1
|
Kwantwi LB. SLAM family-mediated crosstalk between tumor and immune cells in the tumor microenvironment: a promising biomarker and a potential therapeutic target for immune checkpoint therapies. Clin Transl Oncol 2025; 27:901-908. [PMID: 39212911 DOI: 10.1007/s12094-024-03675-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Immune cells infiltrating the tumor microenvironment are physiologically important in controlling cancers. However, emerging studies have shown that cancer cells can evade immune surveillance and establish a balance in which these immune cells support tumor progression and therapeutic resistance. The signaling lymphocytic activation molecule family members have been recognized as mediators of tumor microenvironment interactions, and a promising therapeutic target for cancer immunotherapy. This review is focused on the role of SLAM family in tumor and immune cell interactions and discusses how such crosstalk affects tumor behavior. This will shed insight into the next step toward improving cancer immunotherapy.
Collapse
Affiliation(s)
- Louis Boafo Kwantwi
- Department of Anatomy and Neurobiology, College of Medicine, Northeast Ohio Medical University, Rootstown, OH, 44272, USA.
| |
Collapse
|
2
|
Zhang R, Zhao Y, Chai X, Wang Y, Zhao M, Guo S, Zhang Y, Zhao M. Modified CD15/CD16-CLL1 inhibitory CAR-T cells for mitigating granulocytopenia toxicities in the treatment of acute myeloid leukemia. Transl Oncol 2025; 52:102225. [PMID: 39647325 PMCID: PMC11667018 DOI: 10.1016/j.tranon.2024.102225] [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: 08/01/2024] [Revised: 11/14/2024] [Accepted: 11/28/2024] [Indexed: 12/10/2024] Open
Abstract
CLL1 Chimeric antigen receptor T-cell (CAR-T) therapy, as a promising immunotherapeutic approach, has demonstrated its potential to enhance the prognosis of patients diagnosed with acute myeloid leukemia (AML). However, due to the overexpression of CLL1 on neutrophils, CAR-T cells not only eliminated tumor cells but also eradicated neutrophils simultaneously, resulting in severe granulocytopenia and subsequent infections. Considering the distinct expression levels of CD15/CD16 on neutrophils and AML blasts, we have devised novel modified CD15 /CD16-CLL1 iCAR structures incorporating diverse inhibitory elements. Through extensive screening of structural optimization, we have successfully identified CD16-CLL1 iCAR-T cells that combine PD1 and 2B4 blockade, as well as a single VHH fragment replacing the entire CD16 scFv recognition domain. These modified cells demonstrate enhanced cytotoxicity against blasts while minimizing neutrophil elimination. Furthermore, their functionality has been effectively validated through both in vitro and in vivo experiments. In conclusion, we have successfully engineered innovative CD16-CLL1 iCAR-T cells, which preserves the cytotoxicity against tumor cells while preventing elimination of neutrophils, thereby significantly reducing the incidence of granulocytopenia during CAR-T therapy. Furthermore, our future objectives encompass the meticulous validation of both the efficacy and safety profile of this groundbreaking CAR-T therapy in clinical trials, as well as a comprehensive assessment of its potential to enhance the prognosis of patients diagnosed with AML.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300380, China
| | - Yifan Zhao
- The First Central Clinical College of Tianjin Medical University, Tianjin 300380, China
| | - Xiao Chai
- The First Central Clinical College of Tianjin Medical University, Tianjin 300380, China
| | - Yingshuai Wang
- Department of Internal Medicine III, University Hospital Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Mohan Zhao
- The First Central Clinical College of Tianjin Medical University, Tianjin 300380, China
| | - Shujing Guo
- The First Central Clinical College of Tianjin Medical University, Tianjin 300380, China
| | - Yu Zhang
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300380, China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300380, China.
| |
Collapse
|
3
|
Gunes M, Rosen ST, Shachar I, Gunes EG. Signaling lymphocytic activation molecule family receptors as potential immune therapeutic targets in solid tumors. Front Immunol 2024; 15:1297473. [PMID: 38476238 PMCID: PMC10927787 DOI: 10.3389/fimmu.2024.1297473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/05/2024] [Indexed: 03/14/2024] Open
Abstract
Recently, cancer immunotherapy has revolutionized cancer treatment. Various forms of immunotherapy have a manageable safety profile and result in prolongation of overall survival in patients with solid tumors, but only in a proportion of patients. Various factors in the tumor microenvironment play critical roles and may be responsible for this lack of therapeutic response. Signaling lymphocytic activation molecule family (SLAMF) members are increasingly being studied as factors impacting the tumor immune microenvironment. SLAMF members consist of nine receptors mainly expressed in immune cells. However, SLAMF receptors have also been detected in cancer cells, and they may be involved in a spectrum of anti-tumor immune responses. Here, we review the current knowledge of the expression of SLAMF receptors in solid tumors and tumor-infiltrating immune cells and their association with patient outcomes. Furthermore, we discuss the therapeutic potential of targeting SLAMF receptors to improve outcomes of cancer therapy in solid tumors. We believe the research on SLAMF receptor-targeted strategies may enhance anti-cancer immunity in patients with solid tumors and improve clinical outcomes.
Collapse
Affiliation(s)
- Metin Gunes
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Los Angeles, CA, United States
| | - Steven T. Rosen
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Los Angeles, CA, United States
- Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Los Angeles, CA, United States
| | - Idit Shachar
- Department of System Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - E. Gulsen Gunes
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Los Angeles, CA, United States
- Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Los Angeles, CA, United States
- Toni Stephenson Lymphoma Center, City of Hope, Los Angeles, CA, United States
| |
Collapse
|
4
|
Le VV, Ko SR, Oh HM, Ahn CY. Genomic Insights into Paucibacter aquatile DH15, a Cyanobactericidal Bacterium, and Comparative Genomics of the Genus Paucibacter. J Microbiol Biotechnol 2023; 33:1615-1624. [PMID: 37811910 PMCID: PMC10772561 DOI: 10.4014/jmb.2307.07008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 10/10/2023]
Abstract
Microcystis blooms threaten ecosystem function and cause substantial economic losses. Microorganism-based methods, mainly using cyanobactericidal bacteria, are considered one of the most ecologically sound methods to control Microcystis blooms. This study focused on gaining genomic insights into Paucibacter aquatile DH15 that exhibited excellent cyanobactericidal effects against Microcystis. Additionally, a pan-genome analysis of the genus Paucibacter was conducted to enhance our understanding of the ecophysiological significance of this genus. Based on phylogenomic analyses, strain DH15 was classified as a member of the species Paucibacter aquatile. The genome analysis supported that strain DH15 can effectively destroy Microcystis, possibly due to the specific genes involved in the flagellar synthesis, cell wall degradation, and the production of cyanobactericidal compounds. The pan-genome analysis revealed the diversity and adaptability of the genus Paucibacter, highlighting its potential to absorb external genetic elements. Paucibacter species were anticipated to play a vital role in the ecosystem by potentially providing essential nutrients, such as vitamins B7, B12, and heme, to auxotrophic microbial groups. Overall, our findings contribute to understanding the molecular mechanisms underlying the action of cyanobactericidal bacteria against Microcystis and shed light on the ecological significance of the genus Paucibacter.
Collapse
Affiliation(s)
- Ve Van Le
- Cell Factory Research Centre, Korea Research Institute of Bioscience & Biotechnology, Daejeon 34141, Republic of Korea
| | - So-Ra Ko
- Cell Factory Research Centre, Korea Research Institute of Bioscience & Biotechnology, Daejeon 34141, Republic of Korea
| | - Hee-Mock Oh
- Cell Factory Research Centre, Korea Research Institute of Bioscience & Biotechnology, Daejeon 34141, Republic of Korea
- Department of Environmental Biotechnology, KRIBB School of Biotechnology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Chi-Yong Ahn
- Cell Factory Research Centre, Korea Research Institute of Bioscience & Biotechnology, Daejeon 34141, Republic of Korea
- Department of Environmental Biotechnology, KRIBB School of Biotechnology, University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
5
|
Silva EE, Skon-Hegg C, Badovinac VP, Griffith TS. The Calm after the Storm: Implications of Sepsis Immunoparalysis on Host Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:711-719. [PMID: 37603859 PMCID: PMC10449360 DOI: 10.4049/jimmunol.2300171] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/12/2023] [Indexed: 08/23/2023]
Abstract
The immunological hallmarks of sepsis include the inflammation-mediated cytokine storm, apoptosis-driven lymphopenia, and prolonged immunoparalysis. Although early clinical efforts were focused on increasing the survival of patients through the first phase, studies are now shifting attention to the long-term effects of sepsis on immune fitness in survivors. In particular, the most pertinent task is deciphering how the immune system becomes suppressed, leading to increased incidence of secondary infections. In this review, we introduce the contribution of numerical changes and functional reprogramming within innate (NK cells, dendritic cells) and adaptive (T cells, B cells) immune cells on the chronic immune dysregulation in the septic murine and human host. We briefly discuss how prior immunological experience in murine models impacts sepsis severity, immune dysfunction, and clinical relevance. Finally, we dive into how comorbidities, specifically autoimmunity and cancer, can influence host susceptibility to sepsis and the associated immune dysfunction.
Collapse
Affiliation(s)
- Elvia E Silva
- Department of Pathology, University of Iowa, Iowa City, IA
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA
| | - Cara Skon-Hegg
- Department of Urology, University of Minnesota, Minneapolis, MN
| | - Vladimir P Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA
| | - Thomas S Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
- Minneapolis VA Health Care System, Minneapolis, MN
| |
Collapse
|
6
|
Williams JC, Ford ML, Coopersmith CM. Cancer and sepsis. Clin Sci (Lond) 2023; 137:881-893. [PMID: 37314016 PMCID: PMC10635282 DOI: 10.1042/cs20220713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 06/15/2023]
Abstract
Sepsis is one of the leading causes of death worldwide. While mortality is high regardless of inciting infection or comorbidities, mortality in patients with cancer and sepsis is significantly higher than mortality in patients with sepsis without cancer. Cancer patients are also significantly more likely to develop sepsis than the general population. The mechanisms underlying increased mortality in cancer and sepsis patients are multifactorial. Cancer treatment alters the host immune response and can increase susceptibility to infection. Preclinical data also suggests that cancer, in and of itself, increases mortality from sepsis with dysregulation of the adaptive immune system playing a key role. Further, preclinical data demonstrate that sepsis can alter subsequent tumor growth while tumoral immunity impacts survival from sepsis. Checkpoint inhibition is a well-accepted treatment for many types of cancer, and there is increasing evidence suggesting this may be a useful strategy in sepsis as well. However, preclinical studies of checkpoint inhibition in cancer and sepsis demonstrate results that could not have been predicted by examining either variable in isolation. As sepsis management transitions from a 'one size fits all' model to a more individualized approach, understanding the mechanistic impact of cancer on outcomes from sepsis represents an important strategy towards delivering on the promise of precision medicine in the intensive care unit.
Collapse
Affiliation(s)
- Jeroson C. Williams
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, U.S.A
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA 30322, U.S.A
| | - Mandy L. Ford
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, U.S.A
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA 30322, U.S.A
| | - Craig M. Coopersmith
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, U.S.A
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA 30322, U.S.A
| |
Collapse
|
7
|
Rienzo M, Skirecki T, Monneret G, Timsit JF. Immune checkpoint inhibitors for the treatment of sepsis:insights from preclinical and clinical development. Expert Opin Investig Drugs 2022; 31:885-894. [PMID: 35944174 DOI: 10.1080/13543784.2022.2102477] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Sepsis represents one-fifth of all deaths worldwide and is associated with huge costs. Regarding disease progression, it is now well established that sepsis induces a state of acquired immunosuppression, with an increased risk of secondary infections that contributes to patients' worsening. Thus, tackling sepsis-induced immunosuppression represents a promising perspective. AREAS COVERED Of mechanisms responsible for sepsis-induced immunosuppression, the increased expression of co-inhibitory receptors (aka immune checkpoint) such as PD-1, CTLA4, TIM-3, LAG-3 or BTLA and their ligands recently received considerable interest since their inhibition, thanks to the so-called checkpoint inhibitors (CPI), provided astonishing results in cancer by rebooting immune functions. This review reports on the first landmarks of these molecules in sepsis. We introduce them in terms of basic immunology in line with sepsis pathophysiology both in experimental models and observational works and assess the first human clinical studies. EXPERT OPINION Preclinical results are positive and the first human clinical trials, although currently limited to the early phase, showed a beneficial effect on immunological functions and/or markers and suggested that tolerance of CPIs side effects, mainly auto-immune disorders, is acceptable in sepsis. Elsewhere, in some specific infections leading to ICU admission (or occurring during ICU stay), such as fungal infections, preliminary convincing case reports have been published. Overall, the first results regarding CPIs in sepsis appear encouraging. However, further efforts are warranted, especially in defining the right patients to be treated (i.e., in an individualized approach) and establishing the optimal time to start an immune restoration. Larger trials are now mandatory to confirm CPIs' potential in sepsis.
Collapse
Affiliation(s)
- Mario Rienzo
- AP-HP, Bichat Hospital, Medical and infectious diseases ICU (MI2), F-75018 Paris, France
| | - Tomasz Skirecki
- Laboratory of Flow Cytometry, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Guillaume Monneret
- Immunology Laboratory, Hôpital E. Herriot, Hospices Civils de Lyon, Lyon, F-69003.,Université de Lyon, EA7426, Hôpital E. Herriot, Lyon, F-69003
| | - Jean-François Timsit
- AP-HP, Bichat Hospital, Medical and infectious diseases ICU (MI2), F-75018 Paris, France.,University of Paris, IAME, INSERM, F-75018 Paris, France
| |
Collapse
|
8
|
Gao YL, Yao Y, Zhang X, Chen F, Meng XL, Chen XS, Wang CL, Liu YC, Tian X, Shou ST, Chai YF. Regulatory T Cells: Angels or Demons in the Pathophysiology of Sepsis? Front Immunol 2022; 13:829210. [PMID: 35281010 PMCID: PMC8914284 DOI: 10.3389/fimmu.2022.829210] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
Sepsis is a syndrome characterized by life-threatening organ dysfunction caused by the dysregulated host response to an infection. Sepsis, especially septic shock and multiple organ dysfunction is a medical emergency associated with high morbidity, high mortality, and prolonged after-effects. Over the past 20 years, regulatory T cells (Tregs) have been a key topic of focus in all stages of sepsis research. Tregs play a controversial role in sepsis based on their heterogeneous characteristics, complex organ/tissue-specific patterns in the host, the multi-dimensional heterogeneous syndrome of sepsis, the different types of pathogenic microbiology, and even different types of laboratory research models and clinical research methods. In the context of sepsis, Tregs may be considered both angels and demons. We propose that the symptoms and signs of sepsis can be attenuated by regulating Tregs. This review summarizes the controversial roles and Treg checkpoints in sepsis.
Collapse
Affiliation(s)
- Yu-lei Gao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Yan-fen Chai, ; Yu-lei Gao,
| | - Ying Yao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiang Zhang
- Department of Emergency Medicine, Rizhao People’s Hospital of Shandong Province, Rizhao, China
| | - Fang Chen
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiang-long Meng
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin-sen Chen
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Chao-lan Wang
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan-cun Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Tian
- Department of Medical Research, Beijing Qiansong Technology Development Company, Beijing, China
| | - Song-tao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan-fen Chai
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Yan-fen Chai, ; Yu-lei Gao,
| |
Collapse
|
9
|
Immune checkpoint molecule TIGIT manipulates T cell dysfunction in septic patients. Int Immunopharmacol 2021; 101:108205. [PMID: 34653731 DOI: 10.1016/j.intimp.2021.108205] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 11/23/2022]
Abstract
Sepsis is a dysregulated host response to infection. T cell dysfunction results in the failure to eradicate pathogens and the increased susceptibility to nosocomial infections and mortality during sepsis. Although PD-1 has shown to be a promising target to interfere with T cells dysfunction, the role of other coinhibitory receptors in sepsis remains largely elusive. Here we demonstrated that the immune checkpoint molecule TIGIT on lymphocytes and the critical role of TIGIT in regulating T cell responses in sepsis. Fifty septic patients and seventeen healthy donors were prospectively enrolled. The expression patterns of TIGIT and other molecules on lymphocytes were quantitated by flow cytometry. Ex vivo functional assays were also conducted. Results show that TIGIT expression on T cells was significantly upregulated in sepsis and septic shock patients relative to healthy donors. Elevated frequencies of TIGIT+ T cells correlated with aggravated inflammatory response and organ injuries. Of note, TIGIT expression on CD8+ T cells showed a competitive capability to predict ICU mortality in sepsis. TIGIT+ T cells expressed higher levels of PD-1, lower levels of CD226, and released fewer cytokines. Strikingly, ex vivo blockade of TIGIT using anti-TIGIT antibody restored the frequencies of cytokine-producing T cells from septic patients. These data illustrate that TIGIT on T cells is being used not only as a clinical predictor of poor prognosis but also as a potential target of novel immunotherapeutic intervention during sepsis.
Collapse
|
10
|
Sjaastad FV, Jensen IJ, Berton RR, Badovinac VP, Griffith TS. Inducing Experimental Polymicrobial Sepsis by Cecal Ligation and Puncture. ACTA ACUST UNITED AC 2021; 131:e110. [PMID: 33027848 DOI: 10.1002/cpim.110] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Numerous models are available for the preclinical study of sepsis, and they fall into one of three general categories: (1) administration of exogenous toxins (e.g., lipopolysaccharide, zymosan), (2) virulent bacterial or viral challenge, and (3) host barrier disruption, e.g., cecal ligation and puncture (CLP) or colon ascendens stent peritonitis (CASP). Of the murine models used to study the pathophysiology of sepsis, CLP combines tissue necrosis and polymicrobial sepsis secondary to autologous fecal leakage, as well as hemodynamic and biochemical responses similar to those seen in septic humans. Further, a transient numerical reduction of multiple immune cell types, followed by development of prolonged immunoparalysis, occurs in CLP-induced sepsis just as in humans. Use of the CLP model has led to a vast expansion in knowledge regarding the intricate physiological and cellular changes that occur during and after a septic event. This updated article details the steps necessary to perform this survival surgical technique, as well as some of the obstacles that may arise when evaluating the sepsis-induced changes within the immune system. It also provides representative monoclonal antibody (mAb) panels for multiparameter flow cytometric analysis of the murine immune system in the septic host. © 2020 Wiley Periodicals LLC. Basic Protocol: Cecal ligation and puncture in the mouse.
Collapse
Affiliation(s)
- Frances V Sjaastad
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, Minnesota
| | - Isaac J Jensen
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa
| | - Roger R Berton
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa
| | - Vladimir P Badovinac
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa.,Department of Pathology, University of Iowa, Iowa City, Iowa.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa
| | - Thomas S Griffith
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, Minnesota.,Department of Urology, University of Minnesota, Minneapolis, Minnesota.,Center for Immunology, University of Minnesota, Minneapolis, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Minneapolis VA Health Care System, Minneapolis, Minnesota
| |
Collapse
|
11
|
Zhang W, Anyalebechi JC, Ramonell KM, Chen CW, Xie J, Liang Z, Chihade DB, Otani S, Coopersmith CM, Ford ML. TIGIT modulates sepsis-induced immune dysregulation in mice with preexisting malignancy. JCI Insight 2021; 6:e139823. [PMID: 34100383 PMCID: PMC8262279 DOI: 10.1172/jci.insight.139823] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/05/2021] [Indexed: 12/29/2022] Open
Abstract
TIGIT is a recently identified coinhibitory receptor that is upregulated in the setting of cancer and functionally contributes to the impairment of antitumor immunity. However, its role during sepsis is unknown. Because patients with cancer are 10 times more likely to die of sepsis than previously healthy (PH) patients with sepsis, we interrogated the role of TIGIT during sepsis in the context of preexistent malignancy. PH mice or cancer (CA) mice inoculated with lung carcinoma cells were made septic by cecal ligation and puncture (CLP). We found that sepsis induced TIGIT upregulation predominantly on Tregs and NK cells in both PH and CA mice. Anti-TIGIT Ab improved the 7-d survival of CA septic mice but not PH mice after CLP. Treatment of CA septic animals but not PH septic animals with anti-TIGIT mAb significantly reversed sepsis-induced loss of CD4+ T cells, CD8+ T cells, Foxp3+ Treg, and CD19+ B cells in the spleen, which was the result of decreased caspase-3+ apoptotic cells. In sum, we found that anti-TIGIT Ab reversed sepsis-induced T cell apoptosis in CA septic mice and led to a significant survival benefit, suggesting its use as a potential immunotherapy to improve outcomes in septic patients with cancer.
Collapse
Affiliation(s)
- Wenxiao Zhang
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA.,Department of Critical Care Medicine, People's Hospital of Zhengzhou University (Henan Provincial People's Hospital), Zhengzhou, China
| | - Jerome C Anyalebechi
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kimberly M Ramonell
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ching-Wen Chen
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jianfeng Xie
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA.,Department of Critical Care Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Zhe Liang
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Deena B Chihade
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Shunsuke Otani
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA.,Department of General Medical Science, Chiba University Graduate School of Medicine, Chiba, Japan.,Department of Emergency and Critical Care Medicine, Eastern Chiba Medical Center, Togane, Japan
| | - Craig M Coopersmith
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA.,Emory Critical Care Center and
| | - Mandy L Ford
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA.,Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
12
|
Chen CW, Bennion KB, Swift DA, Morrow KN, Zhang W, Oami T, Coopersmith CM, Ford ML. Tumor-Specific T Cells Exacerbate Mortality and Immune Dysregulation during Sepsis. THE JOURNAL OF IMMUNOLOGY 2021; 206:2412-2419. [PMID: 33911005 DOI: 10.4049/jimmunol.2000865] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 03/08/2021] [Indexed: 11/19/2022]
Abstract
Sepsis induces significant immune dysregulation characterized by lymphocyte apoptosis and alterations in the cytokine milieu. Because cancer patients exhibit a 10-fold greater risk of developing sepsis compared with the general population, we aimed to understand how pre-existing malignancy alters sepsis-induced immune dysregulation. To address this question, we assessed the impact of tumor-specific CD8+ T cells on the immune response in a mouse model of cecal ligation and puncture (CLP)-induced sepsis. Tumor-bearing animals containing Thy1.1+ tumor-specific CD8+ T cells were subjected to CLP, and groups of animals received anti-Thy1.1 mAb to deplete tumor-specific CD8+ T cells or isotype control. Results indicated that depleting tumor-specific T cells significantly improved mortality from sepsis. The presence of tumor-specific CD8+ T cells resulted in increased expression of the 2B4 coinhibitory receptor and increased apoptosis of endogenous CD8+ T cells. Moreover, tumor-specific T cells were not reduced in number in the tumors during sepsis but did exhibit impaired IFN-γ production in the tumor, tumor draining lymph node, and spleen 24 h after CLP. Our research provides novel insight into the mechanisms by which pre-existing malignancy contributes to increased mortality during sepsis.
Collapse
Affiliation(s)
- Ching-Wen Chen
- Immunology and Molecular Pathogenesis Graduate Program, Laney Graduate School, Emory University, Atlanta, GA.,Department of Surgery, Emory University School of Medicine, Atlanta, GA
| | - Kelsey B Bennion
- Department of Surgery, Emory University School of Medicine, Atlanta, GA.,Cancer Biology Graduate Program, Laney Graduate School, Emory University, Atlanta, GA
| | - David A Swift
- Department of Surgery, Emory University School of Medicine, Atlanta, GA
| | - Kristen N Morrow
- Immunology and Molecular Pathogenesis Graduate Program, Laney Graduate School, Emory University, Atlanta, GA.,Department of Surgery, Emory University School of Medicine, Atlanta, GA
| | - Wenxiao Zhang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA.,Department of Critical Care Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Takehiko Oami
- Department of Surgery, Emory University School of Medicine, Atlanta, GA.,Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Craig M Coopersmith
- Department of Surgery, Emory University School of Medicine, Atlanta, GA .,Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA
| | - Mandy L Ford
- Department of Surgery, Emory University School of Medicine, Atlanta, GA .,Emory Transplant Center, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
13
|
Bu Y, Wang H, Ma X, Han C, Jia X, Zhang J, Liu Y, Peng Y, Yang M, Yu K, Wang C. Untargeted Metabolomic Profiling of the Correlation Between Prognosis Differences and PD-1 Expression in Sepsis: A Preliminary Study. Front Immunol 2021; 12:594270. [PMID: 33868224 PMCID: PMC8046931 DOI: 10.3389/fimmu.2021.594270] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 02/24/2021] [Indexed: 12/14/2022] Open
Abstract
Objectives: The mortality rate of sepsis remains very high. Metabolomic techniques are playing increasingly important roles in diagnosis and treatment in critical care medicine. The purpose of our research was to use untargeted metabolomics to identify and analyze the common differential metabolites among patients with sepsis with differences in their 7-day prognosis and blood PD-1 expression and analyze their correlations with environmental factors. Methods: Plasma samples from 18 patients with sepsis were analyzed by untargeted LC-MS metabolomics. Based on the 7-day prognoses of the sepsis patients or their levels of PD-1 expression on the surface of CD4+ T cells in the blood, we divided the patients into two groups. We used a combination of multidimensional and monodimensional methods for statistical analysis. At the same time, the Spearman correlation analysis method was used to analyze the correlation between the differential metabolites and inflammatory factors. Results: In the positive and negative ionization modes, 16 and 8 differential metabolites were obtained between the 7-day death and survival groups, respectively; 5 and 8 differential metabolites were obtained between the high PD-1 and low PD-1 groups, respectively. We identified three common differential metabolites from the two groups, namely, PC (P-18:0/14:0), 2-ethyl-2-hydroxybutyric acid and glyceraldehyde. Then, we analyzed the correlations between environmental factors and the common differences in metabolites. Among the identified metabolites, 2-ethyl-2-hydroxybutyric acid was positively correlated with the levels of IL-2 and lactic acid (Lac) (P < 0.01 and P < 0.05, respectively). Conclusions: These three metabolites were identified as common differential metabolites between the 7-day prognosis groups and the PD-1 expression level groups of sepsis patients. They may be involved in regulating the expression of PD-1 on the surface of CD4+ T cells through the action of related environmental factors such as IL-2 or Lac, which in turn affects the 7-day prognosis of sepsis patients.
Collapse
Affiliation(s)
- Y Bu
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin, China
| | - H Wang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - X Ma
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - C Han
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - X Jia
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - J Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Y Liu
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin, China
| | - Y Peng
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - M Yang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - K Yu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - C Wang
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
14
|
Kumar S, Singh SK, Rana B, Rana A. Tumor-infiltrating CD8 + T cell antitumor efficacy and exhaustion: molecular insights. Drug Discov Today 2021; 26:951-967. [PMID: 33450394 PMCID: PMC8131230 DOI: 10.1016/j.drudis.2021.01.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/20/2020] [Accepted: 01/07/2021] [Indexed: 02/06/2023]
Abstract
Host immunity has an essential role in the clinical management of cancers. Therefore, it is advantageous to choose therapies that can promote tumor cell death and concurrently boost host immunity. The dynamic tumor microenvironment (TME) determines whether an antineoplastic drug will elicit favorable or disparaging immune responses from tumor-infiltrating lymphocytes (TILs). CD8+ T cells are one of the primary tumor-infiltrating immune cells that deliver antitumor responses. Here, we review the influence of various factors in the TME on CD8+ T cell exhaustion and survival, and possible strategies for restoring CD8+ T cell effector function through immunotherapy.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA.
| | - Sunil Kumar Singh
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA
| | - Basabi Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA; University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA; University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
15
|
Sun Y, Anyalebechi JC, Sun H, Yumoto T, Xue M, Liu D, Liang Z, Coopersmith CM, Ford ML. Anti-TIGIT differentially affects sepsis survival in immunologically experienced versus previously naive hosts. JCI Insight 2021; 6:141245. [PMID: 33682797 PMCID: PMC8021109 DOI: 10.1172/jci.insight.141245] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 01/20/2021] [Indexed: 12/29/2022] Open
Abstract
Mounting evidence suggests that the balance of T cell costimulatory and coinhibitory signals contributes to mortality during sepsis. Here, we identified a critical role of the coinhibitory molecule T cell Ig and ITIM domain (TIGIT) in regulating sepsis mortality. Because TIGIT is significantly upregulated on memory T cells, we developed a "memory mouse" model to study the role of TIGIT during sepsis in a more physiologically relevant context. Mice received sequential pathogen exposure and developed memory T cell frequencies, similar to those observed in adult humans, and were then subjected to sepsis induction via cecal ligation and puncture. Our results show that targeting the TIGIT pathway during sepsis is fundamentally different in previously naive versus memory mice, in that αTIGIT Ab had no effect on survival in previously naive septic mice but sharply worsened survival in memory septic mice. Mechanistically, αTIGIT increased apoptosis of memory T cells, decreased T cell function, and downregulated the costimulatory receptor DNAM on memory CD8+ T cells in memory septic mice, but not in previously naive septic mice. Additionally, αTIGIT diminished Helios expression in Tregs in memory but not previously naive septic mice. These data highlight fundamental differences in the pathophysiological impact of targeting TIGIT in immunologically experienced versus previously naive hosts during sepsis.
Collapse
Affiliation(s)
- Yini Sun
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jerome C. Anyalebechi
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - He Sun
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Hepatobiliary Surgery and Transplantation, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
- Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Tetsuya Yumoto
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ming Xue
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Danya Liu
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Zhe Liang
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Craig M. Coopersmith
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mandy L. Ford
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
16
|
Morrow KN, Liang Z, Xue M, Chihade DB, Sun Y, Chen CW, Coopersmith CM, Ford ML. The IL-27 receptor regulates TIGIT on memory CD4 + T cells during sepsis. iScience 2021; 24:102093. [PMID: 33615199 PMCID: PMC7881227 DOI: 10.1016/j.isci.2021.102093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/19/2020] [Accepted: 01/18/2021] [Indexed: 12/19/2022] Open
Abstract
Sepsis is a leading cause of morbidity and mortality associated with significant impairment in memory T cells. These changes include the upregulation of co-inhibitory markers, a decrease in functionality, and an increase in apoptosis. Due to recent studies describing IL-27 regulation of TIGIT and PD-1, we assessed whether IL-27 impacts these co-inhibitory molecules in sepsis. Based on these data, we hypothesized that IL-27 was responsible for T cell dysfunction during sepsis. Using the cecal ligation and puncture (CLP) sepsis model, we found that IL-27Rα was associated with the upregulation of TIGIT on memory CD4+ T cells following CLP. However, IL-27 was not associated with sepsis mortality. Numbers of IL-27Rα+ memory T cells are decreased following cecal ligation and puncture TIGIT is expressed on more IL-27Rα+ versus IL-27Rα− memory CD4+ T cells during sepsis Il27ra−/− and WT T cells exhibit similar effector function and apoptosis during sepsis IL-27 signaling does not impact sepsis mortality
Collapse
Affiliation(s)
- Kristen N Morrow
- Immunology and Molecular Pathogenesis Program, Laney Graduate School, Emory University, Atlanta, GA 30324, USA.,Department of Surgery, Emory University School of Medicine, Atlanta, GA 30324, USA
| | - Zhe Liang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30324, USA
| | - Ming Xue
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30324, USA.,Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Deena B Chihade
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30324, USA
| | - Yini Sun
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30324, USA.,Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang 110000, China
| | - Ching-Wen Chen
- Immunology and Molecular Pathogenesis Program, Laney Graduate School, Emory University, Atlanta, GA 30324, USA.,Department of Surgery, Emory University School of Medicine, Atlanta, GA 30324, USA
| | - Craig M Coopersmith
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30324, USA.,Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA 30324, USA
| | - Mandy L Ford
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30324, USA.,Emory Transplant Center, Emory University School of Medicine, Atlanta, GA 30324, USA
| |
Collapse
|
17
|
Jensen IJ, Jensen SN, Sjaastad FV, Gibson-Corley KN, Dileepan T, Griffith TS, Mangalam AK, Badovinac VP. Sepsis impedes EAE disease development and diminishes autoantigen-specific naive CD4 T cells. eLife 2020; 9:55800. [PMID: 33191915 PMCID: PMC7721438 DOI: 10.7554/elife.55800] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 11/15/2020] [Indexed: 12/14/2022] Open
Abstract
Evaluation of sepsis-induced immunoparalysis has highlighted how decreased lymphocyte number/function contribute to worsened infection/cancer. Yet, an interesting contrast exists with autoimmune disease development, wherein diminishing pathogenic effectors may benefit the post-septic host. Within this framework, the impact of cecal ligation and puncture (CLP)-induced sepsis on the development of experimental autoimmune encephalomyelitis (EAE) was explored. Notably, CLP mice have delayed onset and reduced disease severity, relative to sham mice. Reduction in disease severity was associated with reduced number, but not function, of autoantigen (MOG)-specific pathogenic CD4 T cells in the CNS during disease and draining lymph node during priming. Numerical deficits of CD4 T cell effectors are associated with the loss of MOG-specific naive precursors. Critically, transfer of MOG-TCR transgenic (2D2) CD4 T cells after, but not before, CLP led to EAE disease equivalent to sham mice. Thus, broad impairment of antigenic responses, including autoantigens, is a hallmark of sepsis-induced immunoparalysis. Sepsis is a life-threatening condition that can happen when the immune system overreacts to an infection and begins to damage tissues and organs in the body. It causes an extreme immune reaction called a cytokine storm, where the body releases uncontrolled levels of cytokines, proteins that are involved in coordinating the body’s response to infections. This in turn activates more immune cells, resulting in hyperinflammation. People who survive sepsis may have long-lasing impairments in their immune system that may leave them more vulnerable to infections or cancer. But scientists do not know exactly what causes these lasting immune problems or how to treat them. The fact that people are susceptible to cancer and infection after sepsis may offer a clue. It may suggest that the immune system is not able to attack bacteria or cancer cells. One way to explore this clue would be to test the effects of sepsis on autoimmune diseases, which cause the immune system to attack the body’s own cells. For example, in the autoimmune disease multiple sclerosis, the immune system attacks and destroys cells in the nervous system. If autoimmune disease is reduced after sepsis, it would suggest the cell-destroying abilities of the immune system are lessened. Using this approach, Jensen, Jensen et al. show that sepsis reduces the number of certain immune cells, called CD4 T cells, which are are responsible for an autoimmune attack of the central nervous system. In the experiments, mice that survived sepsis were evaluated for their ability to develop a multiple sclerosis-like disease. Mice that survived sepsis developed less severe or no autoimmune disease. After sepsis, these animals also had fewer CD4 T cells. However, when these immune cells were reinstated, the autoimmune disease emerged. The experiments help explain some of the immune system changes that occur after sepsis. Jensen, Jensen et al. suggest that rather than being completely detrimental, these changes may help to block harmful autoimmune responses. The experiments may also hint at new ways to combat autoimmune diseases by trying to replicate some of the immune-suppressing effects of sepsis. Studying the effect of sepsis on other autoimmune diseases in mice might provide more clues.
Collapse
Affiliation(s)
- Isaac J Jensen
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, United States
| | - Samantha N Jensen
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, United States
| | - Frances V Sjaastad
- Microbiology, Immunology, and Cancer Biology PhD Program, University of Minnesota, Minneapolis, United States
| | - Katherine N Gibson-Corley
- Department of Pathology, University of Iowa, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, United States
| | - Thamothrampillai Dileepan
- Department of Microbiology and Immunology, University of Minnesota, Center for Immunology, Minneapolis, United States
| | - Thomas S Griffith
- Microbiology, Immunology, and Cancer Biology PhD Program, Department of Urology, Center for Immunology, Minneapolis VA Health Care System, University of Minnesota, Minneapolis, United States
| | - Ashutosh K Mangalam
- Interdisciplinary Graduate Program in Immunology, Department of Pathology, University of Iowa, Iowa City, United States
| | - Vladimir P Badovinac
- Interdisciplinary Graduate Program in Immunology, Department of Pathology, Department of Microbiology and Immunology, University of Iowa, Iowa City, United States
| |
Collapse
|
18
|
Martin MD, Badovinac VP, Griffith TS. CD4 T Cell Responses and the Sepsis-Induced Immunoparalysis State. Front Immunol 2020; 11:1364. [PMID: 32733454 PMCID: PMC7358556 DOI: 10.3389/fimmu.2020.01364] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022] Open
Abstract
Sepsis remains a major cause of death in the United States and worldwide, and costs associated with treating septic patients place a large burden on the healthcare industry. Patients who survive the acute phase of sepsis display long-term impairments in immune function due to reductions in numbers and function of many immune cell populations. This state of chronic immunoparalysis renders sepsis survivors increasingly susceptible to infection with newly or previously encountered infections. CD4 T cells play important roles in the development of cellular and humoral immune responses following infection. Understanding how sepsis impacts the CD4 T cell compartment is critical for informing efforts to develop treatments intended to restore immune system homeostasis following sepsis. This review will focus on the current understanding of how sepsis impacts the CD4 T cell responses, including numerical representation, repertoire diversity, phenotype and effector functionality, subset representation (e.g., Th1 and Treg frequency), and therapeutic efforts to restore CD4 T cell numbers and function following sepsis. Additionally, we will discuss recent efforts to model the acute sepsis phase and resulting immune dysfunction using mice that have previously encountered infection, which more accurately reflects the immune system of humans with a history of repeated infection throughout life. A thorough understanding of how sepsis impacts CD4 T cells based on previous studies and new models that accurately reflect the human immune system may improve translational value of research aimed at restoring CD4 T cell-mediated immunity, and overall immune fitness following sepsis.
Collapse
Affiliation(s)
- Matthew D. Martin
- Department of Urology, University of Minnesota, Minneapolis, MN, United States
| | - Vladimir P. Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA, United States
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Thomas S. Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN, United States
- Microbiology, Immunology, and Cancer Biology PhD Program, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Minneapolis VA Healthcare System, Minneapolis, MN, United States
| |
Collapse
|
19
|
Hargadon KM. Tumor microenvironmental influences on dendritic cell and T cell function: A focus on clinically relevant immunologic and metabolic checkpoints. Clin Transl Med 2020; 10:374-411. [PMID: 32508018 PMCID: PMC7240858 DOI: 10.1002/ctm2.37] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer immunotherapy is fast becoming one of the most promising means of treating malignant disease. Cancer vaccines, adoptive cell transfer therapies, and immune checkpoint blockade have all shown varying levels of success in the clinical management of several cancer types in recent years. However, despite the clinical benefits often achieved by these regimens, an ongoing problem for many patients is the inherent or acquired resistance of their cancer to immunotherapy. It is now appreciated that dendritic cells and T lymphocytes both play key roles in antitumor immune responses and that the tumor microenvironment presents a number of barriers to the function of these cells that can ultimately limit the success of immunotherapy. In particular, the engagement of several immunologic and metabolic checkpoints within the hostile tumor microenvironment can severely compromise the antitumor functions of these important immune populations. This review highlights work from both preclinical and clinical studies that has shaped our understanding of the tumor microenvironment and its influence on dendritic cell and T cell function. It focuses on clinically relevant targeted and immunotherapeutic strategies that have emerged from these studies in an effort to prevent or overcome immune subversion within the tumor microenvironment. Emphasis is also placed on the potential of next-generation combinatorial regimens that target metabolic and immunologic impediments to dendritic cell and T lymphocyte function as strategies to improve antitumor immune reactivity and the clinical outcome of cancer immunotherapy going forward.
Collapse
Affiliation(s)
- Kristian M. Hargadon
- Hargadon LaboratoryDepartment of BiologyHampden‐Sydney CollegeHampden‐SydneyVirginiaUSA
| |
Collapse
|