as systemic lupus erythematosus. Multiple single-nucleotide polymorphisms have been located to human Ctla-4 gene, and their association with autoimmune disease is still a matter of controversy. Despite the promising results of abatacept or CTLA-4-Ig in rheumatoid arthritis and murine lupus nephritis, more clinical randomized trials and standardization of outcomes are needed to prove its efficacy and safety in human lupus nephritis.
Collapse
Affiliation(s)
- Jorge Romo-Tena
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Tlalpan 14000, Mexico City, Mexico
| | | | | |
Collapse
|
35
|
Aigner J, Villatoro S, Rabionet R, Roquer J, Jiménez-Conde J, Martí E, Estivill X. A common 56-kilobase deletion in a primate-specific segmental duplication creates a novel butyrophilin-like protein. BMC Genet 2013; 14:61. [PMID: 23829304 PMCID: PMC3729544 DOI: 10.1186/1471-2156-14-61] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 05/21/2013] [Indexed: 12/22/2022] Open
Abstract
Background The Butyrophilin-like (BTNL) proteins are likely to play an important role in inflammation and immune response. Like the B7 protein family, many human and murine BTNL members have been shown to control T lymphocytes response, and polymorphisms in human BTNL2 have been linked to several inflammatory diseases, such as pulmonary sarcoidosis, inflammatory bowel disease and neonatal lupus. Results In this study we provide a comprehensive population, genomic and transcriptomic analysis of a 56-kb deletion copy number variant (CNV), located within two segmental duplications of two genes belonging to the BTNL family, namely BTNL8 and BTNL3. We confirm the presence of a novel BTNL8*3 fusion-protein product, and show an influence of the deletion variant on the expression level of several genes involved in immune function, including BTNL9, another member of the same family. Moreover, by genotyping HapMap and human diversity panel (HGDP) samples, we demonstrate a clear difference in the stratification of the BTNL8_BTNL3-del allele frequency between major continental human populations. Conclusion Despite tremendous progress in the field of structural variation, rather few CNVs have been functionally characterized so far. Here, we show clear functional consequences of a new deletion CNV (BTNL8_BTNL3-del) with potentially important implication in the human immune system and in inflammatory and proliferative disorders. In addition, the marked population differences found of BTNL8_BTNL3-del frequencies suggest that this deletion CNV might have evolved under positive selection due to environmental conditions in some populations, with potential phenotypic consequences.
Collapse
Affiliation(s)
- Johanna Aigner
- Bioinformatics and Genomics Program, Centre for Genomic Regulation (CRG), Barcelona 08003, Spain
| | | | | | | | | | | | | |
Collapse
|
36
|
Mocellin S, Benna C, Pilati P. Coinhibitory molecules in cancer biology and therapy. Cytokine Growth Factor Rev 2013; 24:147-61. [PMID: 23380546 DOI: 10.1016/j.cytogfr.2013.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Accepted: 01/09/2013] [Indexed: 12/31/2022]
Abstract
The adaptive immune response is controlled by checkpoints represented by coinhibitory molecules, which are crucial for maintaining self-tolerance and minimizing collateral tissue damage under physiological conditions. A growing body of preclinical evidence supports the hypothesis that unleashing this immunological break might be therapeutically beneficial in the fight against cancer, as it would elicit an effective antitumor immune response. Remarkably, recent clinical trials have demonstrated that this novel strategy can be highly effective in the treatment of patients with cancer, as shown by the paradigmatic case of ipilimumab (a monoclonal antibody blocking the coinhibitory molecule cytotoxic T lymphocyte associated antigen-4 [CTLA4]) that is opening a new era in the therapeutic approach to a chemoresistant tumor such as cutaneous melanoma. In this review we summarize the biology of coinhibitory molecules, overview the experimental and clinical attempts to interfere with these immune checkpoints to treat cancer and critically discuss the challenges posed by such a promising antitumor modality.
Collapse
Affiliation(s)
- Simone Mocellin
- Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, 35128 Padova, Italy.
| | | | | |
Collapse
|