1
|
Huang S, Liu D, Han L, Deng J, Wang Z, Jiang J, Zeng L. Decoding the potential role of regulatory T cells in sepsis-induced immunosuppression. Eur J Immunol 2024; 54:e2350730. [PMID: 38430202 DOI: 10.1002/eji.202350730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 03/03/2024]
Abstract
Sepsis, a multiorgan dysfunction with high incidence and mortality, is caused by an imbalanced host-to-infection immune response. Organ-support therapy improves the early survival rate of sepsis patients. In the long term, those who survive the "cytokine storm" and its secondary damage usually show higher susceptibility to secondary infections and sepsis-induced immunosuppression, in which regulatory T cells (Tregs) are evidenced to play an essential role. However, the potential role and mechanism of Tregs in sepsis-induced immunosuppression remains elusive. In this review, we elucidate the role of different functional subpopulations of Tregs during sepsis and then review the mechanism of sepsis-induced immunosuppression from the aspects of regulatory characteristics, epigenetic modification, and immunometabolism of Tregs. Thoroughly understanding how Tregs impact the immune system during sepsis may shed light on preclinical research and help improve the translational value of sepsis immunotherapy.
Collapse
Affiliation(s)
- Siyuan Huang
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China
| | - Di Liu
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China
| | - Lei Han
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China
| | - Jin Deng
- Department of Emergency, the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Zhen Wang
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China
| | - Jianxin Jiang
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China
| | - Ling Zeng
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China
| |
Collapse
|
2
|
Nieves-Rosado HM, Jacobs JL, Naqvi A, Mellors JW, Macatangay BJC, Kane LP. TIM-3 signaling contributes to the suppressive capacity of Tregs from people with HIV on antiretroviral therapy. J Leukoc Biol 2023; 114:368-372. [PMID: 37350502 PMCID: PMC10882646 DOI: 10.1093/jleuko/qiad068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 06/24/2023] Open
Abstract
TIM-3 expression is increased on peripheral regulatory T cells (Tregs) of virally suppressed persons with HIV-1 on antiretroviral therapy (PWH-ART). However, the relevance of TIM-3 expression in this setting is unclear. We used flow cytometry to evaluate the suppressive phenotype and signaling pathways in peripheral TIM-3- vs TIM-3+ Tregs in PWH-ART. TIM-3+ Tregs showed increased expression of IL-10 compared with persons without HIV-1. In addition, TIM-3+ Tregs displayed elevated signaling and activation, relative to TIM-3- Tregs from the same PWH-ART. Dramatically, TIM-3 blockade restrained the in vitro suppressive capacity of peripheral Tregs. Therefore, our data demonstrate not only that TIM-3 expression by Tregs is associated with an immunosuppressive response among PWH-ART, but also that TIM-3 contributes directly to the enhanced suppressive activity of Tregs in this setting.
Collapse
Affiliation(s)
- Hector M Nieves-Rosado
- Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, United States
| | - Jana L Jacobs
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, United States
| | - Asma Naqvi
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, United States
| | - John W Mellors
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, United States
| | - Bernard J C Macatangay
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, United States
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, United States
| |
Collapse
|
3
|
Tim-3 blockade enhances the clearance of Chlamydia psittaci in the lung by promoting a cell-mediated immune response. Int Immunopharmacol 2023; 116:109780. [PMID: 36720194 DOI: 10.1016/j.intimp.2023.109780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/10/2023] [Accepted: 01/20/2023] [Indexed: 01/30/2023]
Abstract
Chlamydia psittaci is remarkable at disrupting immunity and thus poses a great risk to the animal industry and public health. Immune inhibitory molecule upregulation and the accumulation of specialized cells play key roles in chlamydial clearance. It is clear that the T-cell immunoglobulin and mucin domain protein 3 receptor (Tim-3) can regulate effector T cells in infectious disease. However, the immunomodulatory effect of Tim-3 in C. psittaci infection remains unknown. Thus, the expression of Tim-3 in effector T cells and its immune regulatory function during C. psittaci infection were investigated. The level of Tim-3 on CD4+ and CD8+ T cells was meaningfully higher in C. psittaci-infected mice. Blockade of Tim-3 signaling by anti-Tim-3 antibody showed accelerated C. psittaci clearance and less pathological changes in the lung than isotype immunoglobulin treatment. Furthermore, treatment with anti-Tim-3 antibody greatly enhanced the levels of IFN-γ and interleukin (IL)-22/IL-17, which were correlated with an improved Th1- and Th17-mediated immune response, and decreased IL-10, which were related with a decreased Treg immune response. In conclusion, Tim-3 expression in effector T cells negatively regulates Th1 and Th17 immune responses against C. psittaci respiratory infection.
Collapse
|
4
|
Chen L, Yu X, Lv C, Dai Y, Wang T, Zheng S, Qin Y, Zhou X, Wang Y, Pei H, Fang H, Huang B. Increase in Serum Soluble Tim-3 Level Is Related to the Progression of Diseases After Hepatitis Virus Infection. Front Med (Lausanne) 2022; 9:880909. [PMID: 35646962 PMCID: PMC9133670 DOI: 10.3389/fmed.2022.880909] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundViral hepatitis is a widespread and serious infectious disease, and most patients with liver cirrhosis and hepatocellular carcinoma are prone to viral infections. T cell immunoglobulin-and mucin-domain-containing molecule-3 (Tim-3) is an immune checkpoint molecule that negatively regulates T cell responses, playing an extremely important role in controlling infectious diseases. However, reports about the role of serum soluble Tim-3 (sTim-3) in hepatitis virus infection are limited. Therefore, this study explored changes in sTim-3 levels in patients infected with hepatitis B virus (HBV), hepatitis C virus (HCV), and hepatitis E virus (HEV).MethodsThis study applied high-sensitivity time-resolved fluorescence immunoassay for the detection of sTim-3 levels. A total of 205 cases of viral hepatitis infection (68 cases of HBV infection, 60 cases of HCV infection, and 77 cases of HEV virus infection) and 88 healthy controls were quantitatively determined. The changes in serum sTim-3 level and its clinical value in hepatitis virus infection were analyzed.ResultsPatients with HBV infection (14.00, 10.78–20.45 ng/mL), HCV infection (15.99, 11.83–27.00 ng/mL), or HEV infection (19.09, 10.85–33.93 ng/mL) had significantly higher sTim-3 levels than that in the healthy control group (7.69, 6.14–10.22 ng/mL, P < 0.0001). Patients with hepatitis and fibrosis infected with HBV (22.76, 12.82–37.53 ng/mL), HCV (33.06, 16.36–39.30 ng/mL), and HEV (28.90, 17.95–35.94 ng/mL) had significantly higher sTim-3 levels than patients with hepatitis without fibrosis (13.29, 7.75–17.28; 13.86, 11.48–18.64; 14.77, 9.79–29.79 ng/mL; P < 0.05).ConclusionsTim-3 level was elevated in patients infected with HBV, HCV, or HEV and gradually increased in patients with either hepatitis or hepatitis with hepatic fibrosis. It has a certain role in the evaluation of the course of a disease after hepatitis virus infection.
Collapse
Affiliation(s)
- Lingli Chen
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiaomei Yu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Chunyan Lv
- Wuxi No.5 People’s Hospital, Wuxi, China
| | - Yaping Dai
- Wuxi No.5 People’s Hospital, Wuxi, China
| | - Tao Wang
- Wuxi No.5 People’s Hospital, Wuxi, China
| | - Shaoxiong Zheng
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yuan Qin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiumei Zhou
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yigang Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Hao Pei
- Wuxi No.5 People’s Hospital, Wuxi, China
- Hao Pei,
| | - Hongming Fang
- Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
- Hongming Fang,
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
- *Correspondence: Biao Huang,
| |
Collapse
|
5
|
Nguyen LN, Nguyen LNT, Zhao J, Schank M, Dang X, Cao D, Khanal S, Thakuri BKC, Zhang J, Lu Z, Wu XY, El Gazzar M, Ning S, Wang L, Moorman JP, Yao ZQ. Immune Activation Induces Telomeric DNA Damage and Promotes Short-Lived Effector T Cell Differentiation in Chronic HCV Infection. Hepatology 2021; 74:2380-2394. [PMID: 34110660 PMCID: PMC8542603 DOI: 10.1002/hep.32008] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/10/2021] [Accepted: 06/01/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Hepatitis C virus (HCV) leads to a high rate of chronic infection and T cell dysfunction. Although it is well known that chronic antigenic stimulation is a driving force for impaired T cell functions, the precise mechanisms underlying immune activation-induced T cell dysfunctions during HCV infection remain elusive. APPROACH AND RESULTS Here, we demonstrated that circulating CD4+ T cells from patients who are chronically HCV-infected exhibit an immune activation status, as evidenced by the overexpression of cell activation markers human leukocyte antigen-antigen D-related, glucose transporter 1, granzyme B, and the short-lived effector marker CD127- killer cell lectin-like receptor G1+ . In contrast, the expression of stem cell-like transcription factor T cell factor 1 and telomeric repeat-binding factor 2 (TRF2) are significantly reduced in CD4+ T cells from patients who are chronically HCV-infected compared with healthy participants (HP). Mechanistic studies revealed that CD4+ T cells from participants with HCV exhibit phosphoinositide 3-kinase/Akt/mammalian target of rapamycin signaling hyperactivation on T cell receptor stimulation, promoting proinflammatory effector cell differentiation, telomeric DNA damage, and cellular apoptosis. Inhibition of Akt signaling during T cell activation preserved the precursor memory cell population and prevented inflammatory effector cell expansion, DNA damage, and apoptotic death. Moreover, knockdown of TRF2 reduced HP T cell stemness and triggered telomeric DNA damage and cellular apoptosis, whereas overexpression of TRF2 in CD4 T cells prevented telomeric DNA damage. CONCLUSIONS These results suggest that modulation of immune activation through inhibiting Akt signaling and protecting telomeres through enhancing TRF2 expression may open therapeutic strategies to fine tune the adaptive immune responses in the setting of persistent immune activation and inflammation during chronic HCV infection.
Collapse
Affiliation(s)
- Lam Nhat Nguyen
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN
| | - Lam Ngoc Thao Nguyen
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN
| | - Juan Zhao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN
| | - Madison Schank
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN
| | - Xindi Dang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN
| | - Dechao Cao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN
| | - Sushant Khanal
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN
| | - Bal Krishna Chand Thakuri
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN
| | - Jinyu Zhang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN
| | - Zeyuan Lu
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN
| | - Xiao Y Wu
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN
| | - Mohamed El Gazzar
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN
| | - Shunbin Ning
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN
| | - Ling Wang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN
| | - Jonathan P Moorman
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN.,Hepatitis (HCV/HBV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, TN
| | - Zhi Q Yao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN.,Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN.,Hepatitis (HCV/HBV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, TN
| |
Collapse
|
6
|
Modabber Z, Shahbazi M, Akbari R, Bagherzadeh M, Firouzjahi A, Mohammadnia-Afrouzi M. TIM-3 as a potential exhaustion marker in CD4 + T cells of COVID-19 patients. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:1707-1715. [PMID: 34499819 PMCID: PMC8589347 DOI: 10.1002/iid3.526] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 12/24/2022]
Abstract
Background COVID‐19 causes a range of clinical symptoms from mild to critical and can be life‐threatening. Up to now, it has led to many deaths. We aimed to evaluate exhausted markers on CD4+ T cells of COVID‐19 patients. Methods In this study, we evaluated 44 patients with confirmed COVID‐19 disease and 16 healthy individuals. Patients were divided into moderate/severe and critical groups. Peripheral blood mononuclear cells (PBMCs) were isolated and stained by anti‐human CD39, PD‐1, TIM‐3, and anti‐human CD4. The percentage of each CD4+ subpopulation was calculated by flow cytometry. Furthermore, we collected clinical information and laboratory data of both control and patient groups. Results We detected overexpression of TIM‐3 on CD4+ T cells in both critical and moderate/severe patients than in healthy individuals (HIs; p < .01 and p < .0001, respectively). CD4+ TIM‐3+ CD39+ lymphocytes were significantly higher in the critical patients than in HI (p < .05). Both Patient groups showed lymphopenia in comparison with HI, but CD4+ lymphocytes did not show any significant difference between study subjects. The increased amount of C‐reactive protein, erythrocyte sedimentation rate, creatinine, blood urea nitrogen, and neutrophil count was observed in patients compared to HI. Conclusion T cell exhaustion occurs during COVID‐19 disease and TIM‐3 is the most important exhausted marker on CD4+ T cells.
Collapse
Affiliation(s)
- Zahra Modabber
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran.,Department of Pathology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mehdi Shahbazi
- Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Roghayeh Akbari
- Department of Internal Medicine, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mojgan Bagherzadeh
- Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Alireza Firouzjahi
- Department of Pathology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mousa Mohammadnia-Afrouzi
- Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
7
|
The TIM3/Gal9 signaling pathway: An emerging target for cancer immunotherapy. Cancer Lett 2021; 510:67-78. [PMID: 33895262 DOI: 10.1016/j.canlet.2021.04.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 03/31/2021] [Accepted: 04/15/2021] [Indexed: 12/20/2022]
Abstract
Immune checkpoint blockade has shown unprecedented and durable clinical response in a wide range of cancers. T cell immunoglobulin and mucin domain 3 (TIM3) is an inhibitory checkpoint protein that is highly expressed in tumor-infiltrating lymphocytes. In various cancers, the interaction of TIM3 and Galectin 9 (Gal9) suppresses anti-tumor immunity mediated by innate as well as adaptive immune cells. Thus, the blockade of the TIM3/Gal9 interaction is a promising therapeutic approach for cancer therapy. In addition, co-blockade of the TIM3/Gal9 pathway along with the PD-1/PD-L1 pathway increases the therapeutic efficacy by overcoming non-redundant immune resistance induced by each checkpoint. Here, we summarize the physiological roles of the TIM3/Gal9 pathway in adaptive and innate immune systems. We highlight the recent clinical and preclinical studies showing the involvement of the TIM3/Gal9 pathway in various solid and blood cancers. In addition, we discuss the potential of using TIM3 and Gal9 as prognostic and predictive biomarkers in different cancers. An in-depth mechanistic understanding of the blockade of the TIM3/Gal9 signaling pathway in cancer could help in identifying patients who respond to this therapy as well as designing combination therapies.
Collapse
|
8
|
Boehne C, Behrendt AK, Meyer-Bahlburg A, Boettcher M, Drube S, Kamradt T, Hansen G. Tim-3 is dispensable for allergic inflammation and respiratory tolerance in experimental asthma. PLoS One 2021; 16:e0249605. [PMID: 33822811 PMCID: PMC8023500 DOI: 10.1371/journal.pone.0249605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/19/2021] [Indexed: 11/18/2022] Open
Abstract
T cell immunoglobulin and mucin domain-containing molecule-3 (Tim-3) has been described as a transmembrane protein, expressed on the surface of various T cells as well as different cells of innate immunity. It has since been associated with Th1 mediated autoimmune diseases and transplantation tolerance studies, thereby indicating a possible role of this receptor in counter-regulation of Th2 immune responses. In the present study we therefore directly examined the role of Tim-3 in allergic inflammation and respiratory tolerance. First, Tim-3-/- mice and wild type controls were immunized and challenged with the model allergen ovalbumin (OVA) to induce an asthma-like phenotype. Analysis of cell numbers and distribution in the bronchoalveolar lavage (BAL) fluid as well as lung histology in H&E stained lung sections demonstrated a comparable degree of eosinophilic inflammation in both mouse strains. Th2 cytokine production in restimulated cell culture supernatants and serum IgE and IgG levels were equally increased in both genotypes. In addition, cell proliferation and the distribution of different T cell subsets were comparable. Moreover, analysis of both mouse strains in our respiratory tolerance model, where mucosal application of the model allergen before immunization, prevents the development of an asthma-like phenotype, revealed no differences in any of the parameters mentioned above. The current study demonstrates that Tim-3 is dispensable not only for the development of allergic inflammation but also for induction of respiratory tolerance in mice in an OVA-based model.
Collapse
Affiliation(s)
- Carolin Boehne
- Department of Pediatrics and Adolescent Medicine, Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Lower Saxony, Germany
| | - Ann-Kathrin Behrendt
- Department of Pediatrics and Adolescent Medicine, Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Lower Saxony, Germany
| | - Almut Meyer-Bahlburg
- Department of Pediatrics and Adolescent Medicine, Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Lower Saxony, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Lower Saxony, Germany
| | - Martin Boettcher
- Institute of Immunology, University Hospital Jena, Jena, Thuringia, Germany
| | - Sebastian Drube
- Institute of Immunology, University Hospital Jena, Jena, Thuringia, Germany
| | - Thomas Kamradt
- Institute of Immunology, University Hospital Jena, Jena, Thuringia, Germany
| | - Gesine Hansen
- Department of Pediatrics and Adolescent Medicine, Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Lower Saxony, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Lower Saxony, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Lower Saxony, Germany
- * E-mail:
| |
Collapse
|
9
|
van der Zwan A, van Unen V, Beyrend G, Laban S, van der Keur C, Kapsenberg HJM, Höllt T, Chuva de Sousa Lopes SM, van der Hoorn MLP, Koning F, Claas FHJ, Eikmans M, Heidt S. Visualizing Dynamic Changes at the Maternal-Fetal Interface Throughout Human Pregnancy by Mass Cytometry. Front Immunol 2020; 11:571300. [PMID: 33193353 PMCID: PMC7649376 DOI: 10.3389/fimmu.2020.571300] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/02/2020] [Indexed: 12/14/2022] Open
Abstract
During healthy pregnancy, a balanced microenvironment at the maternal-fetal interface with coordinated interaction between various immune cells is necessary to maintain immunological tolerance. While specific decidual immune cell subsets have been investigated, a system-wide unbiased approach is lacking. Here, mass cytometry was applied for data-driven, in-depth immune profiling of the total leukocyte population isolated from first, second, and third trimester decidua, as well as maternal peripheral blood at time of delivery. The maternal-fetal interface showed a unique composition of immune cells, different from peripheral blood, with significant differences between early and term pregnancy samples. Profiling revealed substantial heterogeneity in the decidual lymphoid and myeloid cell lineages that shape gestational-specific immune networks and putative differentiation trajectories over time during gestation. Uncovering the overall complexity at the maternal-fetal interface throughout pregnancy resulted in a human atlas that may serve as a foundation upon which comprehension of the immune microenvironment and alterations thereof in pregnancy complications can be built.
Collapse
Affiliation(s)
- Anita van der Zwan
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Vincent van Unen
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Guillaume Beyrend
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Sandra Laban
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Carin van der Keur
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Thomas Höllt
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, Netherlands
- Computer Graphics and Visualization Group, Delft University of Technology, Delft, Netherlands
| | | | | | - Frits Koning
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Frans H. J. Claas
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Michael Eikmans
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
10
|
Immune Checkpoint Inhibitors as Monotherapy or Within a Combinatorial Strategy in Advanced Hepatocellular Carcinoma. Int J Mol Sci 2020; 21:ijms21176302. [PMID: 32878115 PMCID: PMC7504231 DOI: 10.3390/ijms21176302] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/13/2022] Open
Abstract
In advanced-stage hepatocellular carcinoma (HCC), systemic treatment represents the standard therapy. Target therapy has marked a new era based on a greater knowledge of molecular disease signaling. Nonetheless, survival outcomes and long-term response remain unsatisfactory, mostly because of the onset of primary or acquired resistance. More recently, results from clinical trials with immune targeting agents, such as the immune checkpoint inhibitors (ICIs), have shown a promising role for these drugs in the treatment of advanced HCC. In the context of an intrinsic tolerogenic liver environment, since HCC-induced immune tolerance, it is supported by multiple immunosuppressive mechanisms and several clinical trials are now underway to evaluate ICI-based combinations, including their associations with antiangiogenic agents or multikinase kinase inhibitors and multiple ICIs combinations. In this review, we will first discuss the basic principles of hepatic immunogenic tolerance and the evasive mechanism of antitumor immunity in HCC; furthermore we will elucidate the consistent biological rationale for immunotherapy in HCC even in the presence of an intrinsic tolerogenic environment. Subsequently, we will critically report and discuss current literature on ICIs in the treatment of advanced HCC, including a focus on the currently explored combinatorial strategies and their rationales. Finally, we will consider both challenges and future directions in this field.
Collapse
|
11
|
Guo H, Shen Y, Kong YH, Li S, Jiang R, Liu C, Fang C, Hu J. The expression of Tim-1 and Tim-4 molecules in regulatory T cells in type 1 diabetes. Endocrine 2020; 68:64-70. [PMID: 31916216 DOI: 10.1007/s12020-019-02173-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 12/23/2019] [Indexed: 02/05/2023]
Abstract
PURPOSE The TIM family comprises of eight genes in the mouse, three of which are conserved in humans (TIM-1, TIM-3, and TIM-4). Previous studies have revealed the relationships between Tim3+ Tregs and autoimmune disease. There was little study about the expression of Tim1 and Tim4 surface molecules on Tregs. We evaluated the frequency of the Tim1+Tregs and Tim4+Tregs in type 1 diabetes (T1D) in the present study. METHODS A total of 28 patients with T1D and 14 gender-, age-, and ethnically matched healthy volunteers were recruited. PBMCs from these individuals were isolated and analyzed by flow cytometry. Splenocytes from mice were also analyzed by flow cytometry. RESULTS There is no difference in the frequency of Treg cells in peripheral blood isolated from T1D patients. Tim1 on CD4+CD25+ T cells decreased significantly in PBMC of patients with T1D(1.19 ± 0.17% vs 2.78 ± 0.38%, 95% CI:0.87-2.31, P < 0.0001), while expression of Tim4 on CD4+CD25+ T cells in PBMC was less frequent in patients with T1D than healthy people(3.0 ± 0.39% vs 6.25 ± 1.08%, 95% CI:1.08-5.43, P = 0.0044). The frequencies of CD4+CD25+Tim1+ T cells and CD4+CD25+Tim4+ T cells also decreased in spleen of hyperglycemic NOD mice. There were no significant correlations between CD4+CD25+Tim1+T-cells, CD4+CD25+Tim4+T-cells and any clinical features such as age, HbA1c, Fasting C-peptide, diabetic autoantibodies, disease duration, total cholesterol, LDL, HDL, and TG. CONCLUSIONS It is the first report of the expression of Tim1 and Tim4 molecules on Treg cells in T1D in the present study. We also presented evidence that the frequencies of Tim1+Tregs and Tim4+Tregs decreased significantly in both type 1 diabetic patients and hyperglycemic NOD mice. However, the specific functions of Tim1+Tregs and Tim4+Tregs are still unclear.
Collapse
Affiliation(s)
- Heming Guo
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu, China
| | - Yingxiao Shen
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, Jiangsu, China
| | - Ying-Hong Kong
- Department of Endocrinology, Changshu No. 2 People's Hospital, Changshu, 215500, Jiangsu, China
| | - Sicheng Li
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu, China
| | - Rong Jiang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu, China
| | - Cuiping Liu
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, 215002, Jiangsu, China
| | - Chen Fang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu, China.
| | - Ji Hu
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu, China.
| |
Collapse
|
12
|
Szereday L, Meggyes M, Berki T, Miseta A, Farkas N, Gervain J, Par A, Par G. Direct-acting antiviral treatment downregulates immune checkpoint inhibitor expression in patients with chronic hepatitis C. Clin Exp Med 2020; 20:219-230. [PMID: 32108916 PMCID: PMC7181552 DOI: 10.1007/s10238-020-00618-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/18/2020] [Indexed: 12/13/2022]
Abstract
Chronic hepatitis C (CHC) infection is associated with increased TIM-3, PD-1 immune checkpoint receptors expression that inhibits adaptive T cells and increases NK cell cytotoxicity against T helper cells, both resulting T cell exhaustion. Elimination of the virus with direct-acting antivirals (DAAs) may modify host immune response via altering these immune checkpoint receptors’ expression. We conducted a prospective study to analyze changes in TIM-3, PD-1 and their ligands galectin-9, PD-L1 expression by peripheral blood T cell subpopulations, NK cell subpopulations, and monocytes by multicolor flow cytometry in 14 CHC patients successfully treated with 12 weeks of dasabuvir, ombitasvir, and paritaprevir/ritonavir plus ribavirin. Blood samples were collected before, at the end of treatment, and 12 and 24 weeks later. Sustained virological response (SVR) was associated with increased percentage of peripheral blood CD3+ T and CD8+ cytotoxic T lymphocytes and decreased percentage of NKbright cells. After DAA treatment, decreased TIM-3 expression by CD4+ T cells, by NKbright, and by NKT cells was found. Expression of immune checkpoint molecules’ ligand PD-L1 by NK cells and by regulatory T cells and galectin-9 by NK cells and monocytes also decreased significantly at SVR. Our data suggest that DAA treatment not only inhibits viral replication but may alter host adaptive and innate immune responses. A decrease in immune checkpoint molecules and their ligands expression both on adaptive and on innate immune cells may contribute to the recovery of exhausted adaptive immune responses and to sustained virological response.
Collapse
Affiliation(s)
- Laszlo Szereday
- Department of Medical Microbiology and Immunology, University of Pecs, Medical School, 12 Szigeti Street, Pecs, 7624, Hungary. .,Janos Szentagothai Research Centre, Pecs, Hungary.
| | - Matyas Meggyes
- Department of Medical Microbiology and Immunology, University of Pecs, Medical School, 12 Szigeti Street, Pecs, 7624, Hungary.,Janos Szentagothai Research Centre, Pecs, Hungary
| | - Timea Berki
- Department of Biotechnology and Immunology, University of Pecs, Medical School, Pecs, Hungary
| | - Attila Miseta
- Department of Laboratory Medicine, University of Pecs, Medical School, Pecs, Hungary
| | - Nelli Farkas
- Institute for Translational Medicine, University of Pecs, Medical School, Pecs, Hungary
| | - Judit Gervain
- County Hospital Fejér, Szent György Hospital, Szekesfehervar, Hungary
| | - Alajos Par
- Division of Gastroenterology, First Department of Medicine, University of Pecs, Medical School, Pecs, Hungary
| | - Gabriella Par
- Institute for Translational Medicine, University of Pecs, Medical School, Pecs, Hungary.,Division of Gastroenterology, First Department of Medicine, University of Pecs, Medical School, Pecs, Hungary
| |
Collapse
|
13
|
Zhao L, Yu G, Han Q, Cui C, Zhang B. TIM-3: An emerging target in the liver diseases. Scand J Immunol 2020; 91:e12825. [PMID: 31486085 DOI: 10.1111/sji.12825] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/27/2019] [Accepted: 08/31/2019] [Indexed: 12/17/2022]
Abstract
T cell immunoglobulin domain and mucin domain-containing molecule 3 (TIM-3) is found expression in the surface of terminally differentiated T cells and belongs to the TIM family of type Ⅰ transmembrane proteins. It binds to the ligand Galectin-9 and mediates T cell apoptosis. As the research progresses, TIM-3 is also expressed in Th17, NK, monocyte, which binds to ligand and induce immune peripheral tolerance in both mice and man. Numerous researches have demonstrated that TIM-3 influences liver diseases, including liver-associated chronic viral infection, liver fibrosis, liver cancer et al and suggest new approaches to intervention. Currently, targeted therapy of TIM-3 is a new treatment in the field of immunization. Although many studies have proven that TIM-3 has an inhibitory effect in vivo, the specific mechanism is not clear. Herein, we summarize the important role of TIM-3 in the regulation of liver disease and prospects for future clinical research. TIM-3 will provide new targets for improving clinical liver disease.
Collapse
Affiliation(s)
- Lizhen Zhao
- Department of Immunology, Medical College of Qingdao University, Qingdao, China
| | - Guoyi Yu
- Editorial Office of Journal of Qingdao University (Medical Science), Qingdao, China
| | - Qi Han
- Department of Immunology, Medical College of Qingdao University, Qingdao, China
| | - Congxian Cui
- Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Bei Zhang
- Department of Immunology, Medical College of Qingdao University, Qingdao, China
| |
Collapse
|
14
|
Ferrara R, Mezquita L, Texier M, Lahmar J, Audigier-Valette C, Tessonnier L, Mazieres J, Zalcman G, Brosseau S, Le Moulec S, Leroy L, Duchemann B, Lefebvre C, Veillon R, Westeel V, Koscielny S, Champiat S, Ferté C, Planchard D, Remon J, Boucher ME, Gazzah A, Adam J, Bria E, Tortora G, Soria JC, Besse B, Caramella C. Hyperprogressive Disease in Patients With Advanced Non-Small Cell Lung Cancer Treated With PD-1/PD-L1 Inhibitors or With Single-Agent Chemotherapy. JAMA Oncol 2019; 4:1543-1552. [PMID: 30193240 DOI: 10.1001/jamaoncol.2018.3676] [Citation(s) in RCA: 519] [Impact Index Per Article: 103.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Importance Hyperprogressive disease (HPD) is a new pattern of progression recently described in patients with cancer treated with programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1) inhibitors. The rate and outcome of HPD in advanced non-small cell lung cancer (NSCLC) are unknown. Objectives To investigate whether HPD is observed in patients with advanced NSCLC treated with PD-1/PD-L1 inhibitors compared with single-agent chemotherapy and whether there is an association between treatment and HPD. Design, Setting, and Participants In this multicenter retrospective study that included patients treated between August 4, 2011, and April 5, 2017, the setting was pretreated patients with advanced NSCLC who received PD-1/PD-L1 inhibitors (8 institutions) or single-agent chemotherapy (4 institutions) in France. Measurable disease defined by Response Evaluation Criteria in Solid Tumors (RECIST version 1.1) on at least 2 computed tomographic scans before treatment and 1 computed tomographic scan during treatment was required. Interventions The tumor growth rate (TGR) before and during treatment and variation per month (ΔTGR) were calculated. Hyperprogressive disease was defined as disease progression at the first evaluation with ΔTGR exceeding 50%. Main Outcomes and Measures The primary end point was assessment of the HPD rate in patients treated with IO or chemotherapy. Results Among 406 eligible patients treated with PD-1/PD-L1 inhibitors (63.8% male), 46.3% (n = 188) were 65 years or older, 72.4% (n = 294) had nonsquamous histology, and 92.9% (n = 377) received a PD-1 inhibitor as monotherapy in second-line therapy or later. The median follow-up was 12.1 months (95% CI, 10.1-13.8 months), and the median overall survival (OS) was 13.4 months (95% CI, 10.2-17.0 months). Fifty-six patients (13.8%) were classified as having HPD. Pseudoprogression was observed in 4.7% (n = 19) of the population. Hyperprogressive disease was significantly associated with more than 2 metastatic sites before PD-1/PD-L1 inhibitors compared with non-HPD (62.5% [35 of 56] vs 42.6% [149 of 350]; P = .006). Patients experiencing HPD within the first 6 weeks of PD-1/PD-L1 inhibitor treatment had significantly lower OS compared with patients with progressive disease (median OS, 3.4 months [95% CI, 2.8-7.5 months] vs 6.2 months [95% CI, 5.3-7.9 months]; hazard ratio, 2.18 [95% CI, 1.29-3.69]; P = .003). Among 59 eligible patients treated with chemotherapy, 3 (5.1%) were classified as having HPD. Conclusions and Relevance Our study suggests that HPD is more common with PD-1/PD-L1 inhibitors compared with chemotherapy in pretreated patients with NSCLC and is also associated with high metastatic burden and poor prognosis in patients treated with PD-1/PD-L1 inhibitors. Additional studies are needed to determine the molecular mechanisms involved in HPD.
Collapse
Affiliation(s)
- Roberto Ferrara
- Cancer Medicine Department, Gustave Roussy, Villejuif, France
| | - Laura Mezquita
- Cancer Medicine Department, Gustave Roussy, Villejuif, France
| | - Matthieu Texier
- Biostatistics and Epidemiology Department, Gustave Roussy, Villejuif, France
| | - Jihene Lahmar
- Cancer Medicine Department, Gustave Roussy, Villejuif, France
| | | | - Laurent Tessonnier
- Nuclear Medicine Department, Centre Hospitalier Toulon Sainte-Musse, Toulon, France
| | - Julien Mazieres
- Pneumology Department, Centre Hospitalier Universitaire de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Gerard Zalcman
- Thoracic Oncology Department, Hôpital Bichat-Claude Bernard, Université Paris-Diderot, Paris, France
| | - Solenn Brosseau
- Thoracic Oncology Department, Hôpital Bichat-Claude Bernard, Université Paris-Diderot, Paris, France
| | | | - Laura Leroy
- Medical Oncology Department, Institute Bergonié, Bordeaux, France
| | - Boris Duchemann
- Medical Oncology Department, Hôpital Avicenne, Bobigny, France
| | - Corentin Lefebvre
- Service des Maladies Respiratoires, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Remi Veillon
- Service des Maladies Respiratoires, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Virginie Westeel
- Pneumology Department, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | - Serge Koscielny
- Biostatistics and Epidemiology Department, Gustave Roussy, Villejuif, France
| | - Stephane Champiat
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - Charles Ferté
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - David Planchard
- Cancer Medicine Department, Gustave Roussy, Villejuif, France
| | - Jordi Remon
- Cancer Medicine Department, Gustave Roussy, Villejuif, France
| | | | - Anas Gazzah
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - Julien Adam
- Pathology Department, Gustave Roussy, Villejuif, France
| | - Emilio Bria
- Medical Oncology, Fondazione Policlinico Universitario Gemelli IRCCS, Università Cattolica Del Sacro Cuore, Rome, Italy
| | | | | | - Benjamin Besse
- Cancer Medicine Department, Gustave Roussy, Villejuif, France.,Paris-Sud University, Orsay, France
| | | |
Collapse
|
15
|
Expression of costimulatory and inhibitory receptors in FoxP3 + regulatory T cells within the tumor microenvironment: Implications for combination immunotherapy approaches. Adv Cancer Res 2019; 144:193-261. [PMID: 31349899 DOI: 10.1016/bs.acr.2019.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The unprecedented success of immune checkpoint inhibitors has given rise to a rapidly growing number of immuno-oncology agents undergoing preclinical and clinical development and an exponential increase in possible combinations. Defining a clear rationale for combinations by identifying synergies between immunomodulatory pathways has therefore become a high priority. Immunosuppressive regulatory T cells (Tregs) within the tumor microenvironment (TME) represent a major roadblock to endogenous and therapeutic tumor immunity. However, Tregs are also essential for the maintenance of immunological self-tolerance, and share many molecular pathways with conventional T cells including cytotoxic T cells, the primary mediators of tumor immunity. Hence the inability to specifically target and neutralize Tregs within the TME of cancer patients without globally compromising self-tolerance poses a significant challenge. Here we review recent advances in the characterization of tumor-infiltrating Tregs with a focus on costimulatory and inhibitory receptors. We discuss receptor expression patterns, their functional role in Treg biology and mechanistic insights gained from targeting these receptors in preclinical models to evaluate their potential as clinical targets. We further outline a framework of parameters that could be used to refine the assessment of Tregs in cancer patients and increase their value as predictive biomarkers. Finally, we propose modalities to integrate our increasing knowledge on Treg phenotype and function for the rational design of checkpoint inhibitor-based combination therapies. Such combinations have great potential for synergy, as they could concomitantly enhance cytotoxic T cells and inhibit Tregs within the TME, thereby increasing the efficacy of current cancer immunotherapies.
Collapse
|
16
|
Ponziani FR, Nicoletti A, Gasbarrini A, Pompili M. Diagnostic and therapeutic potential of the gut microbiota in patients with early hepatocellular carcinoma. Ther Adv Med Oncol 2019; 11:1758835919848184. [PMID: 31205505 PMCID: PMC6535703 DOI: 10.1177/1758835919848184] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/12/2019] [Indexed: 12/16/2022] Open
Abstract
The gut microbiota is involved in the maintenance of the homeostasis of the human body and its alterations are associated with the development of different pathological conditions. The liver is the organ most exposed to the influence of the gut microbiota, and recently important connections between the intestinal flora and hepatocellular carcinoma (HCC) have been described. In fact, HCC is commonly associated with liver cirrhosis and develops in a microenvironment where inflammation, immunological alterations, and cellular aberrations are dramatically evident. Prevention and diagnosis in the earliest stages are still the most effective weapons in fighting this tumor. Animal models show that the gut microbiota can be involved in the promotion and progression of HCC directly or through different pathogenic mechanisms. Recent data in humans have confirmed these preclinical findings, shedding new light on HCC pathogenesis. Limitations due to the different experimental design, the ethnic and hepatological setting make it difficult to compare the results and draw definitive conclusions, but these studies lay the foundations for a pathogenetic redefinition of HCC. Therefore, it is evident that the characterization of the gut microbiota and its modulation can have an enormous diagnostic, preventive, and therapeutic potential, especially in patients with early stage HCC.
Collapse
Affiliation(s)
- Francesca Romana Ponziani
- Division of Internal Medicine, Gastroenterology and Hepatology, Fondazione Policlinico Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, Rome, 00168, Italy
| | - Alberto Nicoletti
- Internal Medicine, Gastroenterology and Hepatology, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Internal Medicine, Gastroenterology and Hepatology, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy
| | - Maurizio Pompili
- Internal Medicine, Gastroenterology and Hepatology, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
17
|
Cho Y, Han J, Kim W. Recent Advances and Future Directions in Immunotherapeutics for Hepatocellular Carcinoma. ACTA ACUST UNITED AC 2019. [DOI: 10.17998/jlc.19.1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
18
|
Liang S, Cai J, Li Y, Yang R. 1,25‑Dihydroxy‑Vitamin D3 induces macrophage polarization to M2 by upregulating T‑cell Ig‑mucin‑3 expression. Mol Med Rep 2019; 19:3707-3713. [PMID: 30896850 PMCID: PMC6472136 DOI: 10.3892/mmr.2019.10047] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 03/06/2019] [Indexed: 12/14/2022] Open
Abstract
Macrophage polarization serves an important role in immune regulation that is regulated by T-cell immunoglobulin-mucin-3 (Tim-3). The objective of the present study was to explore the role of 1,25-dihydroxy-vitamin D3 [1,25(OH)2D3] in macrophage polarization. Plasmid transfection techniques were applied to prepare RAW264.7 cells with silenced or overexpressed Tim-3 gene. ELISAs were used to examine the level of inflammatory factors secreted by macrophages. Proteins levels were determined by western blot analysis. mRNAs expression levels were assessed using reverse transcription quantitative polymerase chain reaction. It was identified that 1,25(OH)2D3 upregulated Tim-3 levels and promoted the secretion of interleukin (IL)-10. 1,25(OH)2D3 was also observed to increase the level of transforming growth factor-β and to inhibit tumor necrosis factor-α and IL-6. The results also suggested that Tim-3 gene silencing induced macrophages polarization to classically activated macrophages (M1), and that overexpression of the Tim-3 gene induced macrophage polarization to alternatively activated macrophages (M2). 1,25(OH)2D3 treatment upregulated the expression level of Tim-3 in macrophages, which promoted cell polarization to M2 and inhibited polarization to M1. The data from the present study indicated that Tim-3 may induce macrophage polarization to M2, and that 1,25(OH)2D3 produced immunosuppressive effects by upregulating Tim-3.
Collapse
Affiliation(s)
- Shuyu Liang
- Department of Pediatrics, Tianjin First Center Hospital, Tianjin 300192, P.R. China
| | - Jinzhen Cai
- Department of Transplantation Surgery, Tianjin First Center Hospital, Tianjin 300192, P.R. China
| | - Yani Li
- Department of Pediatrics, Tianjin First Center Hospital, Tianjin 300192, P.R. China
| | - Rui Yang
- Department of Pediatrics, Tianjin First Center Hospital, Tianjin 300192, P.R. China
| |
Collapse
|
19
|
Machala EA, McSharry BP, Rouse BT, Abendroth A, Slobedman B. Gal power: the diverse roles of galectins in regulating viral infections. J Gen Virol 2019; 100:333-349. [PMID: 30648945 DOI: 10.1099/jgv.0.001208] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Viruses, as a class of pathogenic microbe, remain a significant health burden globally. Viral infections result in significant morbidity and mortality annually and many remain in need of novel vaccine and anti-viral strategies. The development of effective novel anti-viral therapeutics, in particular, requires detailed understanding of the mechanism of viral infection, and the host response, including the innate and adaptive arms of the immune system. In recent years, the role of glycans and lectins in pathogen-host interactions has become an increasingly relevant issue. This review focuses on the interactions between a specific lectin family, galectins, and the broad range of viral infections in which they play a role. Discussed are the diverse activities that galectins play in interacting directly with virions or the cells they infect, to promote or inhibit viral infection. In addition we describe how galectin expression is regulated both transcriptionally and post-transcriptionally by viral infections. We also compare the contribution of known galectin-mediated immune modulation, across a range of innate and adaptive immune anti-viral responses, to the outcome of viral infections.
Collapse
Affiliation(s)
- Emily A Machala
- 1Discipline of Infectious Diseases and Immunology, University of Sydney, Camperdown, New South Wales, Australia
| | - Brian P McSharry
- 1Discipline of Infectious Diseases and Immunology, University of Sydney, Camperdown, New South Wales, Australia
| | - Barry T Rouse
- 2Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Allison Abendroth
- 1Discipline of Infectious Diseases and Immunology, University of Sydney, Camperdown, New South Wales, Australia
| | - Barry Slobedman
- 1Discipline of Infectious Diseases and Immunology, University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
20
|
Ferrara R, Mezquita L, Texier M, Lahmar J, Audigier-Valette C, Tessonnier L, Mazieres J, Zalcman G, Brosseau S, Le Moulec S, Leroy L, Duchemann B, Lefebvre C, Veillon R, Westeel V, Koscielny S, Champiat S, Ferté C, Planchard D, Remon J, Boucher ME, Gazzah A, Adam J, Bria E, Tortora G, Soria JC, Besse B, Caramella C. Hyperprogressive Disease in Patients With Advanced Non-Small Cell Lung Cancer Treated With PD-1/PD-L1 Inhibitors or With Single-Agent Chemotherapy. JAMA Oncol 2018. [PMID: 30193240 DOI: 10.1001/jamaoncol.2018.3676.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Hyperprogressive disease (HPD) is a new pattern of progression recently described in patients with cancer treated with programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1) inhibitors. The rate and outcome of HPD in advanced non-small cell lung cancer (NSCLC) are unknown. Objectives To investigate whether HPD is observed in patients with advanced NSCLC treated with PD-1/PD-L1 inhibitors compared with single-agent chemotherapy and whether there is an association between treatment and HPD. Design, Setting, and Participants In this multicenter retrospective study that included patients treated between August 4, 2011, and April 5, 2017, the setting was pretreated patients with advanced NSCLC who received PD-1/PD-L1 inhibitors (8 institutions) or single-agent chemotherapy (4 institutions) in France. Measurable disease defined by Response Evaluation Criteria in Solid Tumors (RECIST version 1.1) on at least 2 computed tomographic scans before treatment and 1 computed tomographic scan during treatment was required. Interventions The tumor growth rate (TGR) before and during treatment and variation per month (ΔTGR) were calculated. Hyperprogressive disease was defined as disease progression at the first evaluation with ΔTGR exceeding 50%. Main Outcomes and Measures The primary end point was assessment of the HPD rate in patients treated with IO or chemotherapy. Results Among 406 eligible patients treated with PD-1/PD-L1 inhibitors (63.8% male), 46.3% (n = 188) were 65 years or older, 72.4% (n = 294) had nonsquamous histology, and 92.9% (n = 377) received a PD-1 inhibitor as monotherapy in second-line therapy or later. The median follow-up was 12.1 months (95% CI, 10.1-13.8 months), and the median overall survival (OS) was 13.4 months (95% CI, 10.2-17.0 months). Fifty-six patients (13.8%) were classified as having HPD. Pseudoprogression was observed in 4.7% (n = 19) of the population. Hyperprogressive disease was significantly associated with more than 2 metastatic sites before PD-1/PD-L1 inhibitors compared with non-HPD (62.5% [35 of 56] vs 42.6% [149 of 350]; P = .006). Patients experiencing HPD within the first 6 weeks of PD-1/PD-L1 inhibitor treatment had significantly lower OS compared with patients with progressive disease (median OS, 3.4 months [95% CI, 2.8-7.5 months] vs 6.2 months [95% CI, 5.3-7.9 months]; hazard ratio, 2.18 [95% CI, 1.29-3.69]; P = .003). Among 59 eligible patients treated with chemotherapy, 3 (5.1%) were classified as having HPD. Conclusions and Relevance Our study suggests that HPD is more common with PD-1/PD-L1 inhibitors compared with chemotherapy in pretreated patients with NSCLC and is also associated with high metastatic burden and poor prognosis in patients treated with PD-1/PD-L1 inhibitors. Additional studies are needed to determine the molecular mechanisms involved in HPD.
Collapse
Affiliation(s)
- Roberto Ferrara
- Cancer Medicine Department, Gustave Roussy, Villejuif, France
| | - Laura Mezquita
- Cancer Medicine Department, Gustave Roussy, Villejuif, France
| | - Matthieu Texier
- Biostatistics and Epidemiology Department, Gustave Roussy, Villejuif, France
| | - Jihene Lahmar
- Cancer Medicine Department, Gustave Roussy, Villejuif, France
| | | | - Laurent Tessonnier
- Nuclear Medicine Department, Centre Hospitalier Toulon Sainte-Musse, Toulon, France
| | - Julien Mazieres
- Pneumology Department, Centre Hospitalier Universitaire de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Gerard Zalcman
- Thoracic Oncology Department, Hôpital Bichat-Claude Bernard, Université Paris-Diderot, Paris, France
| | - Solenn Brosseau
- Thoracic Oncology Department, Hôpital Bichat-Claude Bernard, Université Paris-Diderot, Paris, France
| | | | - Laura Leroy
- Medical Oncology Department, Institute Bergonié, Bordeaux, France
| | - Boris Duchemann
- Medical Oncology Department, Hôpital Avicenne, Bobigny, France
| | - Corentin Lefebvre
- Service des Maladies Respiratoires, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Remi Veillon
- Service des Maladies Respiratoires, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Virginie Westeel
- Pneumology Department, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | - Serge Koscielny
- Biostatistics and Epidemiology Department, Gustave Roussy, Villejuif, France
| | - Stephane Champiat
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - Charles Ferté
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - David Planchard
- Cancer Medicine Department, Gustave Roussy, Villejuif, France
| | - Jordi Remon
- Cancer Medicine Department, Gustave Roussy, Villejuif, France
| | | | - Anas Gazzah
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - Julien Adam
- Pathology Department, Gustave Roussy, Villejuif, France
| | - Emilio Bria
- Medical Oncology, Fondazione Policlinico Universitario Gemelli IRCCS, Università Cattolica Del Sacro Cuore, Rome, Italy
| | | | | | - Benjamin Besse
- Cancer Medicine Department, Gustave Roussy, Villejuif, France.,Paris-Sud University, Orsay, France
| | | |
Collapse
|
21
|
Viral hepatitis, inflammation, and cancer: A lesson for autoimmunity. J Autoimmun 2018; 95:58-68. [PMID: 30509387 DOI: 10.1016/j.jaut.2018.10.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 10/22/2018] [Indexed: 12/23/2022]
Abstract
In the present review, we analyzed the various overlapping and non-mutually exclusive mechanisms that intersect and form complex and highly flexible immunological networks allowing the defense against liver infections and tumors. Liver immunity results from the combination of the skills of systemic and local immune system(s) to sense and recognize pathogen or tumor antigens, to sensitize a wide range of innate and adaptive immune cells, and to clear the "invaders", through the establishment of a transient liver immunopathology state undergoing resolution/control of infections or tumors, and memory development. Then, a special emphasis is placed on discussing about the capacity of the immune system(s) to develop a state of chronic low-level immunopathology adapting through the intervention of simultaneous immunoregulatory mechanisms, when the liver is infected by highly mutable viruses (e.g., hepatitis B or C viruses [HBV or HCV]) capable to escape from the immune recognition. The establishment of chronic inflammation represents an advantage for the species survival, because it guarantees the long-term survival of human hosts despite the virus persistence. However, chronic inflammation, in the long run, can evolve towards severe consequences (decompensated cirrhosis and hepatocellular carcinoma) in some individuals, finding requiring the impelling need of discovering new therapeutic anti-viral and immunostimulatory agents addressed, in combination, to fight especially HBV that, in contrast to HCV, lacks antivirals capable to eradicate the virus. Finally, we discussed the concept proposing that the divergent immunoregulatory mechanisms that develop in persisting infections or tumors, on the one hand, and autoimmunity, on the other hand, are the mirror image of each other, whose understanding is also relevant for preparing novel immunotherapeutic approaches in autoimmune diseases.
Collapse
|
22
|
Hyperprogressive disease: recognizing a novel pattern to improve patient management. Nat Rev Clin Oncol 2018; 15:748-762. [DOI: 10.1038/s41571-018-0111-2] [Citation(s) in RCA: 237] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Zhao J, Dang X, Zhang P, Nguyen LN, Cao D, Wang L, Wu X, Morrison ZD, Zhang Y, Jia Z, Xie Q, Wang L, Ning S, EL Gazzar M, Moorman JP, Yao ZQ. Insufficiency of DNA repair enzyme ATM promotes naive CD4 T-cell loss in chronic hepatitis C virus infection. Cell Discov 2018; 4:16. [PMID: 29644094 PMCID: PMC5891503 DOI: 10.1038/s41421-018-0015-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/21/2017] [Indexed: 12/23/2022] Open
Abstract
T cells have a crucial role in viral clearance and vaccine response; however, the mechanisms regulating their responses to viral infections or vaccinations remain elusive. In this study, we investigated T-cell homeostasis, apoptosis, DNA damage, and repair machineries in a large cohort of subjects with hepatitis C virus (HCV) infection. We found that naive CD4 T cells in chronically HCV-infected individuals (HCV T cells) were significantly reduced compared with age-matched healthy subjects. In addition, HCV T cells were prone to apoptosis and DNA damage, as evidenced by increased 8-oxoguanine expression and γH2AX/53BP1-formed DNA damage foci-hallmarks of DNA damage responses. Mechanistically, the activation of DNA repair enzyme ataxia telangiectasia mutated (ATM) was dampened in HCV T cells. ATM activation was also diminished in healthy T cells exposed to ATM inhibitor or to HCV (core protein) that inhibits the phosphoinositide 3 kinase pathway, mimicking the biological effects in HCV T cells. Importantly, ectopic expression of ATM was sufficient to repair the DNA damage, survival deficit, and cell dysfunctions in HCV T cells. Our results demonstrate that insufficient DNA repair enzyme ATM leads to increased DNA damage and renders HCV T cells prone to apoptotic death, which contribute to the loss of naive T cells in HCV infection. Our study reveals a novel mechanism for T-cell dysregulation and viral persistence, providing a new strategy to improve immunotherapy and vaccine responses against human viral diseases.
Collapse
Affiliation(s)
- Juan Zhao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Xindi Dang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Peixin Zhang
- Department of Infectious Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi’an 710038, China
| | - Lam Nhat Nguyen
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Dechao Cao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Lin Wang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Xiaoyuan Wu
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Zheng D Morrison
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Ying Zhang
- Department of Infectious Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi’an 710038, China
| | - Zhansheng Jia
- Department of Infectious Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi’an 710038, China
| | - Qian Xie
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Biomedical Science, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
| | - Ling Wang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Shunbin Ning
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Mohamed EL Gazzar
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Jonathan P Moorman
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
- Department of Veterans Affairs, Hepatitis (HCV/HIV) Program, James H Quillen VA Medical Center, Johnson City, TN 37614 USA
| | - Zhi Q Yao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
- Department of Veterans Affairs, Hepatitis (HCV/HIV) Program, James H Quillen VA Medical Center, Johnson City, TN 37614 USA
| |
Collapse
|
24
|
Granier C, Gey A, Dariane C, Mejean A, Timsit MO, Blanc C, Verkarre V, Radulescu C, Fabre E, Vano Y, Oudard S, Badoual C, Tartour É. [Tim-3: a novel biomarker and therapeutic target in oncology]. Med Sci (Paris) 2018; 34:231-237. [PMID: 29547109 DOI: 10.1051/medsci/20183403011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
T cells harboring multiple co-inhibitory molecules lose their anti-tumoral functionality. PD-1 is a clinically approved target in cancer therapy, but its expression alone does not mean dysfunctionality. The expression of Tim-3 on numerous cell types (T cell, Treg, dendritic cell, myeloid cells) favors tumor escape to immune cells. Within many tumors, PD-1/Tim-3 coexpressing CD8-T cells lose their ability to secrete cytokines (IFNγ, IL-2, TNFα) and their intratumoral infiltration correlates with a bad prognosis. Tim-3 recently appeared as a potential biomarker of anti-PD-1 resistance. Combined blockade of PD-1 and Tim-3 axis demonstrated potent clinical efficacy in preclinical models and reinforced the rationale of using an anti-Tim-3 to override tumor resistance.
Collapse
Affiliation(s)
- Clémence Granier
- Inserm U970, université Paris Descartes Sorbonne Paris-Cité, Paris, France - Équipe labellisée Ligue contre le cancer, Paris, France
| | - Alain Gey
- Inserm U970, université Paris Descartes Sorbonne Paris-Cité, Paris, France - Hôpital européen Georges Pompidou, service d'immunologie biologique, 20, rue Leblanc, 75015 Paris, France
| | - Charles Dariane
- Inserm U970, université Paris Descartes Sorbonne Paris-Cité, Paris, France - Équipe labellisée Ligue contre le cancer, Paris, France - Hôpital européen Georges Pompidou, Service de chirurgie urologique, APHP, Paris, France
| | - Arnaud Mejean
- Hôpital européen Georges Pompidou, Service de chirurgie urologique, APHP, Paris, France
| | - Marc-Olivier Timsit
- Hôpital européen Georges Pompidou, Service de chirurgie urologique, APHP, Paris, France
| | - Charlotte Blanc
- Inserm U970, université Paris Descartes Sorbonne Paris-Cité, Paris, France
| | - Virginie Verkarre
- Hôpital européen Georges Pompidou, Service d'anatomie pathologique, APHP, Paris, France
| | | | - Elisabeth Fabre
- Hôpital européen Georges Pompidou, Service d'oncologie médicale, APHP Paris, France
| | - Yann Vano
- Hôpital européen Georges Pompidou, Service d'oncologie médicale, APHP Paris, France - Inserm U1138, université Paris Descartes Sorbonne Paris-Cité, Paris, France
| | - Stéphane Oudard
- Inserm U970, université Paris Descartes Sorbonne Paris-Cité, Paris, France - Hôpital européen Georges Pompidou, Service d'oncologie médicale, APHP Paris, France
| | - Cécile Badoual
- Inserm U970, université Paris Descartes Sorbonne Paris-Cité, Paris, France - Équipe labellisée Ligue contre le cancer, Paris, France - Hôpital européen Georges Pompidou, Service d'anatomie pathologique, APHP, Paris, France
| | - Éric Tartour
- Inserm U970, université Paris Descartes Sorbonne Paris-Cité, Paris, France - Équipe labellisée Ligue contre le cancer, Paris, France - Hôpital européen Georges Pompidou, service d'immunologie biologique, 20, rue Leblanc, 75015 Paris, France
| |
Collapse
|
25
|
Mysore KR, Ghobrial RM, Kannanganat S, Minze LJ, Graviss EA, Nguyen DT, Perez KK, Li XC. Longitudinal assessment of T cell inhibitory receptors in liver transplant recipients and their association with posttransplant infections. Am J Transplant 2018; 18:351-363. [PMID: 29068155 PMCID: PMC5790618 DOI: 10.1111/ajt.14546] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 09/25/2017] [Accepted: 10/19/2017] [Indexed: 01/25/2023]
Abstract
Current immunosuppression regimens in organ transplantation primarily inhibit T cells. However, T cells are also critical in protective immunity, especially in immune-compromised patients. In this study, we examined the association of T cell dysfunction, as marked by expression of T cell exhaustion molecules, and posttransplant infections in a cohort of liver transplant patients. We focused on Programmed Death 1 (PD-1) and T cell Ig- and mucin-domain molecule 3 (Tim-3), which are potent co-inhibitory receptors, and their persistent expression often leads to T cell dysfunction and compromised protective immunity. We found that patients with the highest expression of PD-1 +Tim-3+ T cells in the memory compartment before transplantation had increased incidence of infections after liver transplantation, especially within the first 90 days. Longitudinal analysis in the first year showed a strong association between variability of PD-1 and Tim-3 expression by T cells and infectious episodes in transplant patients. Furthermore, T cells that expressed PD-1 and Tim-3 had a significantly reduced capacity in producing interferon (IFN)-γ in vitro, and this reduced IFN-γ production could be partially reversed by blocking PD-1 and Tim-3. Interestingly, the percentage of Foxp3+ regulatory T cells in liver transplant patients was stable in the study period. We concluded that the functional status of T cells before and after liver transplantation, as shown by PD-1 and Tim-3 expression, may be valuable in prognosis and management of posttransplant infections.
Collapse
Affiliation(s)
- Krupa R. Mysore
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Immunobiology & Transplant Science Center, Houston Methodist Hospital, Houston, Texas
| | - Rafik M. Ghobrial
- Immunobiology & Transplant Science Center, Houston Methodist Hospital, Houston, Texas
- Department of Surgery, Weill Cornell Medical College of Cornell University, New York, New York
| | - Sunil Kannanganat
- Immunobiology & Transplant Science Center, Houston Methodist Hospital, Houston, Texas
| | - Laurie J. Minze
- Immunobiology & Transplant Science Center, Houston Methodist Hospital, Houston, Texas
| | - Edward A. Graviss
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas
| | - Duc T. Nguyen
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas
| | | | - Xian C. Li
- Immunobiology & Transplant Science Center, Houston Methodist Hospital, Houston, Texas
- Department of Surgery, Weill Cornell Medical College of Cornell University, New York, New York
| |
Collapse
|
26
|
Polymorphisms in TIM-3 and breast cancer susceptibility in Chinese women: A case-control study. Oncotarget 2018; 7:43703-43712. [PMID: 27248321 PMCID: PMC5190054 DOI: 10.18632/oncotarget.9665] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 05/12/2016] [Indexed: 12/29/2022] Open
Abstract
Previous studies have found associations between polymorphisms in T cell immunoglobulin and mucin domain 3 (TIM-3) and increased risks of various cancers. However, the association between TIM-3 polymorphisms and breast cancer (BC) remains uncertain. In this study, a total of 560 BC patients and 583 age, sex, and ethnicity-matched healthy controls from Northwest China were included. The polymorphisms were genotyped using Sequenom MassARRAY. The expression level of TIM-3 protein was detected by immunohistochemistry. We observed rs10053538 had a significantly increased risk of BC, comparing with the wild-type genotype even after Bonferroni correction. In addition, the rs4704853 G>A variants were more frequent among BC patients than the controls (GA + AA vs. GG: OR = 1.32, 95% CI = 1.03-1.69, P = 0.026); However, the significance was lost after Bonferroni correction (P = 0.078). Furthermore, rs10053538 was associated with lymph node metastasis. Age stratification revealed that among patients aged <49 years, those with the rs4704853 GA/AA genotype had a higher risk of BC; But there was no difference when Bonferroni correction was conducted. Immunohistochemical analysis showed that the expression of TIM-3 protein in the breast cancer tissues was higher in patients carrying the rs10053538 GT+TT genotype than those with GG genotype (P = 0.012). However, we failed to find any difference between BC patients and controls in any rs1036199 genetic model. These findings suggested that rs10053538 in TIM-3 might increase susceptibility to BC and promote the progression of BC in Chinese women.
Collapse
|
27
|
Trujillo-Ochoa JL, Corral-Jara KF, Charles-Niño CL, Panduro A, Fierro NA. Conjugated Bilirubin Upregulates TIM-3 Expression on CD4 +CD25 + T Cells: Anti-Inflammatory Implications for Hepatitis A Virus Infection. Viral Immunol 2017; 31:223-232. [PMID: 29099687 DOI: 10.1089/vim.2017.0103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bilirubin (BR), a metabolite with increased concentrations in plasma during viral hepatitis, has been recognized as a potential immune-modulator. We recently reported that conjugated BR (CB) augments regulatory T cell (Treg) suppressor activity during acute hepatitis A virus (HAV) infection. However, the mechanisms related to the effects of CB on Treg function in the course of hepatotropic viral diseases have not been elucidated. T cell immunoglobulin domain and mucin domain 3 (TIM-3), via its interactions with galectin-9 (GAL-9), is a receptor associated with enhanced Treg function. Thus, TIM-3 expression may be related to the crosstalk between CB and Tregs during HAV infection. Herein, in vitro treatment with high concentrations of CB upregulated TIM-3 expression on Tregs from healthy donors. CB treatment in vitro did not induce de novo Treg generation, and in vitro stimulation with TGF-β, which shows increased secretion during HAV infection, resulted in a trend toward increased TIM-3 expression on Tregs and CD4+ T lymphocytes (TLs) from healthy donors. Interestingly, an upregulation of TIM-3 expression on CD4+CD25+ T cells and an increase in the proportion of CD4+ TLs expressing GAL-9 were found in HAV-infected patients with abnormal CB values relative to healthy controls. In addition, a statistically significantly reduction in IL-17F production was observed after treatment of CD4+ TLs from healthy donors with high doses of CB in vitro. In summary, our results suggest that CB might regulate Treg activity via a TIM-3-mediated mechanism, ultimately leading to an anti-inflammatory hepatoprotective effect.
Collapse
Affiliation(s)
- Jorge L Trujillo-Ochoa
- 1 Unidad de Inmunovirología, Servicio de Biología Molecular en Medicina, Hospital Civil de Guadalajara "Fray Antonio Alcalde," Guadalajara, México .,2 Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara , Guadalajara, México
| | - Karla F Corral-Jara
- 1 Unidad de Inmunovirología, Servicio de Biología Molecular en Medicina, Hospital Civil de Guadalajara "Fray Antonio Alcalde," Guadalajara, México .,3 Departamento de Biología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara , Guadalajara, México
| | - Claudia L Charles-Niño
- 4 Departamento de Microbiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara , Guadalajara, México
| | - Arturo Panduro
- 3 Departamento de Biología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara , Guadalajara, México .,5 Servicio de Biología Molecular, Hospital Civil of Guadalajara "Fray Antonio Alcalde," Guadalajara, México
| | - Nora A Fierro
- 1 Unidad de Inmunovirología, Servicio de Biología Molecular en Medicina, Hospital Civil de Guadalajara "Fray Antonio Alcalde," Guadalajara, México .,2 Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara , Guadalajara, México
| |
Collapse
|
28
|
Abstract
The encouraging results in immunotherapy for melanoma also led the way for translational and clinical research about immune-related mechanisms possibly relevant for gastrointestinal tumours. It is in fact now evident that the immune checkpoint modulation and in particular cell-mediated immune-response through programmed cell death-1 (PD-1) and the cytotoxic T-lymphocyte antigen-4 (CTLA4) receptors along with the regulatory T cells activity all have a relevant role in gastrointestinal cancers as well. This review aims to explore the state of the art of immunotherapy for gastrointestinal tumours, deepening recent scientific evidence regarding anti PD-1/PDL-1 and anti CTLA4 monoclonal antibodies, peptide based vaccine, DNA based vaccine, and pulsed dendritic cells, either alone or in combination with other antineoplastic medical therapy and locoregional treatments. Considering the non-negligible toxicity profile deriving from such a treatment approach, predictive biomarkers of response to immunotherapy in gastrointestinal cancer are also urgently needed in order to better select the patients' group with the highest likelihood of benefit.
Collapse
|
29
|
Corneau A, Cosma A, Even S, Katlama C, Le Grand R, Frachet V, Blanc C, Autran B. Comprehensive Mass Cytometry Analysis of Cell Cycle, Activation, and Coinhibitory Receptors Expression in CD4 T Cells from Healthy and HIV-Infected Individuals. CYTOMETRY PART B-CLINICAL CYTOMETRY 2017; 92:21-32. [PMID: 27997758 DOI: 10.1002/cyto.b.21502] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 12/07/2016] [Accepted: 12/09/2016] [Indexed: 01/22/2023]
Abstract
RATIONALE Mass cytometry allows large multiplex analysis of cell cycle stages together with differentiation, activation, and exhaustion markers, allowing further assessment of the quiescence status of resting CD4 T cells. METHODS Peripheral blood CD4 T lymphocytes from 8 individuals, 4 healthy donors, and 4 HIV-infected on antiretroviral treatment (T) were stained with the same 26 monoclonal antibodies and dyes targeting surface and intracellular markers of differentiation, activation, exhaustion, and cell cycle stages. Samples were run on a CYTOF-2. RESULTS Patterns of naïve [TN] CD4 T cells strongly differed from all other memory subsets central-memory (CM), transitional-memory (TM), effector-memory (EM), and terminally differentiated RA-expressing (TEMRA) subsets, while stem-cell memory (SCM) and T follicular-helper cells (TfH) were close to CM and TM cells with the highest percentages in cell cycle. EM and TEMRA were the most altered by HIV infection, with an increased frequency of activated and cycling cells. Activation markers and coinhibitory receptor expression differed among cell cycle stages, with HLA-DR fitting better than CD25 or CD38 with cycle, and opposite PD-1 gradients along differentiation and cell cycle. "Resting" DR-CD25- CD4+ T cells contained similar amounts of cells in G1 than the activated DR ± CD25± ones but three fold lower cells in S-G2-M. CONCLUSION This broad multiplex mass cytometry analysis demonstrates some subsets of the so-called "resting" CD25-DR- CD4+ T cells contain noticeable amounts of cells into cycle or expressing coinhibitory receptors, opening new avenues for a redefinition of resting peripheral blood CD4 T cells harboring the HIV reservoirs. © 2016 International Clinical Cytometry Society.
Collapse
Affiliation(s)
- Aurélien Corneau
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris, UMRS 1135), Paris, F-75013, France.,UPMC Univ Paris 06, Plateforme de Cytométrie (CyPS), UMS30-LUMIC, Faculté de Médecine Pierre et Marie Curie, Site Pitié-Salpêtrière, Paris, F-75013, France.,EPHE, PSL Research University, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Grenoble, France
| | - Antonio Cosma
- Commissariat de l'Energie Atomique (CEA) INSERM, Paris Sud University, UMR 1184, IDMIT Infrastructures, Fontenay aux Roses, F-92265, France
| | - Sophie Even
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris, UMRS 1135), Paris, F-75013, France
| | - Christine Katlama
- AP-HP, Hôpital Pitié-Salpêtrière, Service de Maladies Infectieuses et Tropicales, Paris, F-75013, France.,Sorbonne Universités, UPMC Univ Paris 06, INSERM, U1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique (IPLESP UMRS 1136), Paris, F-75013, France
| | - Roger Le Grand
- Commissariat de l'Energie Atomique (CEA) INSERM, Paris Sud University, UMR 1184, IDMIT Infrastructures, Fontenay aux Roses, F-92265, France
| | - Véronique Frachet
- EPHE, PSL Research University, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Grenoble, France
| | - Catherine Blanc
- UPMC Univ Paris 06, Plateforme de Cytométrie (CyPS), UMS30-LUMIC, Faculté de Médecine Pierre et Marie Curie, Site Pitié-Salpêtrière, Paris, F-75013, France
| | - Brigitte Autran
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris, UMRS 1135), Paris, F-75013, France.,UPMC Univ Paris 06, Plateforme de Cytométrie (CyPS), UMS30-LUMIC, Faculté de Médecine Pierre et Marie Curie, Site Pitié-Salpêtrière, Paris, F-75013, France.,Département d'Immunologie, AP-HP, Hôpital Pitié-Salpêtrière, Paris, F-75013, France
| |
Collapse
|
30
|
唐 映, 徐 加. Tim-3在肝脏疾病中的调节作用. Shijie Huaren Xiaohua Zazhi 2017; 25:2080-2087. [DOI: 10.11569/wcjd.v25.i23.2080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
T淋巴细胞免疫球蛋白黏蛋白分子(T-cell immunoglobulin domain and mucin domain-containing molecule, Tim)-3是Tim家族中的一员, 为近年来新发现的一种在辅助Ⅰ型T淋巴细胞(Help T cell 1, Th1)上特异性表达的Ⅰ型细胞表面分子. Tim-3作为负性调节因子通过与其配体Galectin-9结合引起细胞死亡, 进而调控Th1型细胞功能. Tim-3还表达于其他类型细胞表面, 如自然杀伤细胞、树突状细胞和单核细胞, 对自身免疫性疾病和其他免疫介导的疾病进行免疫调控. 对Tim-3在不同细胞不同免疫条件下的功能以及如何调节进行研究, 将有利于研发Tim-3的潜在治疗作用. 近年来大量研究显示Tim-3通道与肝脏疾病发生发展有着密切关系, 本文就其在肝脏疾病中的调节作用做一总结.
Collapse
|
31
|
Zhuo Y, Zhang YF, Wu HJ, Qin L, Wang YP, Liu AM, Wang XH. Interaction between Galectin-9/TIM-3 pathway and follicular helper CD4 + T cells contributes to viral persistence in chronic hepatitis C. Biomed Pharmacother 2017; 94:386-393. [PMID: 28772217 DOI: 10.1016/j.biopha.2017.07.134] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/12/2017] [Accepted: 07/25/2017] [Indexed: 02/07/2023] Open
Abstract
Both Galectin 9 (Gal-9)/T-cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) pathway and follicular helper CD4+ T (Tfh) cells play important roles in persistent hepatitis C virus (HCV) infection. Thus, we aimed to investigate the regulatory role of interaction between Gal-9/TIM-3 pathway and Tfh cells in chronic hepatitis C. A total of 44 chronic hepatitis C patients and 19 normal controls (NCs) were enrolled in this study. Purified CD4+ T cells were cultured by TIM-3 Fc protein, recombinant Gal-9, or IL-21 for 48h. TIM-3 expression, Tfh proportion, and IL-21 production was measured, respectively. The immunomodulatory role of Gal-9/TIM-3 and IL-21 was also investigated in HCV cell culture system in vitro. We found that the percentage corresponding to both TIM-3-positive and CXCR5+ICOS+ Tfh cells within CD4+ T cells, which correlated with HCV RNA replication, was significantly elevated in patients with chronic hepatitis C in comparison with those in NCs. Moreover, blockade of Gal-9/TIM-3 pathway by TIM-3 Fc protein increased Tfh cells proportion, IL-21 mRNA and protein expression within purified CD4+ T cells, while activation of Gal-9/TIM-3 signaling by Gal-9 stimulation decreased IL-21 production in both patients with chronic HCV infection and healthy individuals. Meanwhile, high concentrations (100 and 200ng/mL) of IL-21 stimulation also elevated TIM-3 expression on CD4+ T cells in chronic hepatitis C. Furthermore, TIM-3 blockage and IL-21 stimulation suppressed mRNA expressions of HCV-induced antiviral proteins (myxovirus resistance A and oligoadenylate synthetase) in Huh7.5 cells without affecting viral replication in HCV cell culture system. The interaction between Gal-9/TIM-3 pathway and Tfh cells contributed to viral persistent in chronic HCV infection, which might be pivotal for development of new therapeutic approaches for chronic hepatitis C.
Collapse
Affiliation(s)
- Ya Zhuo
- Department of Infectious Diseases II, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Xinxiang, Henan Province, China
| | - Yi-Fu Zhang
- Department of Thyroid Breast and Vascular Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Xinxiang, Henan Province, China
| | - Hong-Jie Wu
- Department of Infectious Diseases II, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Xinxiang, Henan Province, China
| | - Lei Qin
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Xinxiang, Henan Province, China
| | - Yan-Ping Wang
- Department of Infectious Diseases III, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Xinxiang, Henan Province, China
| | - A-Min Liu
- Department of Infectious Diseases II, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Xinxiang, Henan Province, China
| | - Xin-Hong Wang
- Department of Infectious Diseases II, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Xinxiang, Henan Province, China.
| |
Collapse
|
32
|
Longo V, Gnoni A, Gardini AC, Pisconti S, Licchetta A, Scartozzi M, Memeo R, Palmieri VO, Aprile G, Santini D, Nardulli P, Silvestris N, Brunetti O. Immunotherapeutic approaches for hepatocellular carcinoma. Oncotarget 2017; 8:33897-33910. [PMID: 28420805 PMCID: PMC5464921 DOI: 10.18632/oncotarget.15406] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/01/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a cancer with a high mortality rate due to the fact that the diagnosis usually occurs at anadvanced stage. Even in case of curative surgical treatment, recurrence is common. Sorafenib and regorafenib are the only therapeutic agents that have been demonstrated to be effective in advanced HCC, thus novel curative approaches are urgently needed. Recent studies focus on the role of immune system in HCC. In fact, the unique immune response in the liver favors tolerance, which can represent a real challenge for conventional immunotherapy in these patients. Spontaneous immune responses against tumor antigens have been detected, and new immune therapies are under investigation: dendritic cell vaccination, immune-modulator strategy, and immune checkpoint inhibition. In recent years different clinical trials examining the use of immunotherapy to treat HCC have been conducted with initial promising results. This review article will summarize the literature data concerning the potential immunotherapeutic approaches in HCC patients.
Collapse
Affiliation(s)
- Vito Longo
- Medical Oncology Unit, Hospital of Taranto, Taranto, Italy
| | - Antonio Gnoni
- Medical Oncology Unit, Hospital of Gallipoli, Gallipoli, Italy
| | - Andrea Casadei Gardini
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, FC, Italy
| | | | | | - Mario Scartozzi
- Department of Medical Oncology, University of Cagliari, Cagliari, Monserrato, CA, Italy
| | - Riccardo Memeo
- Department of Hepatobiliary Surgery, Ospedale Regionale “F.Miulli”, Strada Pr. Acquaviva - Santeramo, Bari, Italy
| | - Vincenzo Ostilio Palmieri
- Department of Biomedical Sciences and Human Oncology, Clinica Medica “A. Murri”, University of Bari Medical School, Bari, Italy
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo Hospital ULSS 6, Vicenza, Italy
| | - Daniele Santini
- Medical Oncology Unit, University Campus Biomedico, Rome, Italy
| | - Patrizia Nardulli
- Pharmacy Unit, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, Viale Orazio Flacco, Bari, Italy
| | - Oronzo Brunetti
- Medical Oncology Unit, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, Viale Orazio Flacco, Bari, Italy
| |
Collapse
|
33
|
Sun H, Gao W, Pan W, Zhang Q, Wang G, Feng D, Geng X, Yan X, Li S. Tim3+ Foxp3 + Treg Cells Are Potent Inhibitors of Effector T Cells and Are Suppressed in Rheumatoid Arthritis. Inflammation 2017; 40:1342-1350. [DOI: 10.1007/s10753-017-0577-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
34
|
Dong J, Yang XF, Wang LX, Wei X, Wang AH, Hao CQ, Shen HJ, Huang CX, Zhang Y, Lian JQ. Modulation of Tim-3 Expression by Antigen-Dependent and -Independent Factors on T Cells from Patients with Chronic Hepatitis B Virus Infection. Front Cell Infect Microbiol 2017; 7:98. [PMID: 28401068 PMCID: PMC5368241 DOI: 10.3389/fcimb.2017.00098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/13/2017] [Indexed: 12/23/2022] Open
Abstract
T-cell immunoglobulin domain and mucin domain-containing molecule-3 (Tim-3) was up-regulated on viral specific T cells and contributed to T cells exhaustion during chronic hepatitis B virus (HBV) infection. However, modulation of Tim-3 expression was still not fully elucidated. To evaluate the potential viral and inflammatory factors involved in the inductor of Tim-3 expression on T cells, 76 patients with chronic HBV infection (including 40 chronic hepatitis B [CHB] and 36 asymptomatic HBV carriers [AsC]) and 40 of normal controls (NCs) were enrolled in this study. Tim-3 expressions on CD4+ and CD8+ T cells were assessed in response to HBV-encoding antigens, HBV peptide pools, and common γ-chain (γc) cytokines stimulation by flow cytometry. HBV peptides and anti-CD3/CD28 directly induced Tim-3 expression on T cells. γc cytokines also drive Tim-3 up-regulations on both CD4+ and CD8+ T cells in patients with chronic HBV infection. However, γc cytokines did not enhance the Tim-3 inductions by either anti-CD3/CD28 or HBV peptides stimulation. Furthermore, γc cytokines-mediated Tim-3 induction could not be abrogated by γc cytokine receptor-neutralizing antibodies. The current results suggested that elevation of Tim-3 expression on T cells could be regulated by both antigen-dependent and -independent manner in patients with chronic HBV infection. The role of γc cytokines in modulation of inhibitory pathway might be evaluated as immunotherapies in humans.
Collapse
Affiliation(s)
- Jie Dong
- Center for Infectious Diseases, Tangdu Hospital, Fourth Military Medical UniversityXi'an, China; Department of Ophthalmology and Otorhinolaryngology, Tenth Hospital of PLAWuwei, China
| | - Xiao-Fei Yang
- Center for Infectious Diseases, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Lin-Xu Wang
- Center for Infectious Diseases, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Xin Wei
- Center for Infectious Diseases, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - An-Hui Wang
- Department of Epidemiology, School of Public Health, Fourth Military Medical University Xi'an, China
| | - Chun-Qiu Hao
- Center for Infectious Diseases, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Huan-Jun Shen
- Center for Infectious Diseases, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Chang-Xing Huang
- Center for Infectious Diseases, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Ye Zhang
- Center for Infectious Diseases, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Jian-Qi Lian
- Center for Infectious Diseases, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| |
Collapse
|
35
|
Greil R, Hutterer E, Hartmann TN, Pleyer L. Reactivation of dormant anti-tumor immunity - a clinical perspective of therapeutic immune checkpoint modulation. Cell Commun Signal 2017; 15:5. [PMID: 28100240 PMCID: PMC5244547 DOI: 10.1186/s12964-016-0155-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/06/2016] [Indexed: 12/17/2022] Open
Abstract
In favor of their outgrowth, cancer cells must resist immune surveillance and edit the immune response. Cancer immunoediting is characterized by fundamental changes in the cellular composition and the inflammatory cytokine profiles in the microenvironment of the primary tumor and metastatic niches, with an ever increasing complexity of interactions between tumor cells and the immune system. Recent data suggest that genetic instability and immunoediting are not necessarily disparate processes. Increasing mutational load may be associated with multiple neoepitopes expressed by the tumor cells and thus increased chances for the immune system to recognize and combat these cells. At the same time the immune system is more and more suppressed and exhausted by this process. Consequently, immune checkpoint modulation may have the potential to be most successful in genetically highly altered and usually extremely unfavorable types of cancer. Moreover, the fact that epitopes recognized by the immune system are preferentially encoded by passenger gene mutations opens windows of synergy in targeting cancer-specific signaling pathways by small molecules simultaneously with antibodies modifying T-cell activation or exhaustion. This review covers some aspects of the current understanding of the immunological basis necessary to understand the rapidly developing therapeutic endeavours in cancer treatment, the clinical achievements made, and raises some burning questions for translational research in this field.
Collapse
Affiliation(s)
- Richard Greil
- Third Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Disease and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, A-5020, Salzburg, Austria. .,Salzburg Cancer Research Institute (SCRI) - Laboratory for Immunological and Molecular Cancer Research (LIMCR), Salzburg, Austria. .,Arbeitsgemeinschaft Medikamentöse Tumortherapie (AGMT) Study Group, Salzburg, Austria. .,Cancer Cluster Salzburg (CCS), Salzburg, Austria.
| | - Evelyn Hutterer
- Third Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Disease and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, A-5020, Salzburg, Austria.,Salzburg Cancer Research Institute (SCRI) - Laboratory for Immunological and Molecular Cancer Research (LIMCR), Salzburg, Austria.,Cancer Cluster Salzburg (CCS), Salzburg, Austria
| | - Tanja Nicole Hartmann
- Third Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Disease and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, A-5020, Salzburg, Austria.,Salzburg Cancer Research Institute (SCRI) - Laboratory for Immunological and Molecular Cancer Research (LIMCR), Salzburg, Austria.,Cancer Cluster Salzburg (CCS), Salzburg, Austria
| | - Lisa Pleyer
- Third Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Disease and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, A-5020, Salzburg, Austria.,Salzburg Cancer Research Institute (SCRI) - Laboratory for Immunological and Molecular Cancer Research (LIMCR), Salzburg, Austria.,Arbeitsgemeinschaft Medikamentöse Tumortherapie (AGMT) Study Group, Salzburg, Austria.,Cancer Cluster Salzburg (CCS), Salzburg, Austria
| |
Collapse
|
36
|
Fouad H, El Raziky M, Hassan EM, Aziz GMA, Darweesh SK, Sayed AR. Regulatory and activated effector T cells in chronic hepatitis C virus: Relation to autoimmunity. World J Hepatol 2016; 8:1287-1294. [PMID: 27843539 PMCID: PMC5084058 DOI: 10.4254/wjh.v8.i30.1287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 07/30/2016] [Accepted: 08/27/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate how Tregs are regulated in chronic hepatitis C virus (HCV) patients via assessment of Tregs markers (granzyme 2, CD69 and FoxP3), Teffs markers [TNFRSF4 (OX40), INFG] and CD4, CD25 genes. METHODS A prospective study was conducted on 120 subjects divided into 4 groups: Group I (n = 30) treatment naïve chronic HCV patients; Group II (n = 30) chronic HCV treated with Peg/Riba; Group III (n = 30) chronic HCV associated with non-organ specific autoantibody and Group IV (n = 30) healthy persons as a control group. Tregs and Teffs markers were assessed in peripheral blood mononuclear cells by quantitative real time reverse transcriptase-polymerase chain reaction. RESULTS Chronic HCV patients exhibited significant higher levels of both Teffs and Tregs in comparison to healthy control group. Tregs markers were significantly decreased in Peg/Riba treated HCV patients in comparison to treatment naïve HCV group. In HCV patients with antinuclear antibody (ANA) +ve, Tregs markers were significantly decreased in comparison to all other studied groups. Teffs markers were significantly elevated in all HCV groups in comparison to control and in HCV group with ANA +ve in comparison to treatment naïve HCV group. CONCLUSION Elevated Tregs cells in chronic HCV patients dampen both CD4+ and CD8+ autologous T cell immune response. Interferon-α and ribavirin therapy suppress proliferation of Tregs. More significant suppression of Tregs was observed in HCV patients with autoantibodies favoring pathological autoimmune response.
Collapse
Affiliation(s)
- Hanan Fouad
- Hanan Fouad, Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Maissa El Raziky
- Hanan Fouad, Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Eman Medhat Hassan
- Hanan Fouad, Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Ghada Mahmoud Abdel Aziz
- Hanan Fouad, Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Samar K Darweesh
- Hanan Fouad, Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Ahmed Reda Sayed
- Hanan Fouad, Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
37
|
Tim-3: Expression on immune cells and roles at the maternal-fetal interface. J Reprod Immunol 2016; 118:92-99. [PMID: 27792886 DOI: 10.1016/j.jri.2016.10.113] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 08/28/2016] [Accepted: 10/19/2016] [Indexed: 12/15/2022]
Abstract
Successful pregnancy relies on the accurate regulation of the maternal-fetal immune system. Without enough tolerance in the uterine microenvironment, the mother and the hemiallogeneic fetus could not peacefully coexist. T cell immunoglobulin and mucin domain (Tim)-3 is a molecule originally regarded as to be expressed on terminally differentiated IFN-γ expressing CD4+ T cells (Th1). The engagement of Tim-3 with its ligand, galectin-9 (Gal-9) could induce the exhaustion or apoptosis of effector T cells, and thus might regulate the tolerance. Tim-3 pathway also participates in regulating the activities of CD4+ regulatory T cells, monocyte-macrophages, dendritic cells and natural killer cells. Dysregulation of Tim-3 expression can elicit excessive or inhibited inflammatory responses and ultimately result in autoimmune diseases, viral or tumor evasion and pregnancy complications. In this review, we will mainly focus on the expression of Tim-3 on local immune cells and its function in pregnancy. In addition, meaningful questions that need further investigation and the potential roles of Tim-3 in fetal tolerance will be discussed. Deeper understanding of the immune checkpoint receptor Tim-3 will shed new light on exploring the pathogenesis of some pregnancy complications, including pre-eclampsia, intrauterine growth restriction, recurrent spontaneous abortion and preterm birth. Tim-3 pathway might be a new target of immune therapy for pregnancy complications in the future.
Collapse
|
38
|
Ren JP, Ying RS, Cheng YQ, Wang L, El Gazzar M, Li GY, Ning SB, Moorman JP, Yao ZQ. HCV-induced miR146a controls SOCS1/STAT3 and cytokine expression in monocytes to promote regulatory T-cell development. J Viral Hepat 2016; 23:755-66. [PMID: 27004559 PMCID: PMC5028233 DOI: 10.1111/jvh.12537] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/19/2016] [Indexed: 02/06/2023]
Abstract
Host innate and adaptive immune responses must be tightly regulated by an intricate balance between positive and negative signals to ensure their appropriate onset and termination while fighting pathogens and avoiding autoimmunity; persistent pathogens may usurp these regulatory machineries to dampen host immune responses for their persistence in vivo. Here, we demonstrate that miR146a is up-regulated in monocytes from hepatitis C virus (HCV)-infected individuals compared to control subjects. Interestingly, miR146a expression in monocytes without HCV infection increased, whereas its level in monocytes with HCV infection decreased, following Toll-like receptor (TLR) stimulation. This miR146a induction by HCV infection and differential response to TLR stimulation were recapitulated in vitro in monocytes co-cultured with hepatocytes with or without HCV infection. Importantly, inhibition of miR146a in monocytes from HCV-infected patients led to a decrease in IL-23, IL-10 and TGF-β expressions through the induction of suppressor of cytokine signalling 1 (SOCS1) and the inhibition of signal transducer and activator transcription 3 (STAT3), and this subsequently resulted in a decrease in regulatory T cells (Tregs) accumulated during HCV infection. These results suggest that miR146a may regulate SOCS1/STAT3 and cytokine signalling in monocytes, directing T-cell differentiation and balancing immune clearance and immune injury during chronic viral infection.
Collapse
Affiliation(s)
- J P Ren
- Center for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - R S Ying
- Center for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
- Department of Hepatology, Guangzhou Number 8 People's Hospital, Guangzhou, China
| | - Y Q Cheng
- Center for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
- International Center for Diagnosis and Treatment of Liver Diseases, 302 Hospital, Beijing, China
| | - L Wang
- Center for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - M El Gazzar
- Center for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - G Y Li
- Center for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - S B Ning
- Center for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - J P Moorman
- Center for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
- Hepatitis (HCV/HIV) Program, James H. Quillen VA Medical Center, Johnson City, TN, USA
| | - Z Q Yao
- Center for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.
- Hepatitis (HCV/HIV) Program, James H. Quillen VA Medical Center, Johnson City, TN, USA.
| |
Collapse
|
39
|
Zhou Y, Li GY, Ren JP, Wang L, Zhao J, Ning SB, Zhang Y, Lian JQ, Huang CX, Jia ZS, Moorman JP, Yao ZQ. Protection of CD4+ T cells from hepatitis C virus infection-associated senescence via ΔNp63-miR-181a-Sirt1 pathway. J Leukoc Biol 2016; 100:1201-1211. [PMID: 27354409 DOI: 10.1189/jlb.5a0316-119rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 06/06/2016] [Indexed: 12/16/2022] Open
Abstract
T cell dysfunction has a crucial role in establishing and maintaining viral persistence. We have previously shown a decline in miR-181a, which regulates CD4+ T cell responses via DUSP6 overexpression, in individuals with hepatitis C virus (HCV) infection. Here, we describe accelerated T cell senescence in HCV-infected individuals compared with age- and sex-matched healthy subjects. Mechanistic studies revealed that up-regulation of transcription factor ΔNp63 led to the decline of miR-181a expression, resulting in an overexpression of the antiaging protein Sirt1, in CD4+ T cells from HCV-infected individuals. Either reconstituting miR-181a or silencing ΔNp63 or Sirt1 expression in CD4+ T cells led to accelerated T cell senescence, as evidenced by an increased senescence-associated β-galactosidase (SA-β-gal) expression, shortened telomere length, and decreased EdU incorporation; this suggests that HCV-induced T cell senescence is counterregulated by the ΔNp63-miR-181a-Sirt1 pathway. An increase of IL-2 production was observed in these senescent CD4+ T cells and was driven by a markedly reduced frequency of Foxp3+ regulatory T (Treg) cells and increased number of Foxp3- effector T (Teff) cells upon manipulating the ΔNp63-miR-181a-Sirt1 pathway. In conclusion, these findings provide novel mechanistic insights into how HCV uses cellular senescent pathways to regulate T cell functions, revealing new targets for rejuvenating impaired T cell responses during chronic viral infection.
Collapse
Affiliation(s)
- Yun Zhou
- Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA.,Center of Diagnosis and Treatment for Infectious Diseases of Chinese PLA, Tangdu Hospital, Fourth Military Medical University, Xian, China
| | - Guang Y Li
- Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA.,Department of Internal Medicine, Division of Infectious Diseases, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA; and
| | - Jun P Ren
- Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA.,Department of Internal Medicine, Division of Infectious Diseases, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA; and
| | - Ling Wang
- Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA.,Department of Internal Medicine, Division of Infectious Diseases, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA; and
| | - Juan Zhao
- Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA.,Department of Internal Medicine, Division of Infectious Diseases, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA; and
| | - Shun B Ning
- Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA.,Department of Internal Medicine, Division of Infectious Diseases, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA; and
| | - Ying Zhang
- Center of Diagnosis and Treatment for Infectious Diseases of Chinese PLA, Tangdu Hospital, Fourth Military Medical University, Xian, China
| | - Jian Q Lian
- Center of Diagnosis and Treatment for Infectious Diseases of Chinese PLA, Tangdu Hospital, Fourth Military Medical University, Xian, China
| | - Chang X Huang
- Center of Diagnosis and Treatment for Infectious Diseases of Chinese PLA, Tangdu Hospital, Fourth Military Medical University, Xian, China
| | - Zhan S Jia
- Center of Diagnosis and Treatment for Infectious Diseases of Chinese PLA, Tangdu Hospital, Fourth Military Medical University, Xian, China;
| | - Jonathan P Moorman
- Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA.,Department of Internal Medicine, Division of Infectious Diseases, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA; and.,Hepatitis/AIDS (HCV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, Tennessee, USA
| | - Zhi Q Yao
- Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA; .,Department of Internal Medicine, Division of Infectious Diseases, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA; and.,Hepatitis/AIDS (HCV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, Tennessee, USA
| |
Collapse
|
40
|
Jensen CE, Loaiza-Bonilla A, Bonilla-Reyes PA. Immune checkpoint inhibitors for hepatocellular carcinoma. Hepat Oncol 2016; 3:201-211. [PMID: 30191042 DOI: 10.2217/hep-2016-0004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 04/25/2016] [Indexed: 12/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer deaths worldwide, and advanced HCC generally caries a poor prognosis. The treatment of advanced disease is limited to sorafenib, which provides only a limited improvement in survival, and novel therapies are, thus, sorely needed. Among emerging alternative approaches, immune checkpoint inhibitors are a particularly promising treatment modality. In this review, we summarize current knowledge of the mechanisms for the two primary targets of immune checkpoint inhibitors and discuss the relevance of these pathways to the immunology of HCC. We also review the state of ongoing and forthcoming trials of immune checkpoint blockade in HCC.
Collapse
Affiliation(s)
- Christopher E Jensen
- Department of Medicine, The Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA.,Department of Medicine, The Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Arturo Loaiza-Bonilla
- Departments of Medicine, Hematology & Oncology, Abramson Cancer Center of the University of Pennsylvania, Perelman Center for Advanced Medicine, 6th Floor South Pavilion, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA.,Departments of Medicine, Hematology & Oncology, Abramson Cancer Center of the University of Pennsylvania, Perelman Center for Advanced Medicine, 6th Floor South Pavilion, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Paula A Bonilla-Reyes
- Facultad de Medicina, Pontificia Universidad Javeriana, Cra. 7 No. 40-62, Hospital Universitario, San Ignacio, Bogota, Colombia.,Facultad de Medicina, Pontificia Universidad Javeriana, Cra. 7 No. 40-62, Hospital Universitario, San Ignacio, Bogota, Colombia
| |
Collapse
|
41
|
Ren JP, Zhao J, Dai J, Griffin JWD, Wang L, Wu XY, Morrison ZD, Li GY, El Gazzar M, Ning SB, Moorman JP, Yao ZQ. Hepatitis C virus-induced myeloid-derived suppressor cells regulate T-cell differentiation and function via the signal transducer and activator of transcription 3 pathway. Immunology 2016; 148:377-86. [PMID: 27149428 DOI: 10.1111/imm.12616] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 04/19/2016] [Accepted: 04/28/2016] [Indexed: 12/13/2022] Open
Abstract
T cells play a pivotal role in controlling viral infection; however, the precise mechanisms responsible for regulating T-cell differentiation and function during infections are incompletely understood. In this study, we demonstrated an expansion of myeloid-derived suppressor cells (MDSCs), in particular the monocytic MDSCs (M-MDSCs; CD14(+) CD33(+) CD11b(+) HLA-DR(-/low) ), in patients with chronic hepatitis C virus (HCV) infection. Notably, HCV-induced M-MDSCs express high levels of phosphorylated signal transducer and activator of transcription 3 (pSTAT3) and interleukin-10 (IL-10) compared with healthy subjects. Blocking STAT3 signalling reduced HCV-mediated M-MDSC expansion and decreased IL-10 expression. Importantly, we observed a significant increase in the numbers of CD4(+) CD25(+) Foxp3(+) regulatory T (Treg) cells following incubation of healthy peripheral blood mononuclear cells (PBMCs) with MDSCs derived from HCV-infected patients or treated with HCV core protein. In addition, depletion of MDSCs from PBMCs led to a significant reduction of Foxp3(+) Treg cells developed during chronic HCV infection. Moreover, depletion of MDSCs from PBMCs significantly increased interferon-γ production by CD4(+) T effector (Teff) cells derived from HCV patients. These results suggest that HCV-induced MDSCs promote Treg cell development and inhibit Teff cell function, suggesting a novel mechanism for T-cell regulation and a new strategy for immunotherapy against human viral diseases.
Collapse
Affiliation(s)
- Jun P Ren
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Juan Zhao
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Jun Dai
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Jeddidiah W D Griffin
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Ling Wang
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Xiao Y Wu
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Zheng D Morrison
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Guang Y Li
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Mohamed El Gazzar
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Shun B Ning
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Jonathan P Moorman
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Hepatitis (HCV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, TN, USA
| | - Zhi Q Yao
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Hepatitis (HCV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, TN, USA
| |
Collapse
|
42
|
Li S, Wan J, Anderson W, Sun H, Zhang H, Peng X, Yu Z, Wang T, Yan X, Smith W. Downregulation of IL-10 secretion by Treg cells in osteoarthritis is associated with a reduction in Tim-3 expression. Biomed Pharmacother 2016; 79:159-65. [DOI: 10.1016/j.biopha.2016.01.036] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 01/26/2016] [Indexed: 12/16/2022] Open
|
43
|
Han Q, Shi H, Liu F. CD163(+) M2-type tumor-associated macrophage support the suppression of tumor-infiltrating T cells in osteosarcoma. Int Immunopharmacol 2016; 34:101-106. [PMID: 26938675 DOI: 10.1016/j.intimp.2016.01.023] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 01/25/2016] [Accepted: 01/26/2016] [Indexed: 01/29/2023]
Abstract
Osteosarcoma is one of the most common childhood cancers with high numbers of cancer-related deaths. Progress in conventional therapies is showing limited improvement. An adaptive T cell-based immunotherapy represents a promising new therapeutic option, but to improve its efficacy, regulatory mechanisms in osteosarcoma need further elucidation. Here, to evaluate the regulatory effect of tumor microenvironment of T cells in osteosarcoma, we examined the peripheral blood (PB) and tumor infiltrating (TI) T cells, and their correlations with PB and tumor immune characteristics. We found that TI T cells contained significantly higher levels of TIM-3(+)PD-1(-) and TIM-3(+)PD-1(+) cells than their PB counterparts. Similar to that in chronic HIV and HCV infections, these TIM-3(+)PD-1(-) and TIM-3(+)PD-1(+) T cells presented reduced proliferation and proinflammatory cytokine secretion in response to stimulation. Presence of M2-type (CD163(+)) macrophages exacerbated T cell immunosuppression, since frequencies of CD163(+) tumor-associated macrophages were directly correlated with the frequencies of suppressed TIM-3(+)PD-1(+) T cells. Moreover, depletion of CD163(+) macrophages significantly improved T cell proliferation and proinflammatory cytokine production. Overall, our data presented an intratumoral T cell-specific immunosuppression that was amplified by M2-type tumor-associated macrophages.
Collapse
Affiliation(s)
- Qinglin Han
- Surgical Research Laboratory, The Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| | - Hongguang Shi
- Surgical Research Laboratory, The Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Fan Liu
- Surgical Research Laboratory, The Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
44
|
Liu Y, Gao LF, Liang XH, Ma CH. Role of Tim-3 in hepatitis B virus infection: An overview. World J Gastroenterol 2016; 22:2294-2303. [PMID: 26900291 PMCID: PMC4735003 DOI: 10.3748/wjg.v22.i7.2294] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 10/08/2015] [Accepted: 12/21/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus (HBV) infection has received increasing public attention. HBV is the prototypical member of hepadnaviruses, which naturally infect only humans and great apes and induce the acute and persistent chronic infection of hepatocytes. A large body of evidence has demonstrated that dysfunction of the host anti-viral immune response is responsible for persistent HBV replication, unresolved inflammation and disease progression. Many regulatory factors are involved in immune dysfunction. Among these, T cell immunoglobulin domain and mucin domain-3 (Tim-3), one of the immune checkpoint proteins, has attracted increasing attention due to its critical role in regulating both adaptive and innate immune cells. In chronic HBV infection, Tim-3 expression is elevated in many types of immune cells, such as T helper cells, cytotoxic T lymphocytes, dendritic cells, macrophages and natural killer cells. Tim-3 over-expression is often accompanied by impaired function of the above-mentioned immunocytes, and Tim-3 inhibition can at least partially rescue impaired immune function and thus promote viral clearance. A better understanding of the regulatory role of Tim-3 in host immunity during HBV infection will shed new light on the mechanisms of HBV-related liver disease and suggest new therapeutic methods for intervention.
Collapse
|
45
|
Hato T, Zhu AX, Duda DG. Rationally combining anti-VEGF therapy with checkpoint inhibitors in hepatocellular carcinoma. Immunotherapy 2016; 8:299-313. [PMID: 26865127 DOI: 10.2217/imt.15.126] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a fatal disease with rising incidence in the world. For advanced HCC, sorafenib, a multikinase inhibitor, is the only systemic therapy with proven survival benefits. Sorafenib is a pan-VEGF receptor inhibitor, and thus many studies have focused its antivascular effects. But VEGF also acts as an immunosuppressive molecule. VEGF can inhibit maturation of dendritic cells, promote immune suppressive cell infiltration and enhance immune checkpoint molecules expression. On the other hand, potent VEGF inhibition may increase tumor hypoxia, which could hinder antitumor immunity or immunotherapy. Thus, achieving synergy when combining anti-VEGF therapy with immunotherapy may require proper polarization of the tumor microenvironment by dose titration or combination with other immunomodulating agents.
Collapse
Affiliation(s)
- Tai Hato
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.,Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Andrew X Zhu
- Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | - Dan G Duda
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
46
|
Wang Y, Zheng Z, Zhu X, Han J, Dong M, Tao K, Wang H, Wang Y, Hu D. The amelioration of composite tissue allograft rejection by TIM-3-modified dendritic cell: Regulation of the balance of regulatory and effector T cells. Immunol Lett 2016; 169:15-22. [DOI: 10.1016/j.imlet.2015.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/19/2015] [Accepted: 11/04/2015] [Indexed: 11/29/2022]
|
47
|
Martínez-Esparza M, Tristán-Manzano M, Ruiz-Alcaraz AJ, García-Peñarrubia P. Inflammatory status in human hepatic cirrhosis. World J Gastroenterol 2015; 21:11522-11541. [PMID: 26556984 PMCID: PMC4631958 DOI: 10.3748/wjg.v21.i41.11522] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 07/31/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
This review focuses on new findings about the inflammatory status involved in the development of human liver cirrhosis induced by the two main causes, hepatitis C virus (HCV) infection and chronic alcohol abuse, avoiding results obtained from animal models. When liver is faced to a persistent and/or intense local damage the maintained inflammatory response gives rise to a progressive replacement of normal hepatic tissue by non-functional fibrotic scar. The imbalance between tissue regeneration and fibrosis will determine the outcome toward health recovery or hepatic cirrhosis. In all cases progression toward liver cirrhosis is caused by a dysregulation of mechanisms that govern the balance between activation/homeostasis of the immune system. Detecting differences between the inflammatory status in HCV-induced vs alcohol-induced cirrhosis could be useful to identify specific targets for preventive and therapeutic intervention in each case. Thus, although survival of patients with alcoholic cirrhosis seems to be similar to that of patients with HCV-related cirrhosis (HCV-C), there are important differences in the altered cellular and molecular mechanisms implicated in the progression toward human liver cirrhosis. The predominant features of HCV-C are more related with those that allow viral evasion of the immune defenses, especially although not exclusively, inhibition of interferons secretion, natural killer cells activation and T cell-mediated cytotoxicity. On the contrary, the inflammatory status of alcohol-induced cirrhosis is determined by the combined effect of direct hepatotoxicity of ethanol metabolites and increases of the intestinal permeability, allowing bacteria and bacterial products translocation, into the portal circulation, mesenteric lymph nodes and peritoneal cavity. This phenomenon generates a stronger pro-inflammatory response compared with HCV-related cirrhosis. Hence, therapeutic intervention in HCV-related cirrhosis must be mainly focused to counteract HCV-immune system evasion, while in the case of alcohol-induced cirrhosis it must try to break the inflammatory loop established at the gut-mesenteric lymph nodes-peritoneal-systemic axis.
Collapse
|
48
|
Bu M, Shen Y, Seeger WL, An S, Qi R, Sanderson JA, Cai Y. Ovarian carcinoma-infiltrating regulatory T cells were more potent suppressors of CD8(+) T cell inflammation than their peripheral counterparts, a function dependent on TIM3 expression. Tumour Biol 2015; 37:3949-56. [PMID: 26482613 DOI: 10.1007/s13277-015-4237-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 10/12/2015] [Indexed: 01/15/2023] Open
Abstract
Ovarian carcinoma is one of the most severe cancers in women, with a high relapse rate and limited secondary treatment options. To assist research in novel treatment technologies, including CD8(+) T cell-base immunotherapy, we examined the effect of tumor-infiltrating regulatory T cells (Tregs) in inhibiting CD8(+) T cell inflammation. We found that compared to their peripheral blood counterparts, tumor-infiltrating Tregs exhibited more potent inhibitory function, which was associated with higher interleukin 10 (IL-10) production in tumor-infiltrating Tregs. Blockade of T cell immunoglobulin mucin 3 (TIM3), a regulatory molecule overrepresented on tumor-infiltrating Tregs, had significantly reverted Treg-mediated suppression. Moreover, expression of TIM3 on tumor-infiltrating Tregs was directly correlated with tumor size. Together, our results demonstrated that ovarian tumor-infiltrating Treg cells were more immunosuppressive than their peripheral blood counterparts in a TIM3-dependent fashion.
Collapse
Affiliation(s)
- Meimei Bu
- Department of Anesthesiology, The Maternal and Child Health Hospital of Jinan City, Jinan, Shandong, 250001, China
| | - Yizhen Shen
- Department of Urology, General Hospital of Jinan Military Command, Jinan, Shandong, 250031, China
| | | | - Shizhi An
- Department of Anesthesiology, The Maternal and Child Health Hospital of Jinan City, Jinan, Shandong, 250001, China
| | - Rongqin Qi
- Department of Anesthesiology, The Maternal and Child Health Hospital of Jinan City, Jinan, Shandong, 250001, China
| | | | - Yan Cai
- Department of Prenatal Diagnosis, The Maternal and Child Health Hospital of Jinan City, No.2 Jian Guo Xiao Jing San Road, Jinan, Shandong, 250001, China.
| |
Collapse
|
49
|
Dinney CM, Zhao LD, Conrad CD, Duker JM, Karas RO, Hu Z, Hamilton MA, Gillis TR, Parker TM, Fan B, Advani AH, Poordad FB, Fauceglia PL, Kirsch KM, Munk PT, Ladanyi MP, Bochner BA, Bekelman JA, Grandori CM, Olson JC, Lechan RD, Abou GMA, Goodarzi MA. Regulation of HBV-specific CD8(+) T cell-mediated inflammation is diversified in different clinical presentations of HBV infection. J Microbiol 2015; 53:718-24. [PMID: 26428923 DOI: 10.1007/s12275-015-5314-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 07/13/2015] [Accepted: 08/12/2015] [Indexed: 02/07/2023]
Abstract
Chronic HBV infection is the leading cause of liver cirrhosis and hepatic cancer, but the individual responses toward HBV infection are highly variable, ranging from asymptomatic to chronic active hepatitis B inflammation. In this study, we hypothesized that the different individual responses to HBV infection was associated with differences in HBV-specific CD8(+) T cell-mediated inflammation and cytotoxicity. Blood samples were collected from subjects with asymptomatic HBV-infection, subjects undergoing active chronic HBV flares (active CHB), and subjects with HBV-infected hepatocellular carcinoma (HBV-HCC). By tetramer staining, we found that all three groups had similar frequencies of HBVspecific CD8(+) T cells. However, after HBV peptide stimulation, the HBV-specific CD8(+) T cells in asymptomatic subjects had significantly stronger interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), and CD107a expression than those in active CHB and HBV-HCC patients. Examination of surface marker expression revealed that the PD-1(-)Tim-3(-) double-negative cell population was the main contributor to HBV-specific inflammation. In active CHB patients and HBV-HCC patients, however, the frequencies of activated PD-1(-)Tim-3(-) cells were significantly reduced. Moreover, the serum HBV DNA titer was not correlated with the frequencies of HBV-specific CD8(+) T cells but was inversely correlated with the frequencies of IFN-g-expressing and CD107a-express cells in response to HBV stimulation. Together, our data demonstrated that the status of HBVspecific CD8(+) T cell exhaustion was associated with different clinical outcomes of chronic HBV infection.
Collapse
Affiliation(s)
- Colin M Dinney
- Wayne State University Medical Center, Detroit, Michigan, 48201, USA
| | - Lu-Dong Zhao
- Department of Hepatobiliary Surgery, Linyi People's Hospital, Shandong, 276000, P. R. China
| | - Charles D Conrad
- Wayne State University Medical Center, Detroit, Michigan, 48201, USA
| | - Jay M Duker
- Wayne State University Medical Center, Detroit, Michigan, 48201, USA
| | - Richard O Karas
- Wayne State University Medical Center, Detroit, Michigan, 48201, USA
| | - Zhibin Hu
- Wayne State University Medical Center, Detroit, Michigan, 48201, USA
| | - Michele A Hamilton
- Department of Medicine, University of Maryland, Medscientist Group, Baltimore, MD, 21201, USA
| | - Thomas R Gillis
- Department of Medicine, University of Maryland, Medscientist Group, Baltimore, MD, 21201, USA
| | | | - Bing Fan
- Tufts University, Boston, MA, 02111, USA
| | | | - Fred B Poordad
- University of British Columbia, Vancouver, BC, V6T 2B5, Canada
| | | | | | - Peter T Munk
- Georgetown University, Washington, DC, 20057, USA
| | | | | | | | | | - James C Olson
- University of British Columbia, Vancouver, BC, V6T 2B5, Canada.
| | | | - Ghassan M A Abou
- Department of Medicine, University of Maryland, Medscientist Group, Baltimore, MD, 21201, USA.
| | - Mark A Goodarzi
- Department of Medicine, University of Maryland, Medscientist Group, Baltimore, MD, 21201, USA.
| |
Collapse
|
50
|
Jia YH, Fan ZJ, Guo H, Zhou SF, Liu SY. Correlations between HCV RNA load and titers of anti-HCV antibody and anti-nuclear antibody in chronic hepatitis C patients. Shijie Huaren Xiaohua Zazhi 2015; 23:3766-3770. [DOI: 10.11569/wcjd.v23.i23.3766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the correlations between hepatitis C virus (HCV) RNA load and the titers of anti-HCV antibody and anti-nuclear antibody (ANA), and the change in ANA titers before and after interferon treatment, to provide guidance for the treatment of chronic hepatitis C (CHC).
METHODS: Serum samples were collected from 112 patients with CHC from January 2013 to February 2014 at Tianjin Third Central Hospital. Real-time fluorescence quantitative polymerase chain reaction (FQ-PCR) was performed to detect HCV RNA, ELISA was used for the detection of anti-HCV antibody, and indirect immunofluorescence (IIF) was performed for the detection of ANA. Spearman correlation analysis was used to analyse the correlations between Log HCV RNA and the titers of anti-HCV antibody and ANA. Independent samples t-test and K independent samples test analysis were used to analyse the correlation between Log HCV RNA and the titer of ANA. Rank sum test was used to compare ANA titers before and after treatment with interferon for one month.
RESULTS: There was no correlation between HCV RNA load and the titer of anti-HCV antibody (r = 0.078, P = 0.465). There was a correlation between HCV RNA load and ANA titer (r = 0.744, P = 0.001). There was a significant difference in HCV RNA between the ANA(+)/(-) groups (P = 0.001). HCV RNA varied among different titers of ANA (χ2 = 32.58, P = 0.001). The change in ANA titers before and after interferon treatment was statistically significant (Z = 2.60, P = 0.001). Most CHC patients had a low titer of ANA. With the increase in viral load, the number of patients with a high titer of ANA had an increasing trend.
CONCLUSION: HCV RNA can be used to evaluate the antiviral effect. There is a certain degree of autoimmune phenomenon in patients with CHC. Interferon can enhance autoimmunity in the course of therapy.
Collapse
|