1
|
Lyu J, Narum DE, Baldwin SL, Larsen SE, Bai X, Griffith DE, Dartois V, Naidoo T, Steyn AJC, Coler RN, Chan ED. Understanding the development of tuberculous granulomas: insights into host protection and pathogenesis, a review in humans and animals. Front Immunol 2024; 15:1427559. [PMID: 39717773 PMCID: PMC11663721 DOI: 10.3389/fimmu.2024.1427559] [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: 05/04/2024] [Accepted: 11/18/2024] [Indexed: 12/25/2024] Open
Abstract
Granulomas, organized aggregates of immune cells which form in response to Mycobacterium tuberculosis (Mtb), are characteristic but not exclusive of tuberculosis (TB). Despite existing investigations on TB granulomas, the determinants that differentiate host-protective granulomas from granulomas that contribute to TB pathogenesis are often disputed. Thus, the goal of this narrative review is to help clarify the existing literature on such determinants. We adopt the a priori view that TB granulomas are host-protective organelles and discuss the molecular and cellular determinants that induce protective granulomas and those that promote their failure. While reports about protective TB granulomas and their failure may initially seem contradictory, it is increasingly recognized that either deficiencies or excesses of the molecular and cellular components in TB granuloma formation may be detrimental to the host. More specifically, insufficient or excessive expression/representation of the following components have been reported to skew granulomas toward the less protective phenotype: (i) epithelioid macrophages; (ii) type 1 adaptive immune response; (iii) type 2 adaptive immune response; (iv) tumor necrosis factor; (v) interleukin-12; (vi) interleukin-17; (vii) matrix metalloproteinases; (viii) hypoxia in the TB granulomas; (ix) hypoxia inducible factor-1 alpha; (x) aerobic glycolysis; (xi) indoleamine 2,3-dioxygenase activity; (xii) heme oxygenase-1 activity; (xiii) immune checkpoint; (xiv) leukotriene A4 hydrolase activity; (xv) nuclear-factor-kappa B; and (xvi) transforming growth factor-beta. Rather, more precise and timely coordinated immune responses appear essential for eradication or containment of Mtb infection. Since there are several animal models of infection with Mtb, other species within the Mtb complex, and the surrogate Mycobacterium marinum - whether natural (cattle, elephants) or experimental (zebrafish, mouse, guinea pig, rabbit, mini pig, goat, non-human primate) infections - we also compared the TB granulomatous response and other pathologic lung lesions in various animals infected with one of these mycobacteria with that of human pulmonary TB. Identifying components that dictate the formation of host-protective granulomas and the circumstances that result in their failure can enhance our understanding of the macrocosm of human TB and facilitate the development of novel remedies - whether they be direct therapeutics or indirect interventions - to efficiently eliminate Mtb infection and prevent its pathologic sequelae.
Collapse
Affiliation(s)
- Jiwon Lyu
- Division of Pulmonary and Critical Medicine, Soon Chun Hyang University Cheonan Hospital, Seoul, Republic of Korea
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
| | - Drew E. Narum
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
| | - Susan L. Baldwin
- Center for Global Infectious Diseases, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Sasha E. Larsen
- Center for Global Infectious Diseases, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Xiyuan Bai
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - David E. Griffith
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine, Nutley, NJ, United States
| | - Threnesan Naidoo
- Departments of Forensic & Legal Medicine and Laboratory Medicine & Pathology, Faculty of Medicine & Health Sciences, Walter Sisulu University, Mthatha, South Africa
| | - Adrie J. C. Steyn
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Microbiology and Centers for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rhea N. Coler
- Center for Global Infectious Diseases, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Edward D. Chan
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| |
Collapse
|
2
|
Arbous SM, Termorshuizen F, Brinkman S, de Lange DW, Bosman RJ, Dekkers OM, de Keizer NF. Three-year mortality of ICU survivors with sepsis, an infection or an inflammatory illness: an individually matched cohort study of ICU patients in the Netherlands from 2007 to 2019. Crit Care 2024; 28:374. [PMID: 39563453 PMCID: PMC11577713 DOI: 10.1186/s13054-024-05165-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 11/08/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Sepsis is a frequent reason for ICU admission and a leading cause of death. Its incidence has been increasing over the past decades. While hospital mortality is decreasing, it is recognized that the sequelae of sepsis extend well beyond hospitalization and are associated with a high mortality rate that persists years after hospitalization. The aim of this study was to disentangle the relative contribution of sepsis (infection with multi-organ failure), of infection and of inflammation, as reasons for ICU admission to long-term survival. This was done as infection and inflammation are both cardinal features of sepsis. We assessed the 3-year mortality of ICU patients admitted with sepsis, with individually matched ICU patients with an infection but not sepsis, and with an inflammatory illness not caused by infection, discharged alive from hospital. METHODS A multicenter cohort study of adult ICU survivors admitted between January 1st 2007 and January 1st 2019, with sepsis, an infection or an inflammatory illness. Patients were classified within the first 24 h of ICU admission according to APACHE IV admission diagnoses. Dutch ICUs (n = 78) prospectively recorded demographic and clinical data of all admissions in the NICE registry. These data were linked to a health care insurance claims database to obtain 3-year mortality data. To better understand and distinct the sepsis cohort from the non-sepsis infection and inflammatory condition cohorts, we performed several sensitivity analyses with varying definitions of the infection and inflammatory illness cohort. RESULTS Three-year mortality after discharge was 32.7% in the sepsis (N = 10,000), 33.6% in the infectious (N = 10,000), and 23.8% in the inflammatory illness cohort (N = 9997). Compared with sepsis patients, the adjusted HR for death within 3 years after hospital discharge was 1.00 (95% CI 0.95-1.05) for patients with an infection and 0.88 (95% CI 0.83-0.94) for patients with an inflammatory illness. CONCLUSIONS Both sepsis and non-sepsis infection patients had a significantly increased hazard rate of death in the 3 years after hospital discharge compared with patients with an inflammatory illness. Among sepsis and infection patients, one third died in the next 3 years, approximately 10% more than patients with an inflammatory illness. The fact that we did not find a difference between patients with sepsis or an infection suggests that the necessity for an ICU admission with an infection increases the risk of long-term mortality. This result emphasizes the need for greater attention to the post-ICU management of sepsis, infection, and severe inflammatory illness survivors.
Collapse
Affiliation(s)
- Sesmu M Arbous
- Department of Intensive Care, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.
- Department of Clinical Epidemiology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.
- NICE (National Intensive Care Evaluation) Foundation, Amsterdam, The Netherlands.
| | - Fabian Termorshuizen
- Department of Medical Informatics, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
- NICE (National Intensive Care Evaluation) Foundation, Amsterdam, The Netherlands
- Amsterdam Public Health, Digital Health and Quality of Care, Amsterdam, The Netherlands
| | - Sylvia Brinkman
- Department of Medical Informatics, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
- NICE (National Intensive Care Evaluation) Foundation, Amsterdam, The Netherlands
- Amsterdam Public Health, Digital Health and Quality of Care, Amsterdam, The Netherlands
| | - Dylan W de Lange
- Department of Intensive Care, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
- NICE (National Intensive Care Evaluation) Foundation, Amsterdam, The Netherlands
| | - Rob J Bosman
- Department of Intensive Care, OLVG, Amsterdam, The Netherlands
- NICE (National Intensive Care Evaluation) Foundation, Amsterdam, The Netherlands
| | - Olaf M Dekkers
- Department of Clinical Epidemiology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Nicolette F de Keizer
- Department of Medical Informatics, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
- NICE (National Intensive Care Evaluation) Foundation, Amsterdam, The Netherlands
- Amsterdam Public Health, Digital Health and Quality of Care, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Hayashi M, Yoshikawa T, Mizusawa J, Hato S, Iwasaki Y, Sasako M, Kawachi Y, Iishi H, Choda Y, Boku N, Terashima M. Prognostic Impact of Post-operative Infectious Complications in Gastric Cancer Patients Receiving Neoadjuvant Chemotherapy: Post Hoc Analysis of a Randomized Controlled Trial, JCOG0501. J Gastrointest Cancer 2024; 55:1125-1133. [PMID: 38703333 DOI: 10.1007/s12029-024-01061-3] [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] [Accepted: 04/20/2024] [Indexed: 05/06/2024]
Abstract
PURPOSE Post-operative infectious complication (IC) is a well-known negative prognostic factor, while showing neoadjuvant chemotherapy (NAC) may cancel out the negative influence of IC. This analysis compared the clinical impacts of IC according to the presence or absence of NAC in gastric cancer patients enrolled in the phase III clinical trial (JCOG0501) which compared upfront surgery (arm A) and NAC followed by surgery (arm B) in type 4 and large type 3 gastric cancer. METHODS The subjects were 224 patients who underwent R0 resection out of 316 patients enrolled in JCOG0501. The prognoses of the patients with or without ICs in each arm were investigated by univariable and multivariable Cox regression analyses. RESULTS There were 21 (20.0%) IC occurrences in arm A and 15 (12.6%) in arm B. In arm A, the overall survival (OS) of patients with ICs was slightly worse than those without IC (3-year OS, 57.1% in patients with ICs, 79.8% in those without ICs; adjusted hazard ratio (95% confidence interval), 1.292 (0.655-2.546)). In arm B, patients with ICs showed a trend of better survival than those without ICs (3-year OS, 80.0% in patients with IC, 74.0% in those without IC; adjusted hazard ratio, 0.573 (0.226-1.456)). CONCLUSION This study could not indicate the negative prognostic influence of ICs in gastric cancer patients receiving NAC, which might be canceled by NAC. To build exact evidence, further investigation with prospective and large numbers of data might be expected.
Collapse
Affiliation(s)
- Masato Hayashi
- Department of Surgery, Tochigi Cancer Center, Utsunomiya, Tochigi, Japan
| | - Takaki Yoshikawa
- Department of Gastric Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| | - Junki Mizusawa
- JCOG Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Shinji Hato
- Department of Gastroenterological Surgery, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Yoshiaki Iwasaki
- Department of Surgery, IMS Tokyo Katsushika General Hospital, Tokyo, Japan
| | - Mitsuru Sasako
- Department of Surgery, Yodogawa Christian Hospital, Osaka, Japan
| | - Yasuyuki Kawachi
- Department of Surgery, Nagaoka Chuo General Hospital, Niigata, Japan
| | - Hiroyasu Iishi
- Department of Gastrointestinal Oncology, Osaka International Cancer Institute, Osaka, Japan
- Department of Gastroenterology, Itami City Hospital, Itami, Japan
| | - Yasuhiro Choda
- Department of Surgery, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Narikazu Boku
- Department of Medical Oncology and General Medicine, IMSUT Hospital, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
4
|
Christensen LM, Akimova T, Wang L, Han R, Samanta A, Di Giorgio E, Hancock WW. T-regulatory cells require Sin3a for stable expression of Foxp3. Front Immunol 2024; 15:1444937. [PMID: 39156895 PMCID: PMC11327135 DOI: 10.3389/fimmu.2024.1444937] [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: 06/06/2024] [Accepted: 07/08/2024] [Indexed: 08/20/2024] Open
Abstract
Histone deacetylases 1 and 2 play a major role in the transcriptional regulation of T-regulatory (Treg) cells via interactions with a myriad of coregulatory factors. Sin3a has been well established as a Hdac1/2 cofactor, while its role within Tregs has not been established. In this study, the effects of conditional deletion of Sin3a within Foxp3+ Tregs were evaluated. Developmental deletion of Sin3a from Foxp3+ Tregs resulted in the rapid onset of fatal autoimmunity. Treg numbers were greatly reduced, while residual Tregs had impaired suppressive function. Mice also showed effector T-cell activation, autoantibody production, and widespread tissue injury. Mechanistically, Sin3a deletion resulted in decreased transcription of Foxp3 with a complete lack of CNS2 CpG demethylation. In addition, Foxp3 protein stability was impaired with an increased ex-Treg population. Thus, Sin3a plays a critical role in the maintenance of Treg identity and function and is essential for the expression and stability of Foxp3.
Collapse
Affiliation(s)
- Lanette M. Christensen
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Tatiana Akimova
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Liqing Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Rongxiang Han
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Arabinda Samanta
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | | | - Wayne W. Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
5
|
Liu X, Chen L, Peng W, Deng H, Ni H, Tong H, Hu H, Wang S, Qian J, Liang A, Chen K. Th17/Treg balance: the bloom and wane in the pathophysiology of sepsis. Front Immunol 2024; 15:1356869. [PMID: 38558800 PMCID: PMC10978743 DOI: 10.3389/fimmu.2024.1356869] [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: 12/16/2023] [Accepted: 02/20/2024] [Indexed: 04/04/2024] Open
Abstract
Sepsis is a multi-organ dysfunction characterized by an unregulated host response to infection. It is associated with high morbidity, rapid disease progression, and high mortality. Current therapies mainly focus on symptomatic treatment, such as blood volume supplementation and antibiotic use, but their effectiveness is limited. Th17/Treg balance, based on its inflammatory property, plays a crucial role in determining the direction of the inflammatory response and the regression of organ damage in sepsis patients. This review provides a summary of the changes in T-helper (Th) 17 cell and regulatory T (Treg) cell differentiation and function during sepsis, the heterogeneity of Th17/Treg balance in the inflammatory response, and the relationship between Th17/Treg balance and organ damage. Th17/Treg balance exerts significant control over the bloom and wanes in host inflammatory response throughout sepsis.
Collapse
Affiliation(s)
- Xinyong Liu
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Longwang Chen
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei Peng
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Hongsheng Deng
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Hongying Ni
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Hongjie Tong
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Hangbo Hu
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Shengchao Wang
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jin Qian
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Andong Liang
- Nursing Faculty, School of Medicine, Jinhua Polytechnic, Jinhua, China
| | - Kun Chen
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
6
|
Yang J, Chen J, Zhang M, Zhou Q, Yan B. Prognostic impacts of repeated sepsis in intensive care unit on autoimmune disease patients: a retrospective cohort study. BMC Infect Dis 2024; 24:197. [PMID: 38350868 PMCID: PMC10863122 DOI: 10.1186/s12879-024-09072-y] [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: 07/24/2023] [Accepted: 01/29/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Autoimmune diseases (ADs) may be complicated by sepsis when intensive care unit (ICU) admission. But repeated sepsis among AD patients has not been studied yet. The aim of this study is to investigate the impact of repeated in-ICU sepsis on the 1-year overall-cause mortality, septic shock and in-ICU death of AD patients. METHODS Data of AD patients with sepsis retrieved from Medical Information Mart for Intensive Care IV (MIMIC-IV) database were divided into the single group and the repeated group according to the frequency of in-ICU sepsis. Propensity score matching was used to balance inter-group bias. Cox proportional hazard regression and sensitivity analysis were utilized to assess the variables on mortality. RESULTS The incidence of repeated in-ICU sepsis in baseline was 19.8%. The repeated in-ICU sepsis was a risk factor for 1-year overall-cause mortality among AD patients (adjusted hazard ratio [HR] = 1.50, 95% CI: 1.16-1.93, P = 0.002), with robust adjusted HRs by the adjustment for confounders in the sensitivity analysis (all P < 0.01). Maximum Sequential Organ Failure Assessment (Max SOFA), Charlson comorbidity index (CCI) and Simplified Acute Physiology Score-II (SAPS-II) were risk factors for 1-year overall-cause mortality among AD with repeated sepsis (Max SOFA: HR = 1.09, P = 0.002; CCI: HR = 1.08, P = 0.039; SAPS-II: HR = 1.03, P < 0.001). CONCLUSIONS Compared to single hit, repeated in-ICU sepsis was independently related to a higher risk of 1-year overall-cause mortality among AD patients. Assessment tools (Higher SOFA, CCI and SAPS-II scores) were closely linked to poor prognosis of AD with repeated sepsis and helped to reflect ill physical conditions for the patients.
Collapse
Affiliation(s)
- Jinming Yang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue Alley, 610041, Chengdu, China
| | - Jie Chen
- Department of Rheumatology, People's Hospital of Leshan, Leshan, China
| | - Min Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue Alley, 610041, Chengdu, China
| | - Qingsa Zhou
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue Alley, 610041, Chengdu, China
| | - Bing Yan
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue Alley, 610041, Chengdu, China.
| |
Collapse
|
7
|
Bai X, Verma D, Garcia C, Musheyev A, Kim K, Fornis L, Griffith DE, Li L, Whittel N, Gadwa J, Ohanjanyan T, Eggleston MJ, Galvan M, Freed BM, Ordway D, Chan ED. Ex vivo and in vivo evidence that cigarette smoke-exposed T regulatory cells impair host immunity against Mycobacterium tuberculosis. Front Cell Infect Microbiol 2023; 13:1216492. [PMID: 37965256 PMCID: PMC10641287 DOI: 10.3389/fcimb.2023.1216492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/08/2023] [Indexed: 11/16/2023] Open
Abstract
Introduction A strong epidemiologic link exists between cigarette smoke (CS) exposure and susceptibility to tuberculosis (TB). Macrophage and murine studies showed that CS and nicotine impair host-protective immune cells against Mycobacterium tuberculosis (MTB) infection. While CS and nicotine may activate T regulatory cells (Tregs), little is known about how CS may affect these immunosuppressive cells with MTB infection. Methods We investigated whether CS-exposed Tregs could exacerbate MTB infection in co-culture with human macrophages and in recipient mice that underwent adoptive transfer of Tregs from donor CS-exposed mice. Results We found that exposure of primary human Tregs to CS extract impaired the ability of unexposed human macrophages to control an MTB infection by inhibiting phagosome-lysosome fusion and autophagosome formation. Neutralizing CTLA-4 on the CS extract-exposed Tregs abrogated the impaired control of MTB infection in the macrophage and Treg co-cultures. In Foxp3+GFP+DTR+ (Thy1.2) mice depleted of endogenous Tregs, adoptive transfer of Tregs from donor CS-exposed B6.PL(Thy1.1) mice with subsequent MTB infection of the Thy1.2 mice resulted in a greater burden of MTB in the lungs and spleens than those that received Tregs from air-exposed mice. Mice that received Tregs from donor CS-exposed mice and infected with MTB had modest but significantly reduced numbers of interleukin-12-positive dendritic cells and interferon-gamma-positive CD4+ T cells in the lungs, and an increased number of total programmed cell death protein-1 (PD-1) positive CD4+ T cells in both the lungs and spleens. Discussion Previous studies demonstrated that CS impairs macrophages and host-protective T effector cells in controlling MTB infection. We now show that CS-exposed Tregs can also impair control of MTB in co-culture with macrophages and in a murine model.
Collapse
Affiliation(s)
- Xiyuan Bai
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Department of Medicine, National Jewish Health, Denver, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Deepshikha Verma
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Cindy Garcia
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Ariel Musheyev
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Kevin Kim
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Lorelenn Fornis
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - David E. Griffith
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Li Li
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Nicholas Whittel
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Jacob Gadwa
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Tamara Ohanjanyan
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Matthew J. Eggleston
- Complement Laboratory, Advance Diagnostics, National Jewish Health, Denver, CO, United States
| | - Manuel Galvan
- Complement Laboratory, Advance Diagnostics, National Jewish Health, Denver, CO, United States
| | - Brian M. Freed
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Diane Ordway
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Edward D. Chan
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Department of Medicine, National Jewish Health, Denver, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| |
Collapse
|
8
|
Tripathi S, Tsang JS, Park K. Systems immunology of regulatory T cells: can one circuit explain it all? Trends Immunol 2023; 44:766-781. [PMID: 37690962 PMCID: PMC10543564 DOI: 10.1016/j.it.2023.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 09/12/2023]
Abstract
Regulatory T (Treg) cells play vital roles in immune homeostasis and response, including discrimination between self- and non-self-antigens, containment of immunopathology, and inflammation resolution. These diverse functions are orchestrated by cellular circuits involving Tregs and other cell types across space and time. Despite dramatic progress in our understanding of Treg biology, a quantitative framework capturing how Treg-containing circuits give rise to these diverse functions is lacking. Here, we propose that different facets of Treg function can be interpreted as distinct operating regimes of the same underlying circuit. We discuss how a systems immunology approach, involving quantitative experiments, computational modeling, and machine learning, can advance our understanding of Treg function, and help identify general operating and design principles underlying immune regulation.
Collapse
Affiliation(s)
- Shubham Tripathi
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA.
| | - John S Tsang
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA; Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA.
| | - Kyemyung Park
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA; Graduate School of Health Science and Technology and Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea.
| |
Collapse
|
9
|
Hästbacka J, But A, Strandberg G, Lipcsey M. Risk of malignant disease in 1-year sepsis survivors, a registry-based nationwide follow-up study. Crit Care 2023; 27:376. [PMID: 37773171 PMCID: PMC10543324 DOI: 10.1186/s13054-023-04654-9] [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: 08/15/2023] [Accepted: 09/19/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Cancer and sepsis share risk factors, and sepsis patients may have impaired immune response and increased morbidity long after intensive care. This study aimed to assess whether sepsis survivors are at increased risk for cancer. Our objective was to assess the incidence of new cancer in 1-year sepsis survivors and test the hypothesis that it is higher than that of the general population. METHODS We obtained data on ICU admissions of adult patients from Swedish Intensive care registry (SICR) from 2005 to 2017. We included patients with an explicit ICD-10 code for sepsis for the primary ICU admission. We obtained data on cancer diagnoses (2001-2018), death (2005-2018) and emigration (2005-2018) from Cancer and Cause of death and National Patient Registry databases of the National Board of Health and Welfare; age and sex-specific cancer incidence rates in Sweden from NORDCAN registry from 2006 to 2018. One-year survivors formed the final cohort, that was followed for new cancer diagnoses until death, emigration, or end of 2018, whichever came first. The main outcome measure was standardized incidence rate ratio (SIR) to compare the incidence of cancer in 1-year sepsis survivors to that in the general population (NORDCAN). We also performed several sensitivity analyses. RESULTS In a cohort of 18,550 1-year survivors, 75,427 person years accumulated during a median follow-up (FU) of 3.36 years (IQR 1.72-5.86), 6366 (34.3%) patients died, and 1625 (8.8%) patients were diagnosed with a new cancer after a median FU of 2.51 (IQR 1.09-4.48) years. The incidence ratio of any new cancer over the whole FU was 1.31 (95% CI 1.23-1.40) for men and 1.74 (95% CI 1.61-1.88) for women. The difference in incidence rates persisted in several sensitivity analyses. The SIRs were highest in cancers of gastrointestinal tract, genital organs, and skin. CONCLUSION AND RELEVANCE Compared to general population, incidence of cancer is increased in 1-year sepsis survivors. Variation in the findings depending on follow-up time suggests that factors other than sepsis alone are involved. Surveillance for malignant disease may be warranted in sepsis survivors.
Collapse
Affiliation(s)
- Johanna Hästbacka
- Department of Intensive Care, Tampere University, Faculty of Medicine and Health Technology, and Tampere University Hospital, Tampere, Finland.
| | - Anna But
- Biostatistics Consulting, Department of Public Health, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Gunnar Strandberg
- Section of Anesthesiology and Intensive Care, Department of Surgical Sciences, Uppsala University Hospital, Uppsala, Sweden
| | - Miklós Lipcsey
- Section of Anesthesiology and Intensive Care, Department of Surgical Sciences, Uppsala University Hospital, Uppsala, Sweden
- Hedenstierna Laboratory, Department of Surgical Science, Uppsala University, Uppsala, Sweden
| |
Collapse
|
10
|
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
|
11
|
Christensen LM, Hancock WW. Nuclear Coregulatory Complexes in Tregs as Targets to Promote Anticancer Immune Responses. Front Immunol 2022; 13:909816. [PMID: 35795673 PMCID: PMC9251111 DOI: 10.3389/fimmu.2022.909816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/18/2022] [Indexed: 12/17/2022] Open
Abstract
T-regulatory (Treg) cells display considerable heterogeneity in their responses to various cancers. The functional differences among this cell type are heavily influenced by multiprotein nuclear complexes that control their gene expression. Many such complexes act mechanistically by altering epigenetic profiles of genes important to Treg function, including the forkhead P3 (Foxp3) transcription factor. Complexes that form with certain members of the histone/protein deacetylase (HDAC) class of enzymes, like HDACs 1, 2, and 3, along with histone methyltransferase complexes, are important in the induction and stabilization of Foxp3 and Treg identity. The functional behavior of both circulating and intratumoral Tregs greatly impacts the antitumor immune response and can be predictive of patient outcome. Thus, targeting these regulatory complexes within Tregs may have therapeutic potential, especially in personalized immunotherapies.
Collapse
Affiliation(s)
- Lanette M. Christensen
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Wayne W. Hancock
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
- *Correspondence: Wayne W. Hancock,
| |
Collapse
|
12
|
Park J, Kang GH, Kim Y, Lee JY, Song JA, Hwang JH. Formaldehyde exposure induces differentiation of regulatory T cells via the NFAT-mediated T cell receptor signalling pathway in Yucatan minipigs. Sci Rep 2022; 12:8149. [PMID: 35581361 PMCID: PMC9114421 DOI: 10.1038/s41598-022-12183-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 05/05/2022] [Indexed: 12/31/2022] Open
Abstract
The use of minipigs (Sus scrofa) as a platform for toxicological and pharmacological research is well established. In the present study, we investigated the effect of formaldehyde (FA) exposure on helper T cell-mediated splenic immune responses in Yucatan minipigs. The minipigs were exposed to different inhaled concentrations of FA (0, 2.16, 4.62, or 10.48 mg/m3) for a period of 2 weeks. Immune responses elicited by exposure to FA were determined by assessing physiological parameters, mRNA expression, and cytokine production. Additionally, the distribution of helper T cells and regulatory T (Treg) cells and expression of NFAT families, which are well-known T cell receptor signalling proteins associated with regulatory T cell development, were evaluated. Exposure to FA suppressed the expression of genes associated with Th1 and Th2 cells in minipigs in a concentration-dependent manner. The subsequent production of cytokines also declined post-FA exposure. Furthermore, exposure to FA induced the differentiation of CD4+ Foxp3+ Treg cells with divergent expression levels of NFAT1 and NFAT2. These results indicated that exposure to FA increased the Treg cell population via the NFAT-mediated T cell receptor signalling pathway, leading to suppression of effector T cell activity with a decline in T cell-related cytokine production.
Collapse
Affiliation(s)
- Jeongsik Park
- Animal Model Research Group, Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, 30 Baehak 1-gil, Jeonguep, Jeollabuk-do, 56212, Republic of Korea
| | - Goo-Hwa Kang
- Animal Model Research Group, Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, 30 Baehak 1-gil, Jeonguep, Jeollabuk-do, 56212, Republic of Korea
| | - Youngkyu Kim
- Animal Model Research Group, Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, 30 Baehak 1-gil, Jeonguep, Jeollabuk-do, 56212, Republic of Korea.,Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul-si, 27447, Republic of Korea
| | - Ju Young Lee
- Animal Model Research Group, Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, 30 Baehak 1-gil, Jeonguep, Jeollabuk-do, 56212, Republic of Korea.,Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, 34113, Republic of Korea
| | - Jeong Ah Song
- Animal Model Research Group, Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, 30 Baehak 1-gil, Jeonguep, Jeollabuk-do, 56212, Republic of Korea
| | - Jeong Ho Hwang
- Animal Model Research Group, Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, 30 Baehak 1-gil, Jeonguep, Jeollabuk-do, 56212, Republic of Korea.
| |
Collapse
|
13
|
Weinberg SE, Singer BD. Toward a Paradigm to Distinguish Distinct Functions of FOXP3 + Regulatory T Cells. Immunohorizons 2021; 5:944-952. [PMID: 34893512 DOI: 10.4049/immunohorizons.2100046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/22/2021] [Indexed: 12/13/2022] Open
Abstract
FOXP3+ regulatory T (Treg) cells are a unique subset of CD4+ T cells that classically function as master regulators of immune homeostasis. Besides this canonical suppressive role, which is required to maintain self-tolerance, a growing body of literature has identified Treg cells as critical orchestrators of tissue protection during acute stress and as effector cells that drive repair following tissue injury. Despite substantial interest in these distinct roles, the field has struggled to disentangle Treg cell suppressive functions from those that promote tissue defense and repair. In this article, we will examine the literature in the context of specific physiologic settings, contrasting the suppressive function of Treg cells with their emerging roles in promoting tissue homeostasis and tissue repair. Further, we will discuss a new paradigm differentiating tissue defense from tissue repair-a paradigm needed to translate Treg cell-based therapies to the clinic.
Collapse
Affiliation(s)
- Samuel E Weinberg
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Benjamin D Singer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; .,Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL; and.,Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
14
|
Abstract
Sepsis is expected to have a substantial impact on public health and cost as its prevalence increases. Factors contributing to increased prevalence include a progressively aging population, advances in the use of immunomodulatory agents to treat a rising number of diseases, and immune-suppressing therapies in organ transplant recipients and cancer patients. It is now recognized that sepsis is associated with profound and sustained immunosuppression, which has been implicated as a predisposing factor in the increased susceptibility of patients to secondary infections and mortality. In this review, we discuss mechanisms of sepsis-induced immunosuppression and biomarkers that identify a state of impaired immunity. We also highlight immune-enhancing strategies that have been evaluated in patients with sepsis, as well as therapeutics under current investigation. Finally, we describe future challenges and the need for a new treatment paradigm, integrating predictive enrichment with patient factors that may guide the future selection of tailored immunotherapy. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Lisa K Torres
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, New York-Presbyterian Hospital-Weill Cornell Medicine, New York, NY, USA;
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands;
| | - Tom van der Poll
- Center of Experimental and Molecular Medicine, Division of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands;
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW This study reviews the mechanisms of HDL cholesterol immunomodulation in the context of the mechanisms of chronic inflammation and immunosuppression causing persistent inflammation, immunosuppression and catabolism syndrome (PICS) and describes potential therapies and gaps in current research. RECENT FINDINGS Low HDL cholesterol is predictive of acute sepsis severity and outcome. Recent research has indicated apolipoprotein is a prognostic indicator of long-term outcomes. The pathobiologic mechanisms of PICS have been elucidated in the past several years. Recent research of the interaction of HDL pathways in related chronic inflammatory diseases may provide insights into further mechanisms and therapeutic targets. SUMMARY HDL significantly influences innate and adaptive immune pathways relating to chronic disease and inflammation. Further research is needed to better characterize these interactions in the setting of PICS.
Collapse
Affiliation(s)
- Grant Barker
- Department of Emergency Medicine, University of Florida College of Medicine, Jacksonville
| | - Julia R Winer
- University of Florida College of Medicine, Gainesville, Florida
| | - Faheem W Guirgis
- Department of Emergency Medicine, University of Florida College of Medicine, Jacksonville
| | - Srinivasa Reddy
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, USA
| |
Collapse
|
16
|
Liu X, Yin L, Shen S, Hou Y. Inflammation and cancer: paradoxical roles in tumorigenesis and implications in immunotherapies. Genes Dis 2021; 10:151-164. [PMID: 37013041 PMCID: PMC10066281 DOI: 10.1016/j.gendis.2021.09.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/29/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation caused by persistent infections and metabolic disorders is thought to contribute to the increased cancer risk and the accelerated cancer progression. Oppositely, acute inflammation induced by bacteria-based vaccines or that is occurring after cancer selectively inhibits cancer progression and metastasis. However, the interaction between inflammation and cancer may be more complex than the current explanations for the relationship between chronic and acute inflammation and cancer. In this review, we described the impact of inflammation on cancer on the basis of three perspectives, including inflammation with different durations (chronic and acute inflammation), different scopes (systemic and local inflammation) and different occurrence sequences (inflammation occurring after and before cancer). In addition, we also introduced bacteria/virus-based cancer immunotherapies. We perceive that inflammation may be a double-edged sword with cancer-promoting and cancer-suppressing functions in certain cases. We expect to further improve the understanding of the relationship between inflammation and cancer and provide a theoretical basis for further research on their complex interaction.
Collapse
Affiliation(s)
- Xinghan Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Lijie Yin
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Sunan Shen
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
- Corresponding author. The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China. Fax: +86 25 8968 8441.
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
- Corresponding author. The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China. Fax: +86 25 8968 8441.
| |
Collapse
|
17
|
de Lima MHF, Hiroki CH, de Fátima Borges V, Cebinelli GCM, Santos J, Rosa MH, Silva CMS, Wanderley CWS, Gonçalves AV, Silva Quirino GF, Zamboni DS, Cunha TM, Filho JCA, Cunha FQ. Sepsis-induced immunosuppression is marked by an expansion of a highly suppressive repertoire of FOXP3 +T regulatory cells-expressing TIGIT. J Infect Dis 2021; 225:531-541. [PMID: 34379757 DOI: 10.1093/infdis/jiab405] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 08/10/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Although literature demonstrates that an increase in both the number and suppressive function of CD4+FOXP3+T regulatory cells (Tregs) during sepsis contributes to an immunosuppressed state, little is known about the identity of these cells. METHODS Using the sepsis mouse model of cecal ligation and puncture (CLP), we analysed the frequency and molecular signature of the TIGIT + and TIGIT- Tregs subsets by flow cytometry and qPCR. Additionally, ST2-/- and STAT6-/- mice were submitted to CLP or rIL-33 treatment to investigate the mechanism through which TIGIT+Tregs differentiate during sepsis. RESULTS Sepsis was marked by the sustained expansion of the highly suppressive TIGIT +Tregs subset, which expresses Helios, Neuropilin-1, and high levels of Tnfrsf18 and Pdcd1 at 15d after CLP. The increase of the TIGIT +Tregs was accompanied by higher susceptibility to nosocomial bacteria challenge, suggesting its involvement with post-sepsis immunosuppression. Mechanistically, we found that the ST2 deletion abrogated the expansion of the TIGIT+Treg subset during sepsis. Furthermore, the treatment with rIL-33 resulted in the expansion of TIGIT+Tregs depending on the STAT6 and M2 macrophages. CONCLUSIONS These findings demonstrated that only the TIGIT+Tregs remain stably expanded at the late phase of sepsis. Moreover, the expansion of TIGIT +Tregs is dependent on the IL-33/ST2/STAT6/M2 macrophages axis.
Collapse
Affiliation(s)
- Mikhael Haruo Fernandes de Lima
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Carlos Hiroji Hiroki
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Vanessa de Fátima Borges
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Guilherme Cesar Martelossi Cebinelli
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Jessica Santos
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marcos Henrique Rosa
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Camilla Meireles S Silva
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Carlos Wagner S Wanderley
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Augusto Velozo Gonçalves
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Gustavo Fernando Silva Quirino
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Dario S Zamboni
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thiago M Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - José-Carlos A Filho
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fernando Q Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
18
|
Miljković Đ, Stanisavljević S, Jensen IJ, Griffith TS, Badovinac VP. Sepsis and multiple sclerosis: Causative links and outcomes. Immunol Lett 2021; 238:40-46. [PMID: 34320384 DOI: 10.1016/j.imlet.2021.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 06/08/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022]
Abstract
Sepsis is a life-threatening condition characterized by an acute cytokine storm followed by prolonged dysfunction of the immune system in the survivors. Post-septic lymphopenia and functional deficits of the remaining immune cells lead to increased susceptibility to secondary infections and other morbid conditions causing late death in the patients. This state of post-septic immunoparalysis may also influence disorders stemming from inappropriate or overactive immune responses, such as autoimmune and immunoinflammatory diseases, including multiple sclerosis. In addition, ongoing autoimmunity likely influences the susceptibility to and outcome of sepsis. This review article addresses the bidirectional relationship between sepsis and multiple sclerosis, with a focus on the immunologic mechanisms of the interaction and potential directions for future studies.
Collapse
Affiliation(s)
- Đorđe Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia.
| | - Suzana Stanisavljević
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Isaac J Jensen
- Department of Pathology, Department of Microbiology and Immunology, Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Thomas S Griffith
- Microbiology, Immunology, and Cancer Biology PhD Program, Department of Urology, Center for Immunology, Masonic Cancer Center, University of Minnesota, Minneapolis VA Health Care System, Minneapolis, MN 55417, USA
| | - Vladimir P Badovinac
- Department of Pathology, Department of Microbiology and Immunology, Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
19
|
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
|
20
|
Gudiol C, Albasanz-Puig A, Cuervo G, Carratalà J. Understanding and Managing Sepsis in Patients With Cancer in the Era of Antimicrobial Resistance. Front Med (Lausanne) 2021; 8:636547. [PMID: 33869250 PMCID: PMC8044357 DOI: 10.3389/fmed.2021.636547] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/08/2021] [Indexed: 12/23/2022] Open
Abstract
Sepsis is a frequent complication in immunosuppressed cancer patients and hematopoietic stem cell transplant recipients that is associated with high morbidity and mortality rates. The worldwide emergence of antimicrobial resistance is of special concern in this population because any delay in starting adequate empirical antibiotic therapy can lead to poor outcomes. In this review, we aim to address: (1) the mechanisms involved in the development of sepsis and septic shock in these patients; (2) the risk factors associated with a worse prognosis; (3) the impact of adequate initial empirical antibiotic therapy given the current era of widespread antimicrobial resistance; and (4) the optimal management of sepsis, including adequate and early source control of infection, optimized antibiotic use based on the pharmacokinetic and pharmacodynamics changes in these patients, and the role of the new available antibiotics.
Collapse
Affiliation(s)
- Carlota Gudiol
- Infectious Diseases Department, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), University of Barcelona, Barcelona, Spain.,Institut Català d'Oncologia (ICO), Hospital Duran i Reynals, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain.,Spanish Network for Research in Infectious Diseases (REIPI RD16/0016/0001), Instituto de Salud Carlos III, Madrid, Spain
| | - Adaia Albasanz-Puig
- Infectious Diseases Department, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), University of Barcelona, Barcelona, Spain.,Spanish Network for Research in Infectious Diseases (REIPI RD16/0016/0001), Instituto de Salud Carlos III, Madrid, Spain
| | - Guillermo Cuervo
- Infectious Diseases Department, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), University of Barcelona, Barcelona, Spain.,Spanish Network for Research in Infectious Diseases (REIPI RD16/0016/0001), Instituto de Salud Carlos III, Madrid, Spain
| | - Jordi Carratalà
- Infectious Diseases Department, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), University of Barcelona, Barcelona, Spain.,Spanish Network for Research in Infectious Diseases (REIPI RD16/0016/0001), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
21
|
Leite CA, Mota JM, de Lima KA, Wanderley CW, Nascimento LA, Ferreira MD, Silva CMS, Colon DF, Sakita JY, Kannen V, Viacava PR, Begnami MD, Lima-Junior RCP, Cordeiro de Lima VC, Alves-Filho JC, Cunha FQ, Ribeiro RA. Paradoxical interaction between cancer and long-term postsepsis disorder: impairment of de novo carcinogenesis versus favoring the growth of established tumors. J Immunother Cancer 2021; 8:jitc-2019-000129. [PMID: 32376720 PMCID: PMC7223471 DOI: 10.1136/jitc-2019-000129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2020] [Indexed: 02/06/2023] Open
Abstract
Background Previous data have reported that the growth of established tumors may be facilitated by postsepsis disorder through changes in the microenvironment and immune dysfunction. However, the influence of postsepsis disorder in initial carcinogenesis remains elusive. Methods In the present work, the effect of postsepsis on inflammation-induced early carcinogenesis was evaluated in an experimental model of colitis-associated colorectal cancer (CAC). We also analyzed the frequency and role of intestinal T regulatory cells (Treg) in CAC carcinogenesis. Results The colitis grade and the tumor development rate were evaluated postmortem or in vivo through serial colonoscopies. Sepsis-surviving mice (SSM) presented with a lower colonic DNA damage, polyp incidence, reduced tumor load, and milder colitis than their sham-operated counterparts. Ablating Treg led to restoration of the ability to develop colitis and tumor polyps in the SSM, in a similar fashion to that in the sham-operated mice. On the other hand, the growth of subcutaneously inoculated MC38luc colorectal cancer cells or previously established chemical CAC tumors was increased in SSM. Conclusion Our results provide evidence that postsepsis disorder has a dual effect in cancer development, inhibiting inflammation-induced early carcinogenesis in a Treg-dependent manner, while increasing the growth of previously established tumors.
Collapse
Affiliation(s)
- Caio Abner Leite
- A.C. Camargo Cancer Center, Sao Paulo, Brazil.,Center for Research in Inflammatory Diseases (CRID), University of Sao Paulo, Ribeirao Preto, Brazil.,Cancer Institute of Ceara, Fortaleza, Brazil
| | - Jose Mauricio Mota
- Instituto do Cancer do Estado de Sao Paulo, University of Sao Paulo, Sao Paulo, Brazil
| | - Kalil Alves de Lima
- Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | | | | | | | | | - Juliana Yumi Sakita
- Department of Toxicology, Bromatology, and Clinical Analysis, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Vinicius Kannen
- Department of Toxicology, Bromatology, and Clinical Analysis, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Paula Ramos Viacava
- Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | | | | | | | - Fernando Queiroz Cunha
- Center for Research in Inflammatory Diseases (CRID), University of Sao Paulo, Ribeirao Preto, Brazil .,Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Ronaldo Albuquerque Ribeiro
- Cancer Institute of Ceara, Fortaleza, Brazil.,Federal University of Ceara, Faculty of Medicine, Fortaleza, Brazil
| |
Collapse
|
22
|
Mirouse A, Vigneron C, Llitjos JF, Chiche JD, Mira JP, Mokart D, Azoulay E, Pène F. Sepsis and Cancer: An Interplay of Friends and Foes. Am J Respir Crit Care Med 2020; 202:1625-1635. [PMID: 32813980 DOI: 10.1164/rccm.202004-1116tr] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Sepsis and cancer share a number of pathophysiological features, and both result from the inability of the host's immune system to cope with the initial insult (tissue invasion by pathogens and malignant cell transformation, respectively). The common coexistence of both disorders and the profound related alterations in immune homeostasis raise the question of their mutual impact on each other's course. This translational review aims to discuss the interactions between cancer and sepsis supported by clinical data and the translation to experimental models. The dramatic improvement in cancer has come at a cost of increased risks of life-threatening infectious complications. Investigating the long-term outcomes of sepsis survivors has revealed an unexpected susceptibility to cancer long after discharge from the ICU. Nonetheless, it is noteworthy that an acute septic episode may harbor antitumoral properties under particular circumstances. Relevant double-hit animal models have provided clues to whether and how bacterial sepsis may impact malignant tumor growth. In sequential sepsis-then-cancer models, postseptic mice exhibited accelerated tumor growth. When using reverse cancer-then-sepsis models, bacterial sepsis applied to mice with cancer conversely resulted in inhibition or even regression of tumor growth. Experimental models thus highlight dual effects of sepsis on tumor growth, mostly depending on the sequence of insults, and allow deciphering the immune mechanisms and their relation with microorganisms.
Collapse
Affiliation(s)
- Adrien Mirouse
- Université de Paris, Paris, France.,Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris, France.,Médecine Intensive et Réanimation, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP) Nord, Paris, France
| | - Clara Vigneron
- Université de Paris, Paris, France.,Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris, France.,Médecine Intensive et Réanimation, Hôpital Cochin, AP-HP Centre, Paris, France; and
| | - Jean-François Llitjos
- Université de Paris, Paris, France.,Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris, France.,Médecine Intensive et Réanimation, Hôpital Cochin, AP-HP Centre, Paris, France; and
| | - Jean-Daniel Chiche
- Université de Paris, Paris, France.,Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris, France.,Médecine Intensive et Réanimation, Hôpital Cochin, AP-HP Centre, Paris, France; and
| | - Jean-Paul Mira
- Université de Paris, Paris, France.,Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris, France.,Médecine Intensive et Réanimation, Hôpital Cochin, AP-HP Centre, Paris, France; and
| | - Djamel Mokart
- Réanimation Polyvalente, Département d'Anesthésie et de Réanimation, Institut Paoli Calmettes, Marseille, France
| | - Elie Azoulay
- Université de Paris, Paris, France.,Médecine Intensive et Réanimation, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP) Nord, Paris, France
| | - Frédéric Pène
- Université de Paris, Paris, France.,Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris, France.,Médecine Intensive et Réanimation, Hôpital Cochin, AP-HP Centre, Paris, France; and
| |
Collapse
|
23
|
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
|
24
|
Park J, Yang HS, Song MK, Kim DI, Lee K. Formaldehyde exposure induces regulatory T cell-mediated immunosuppression via calcineurin-NFAT signalling pathway. Sci Rep 2020; 10:17023. [PMID: 33046725 PMCID: PMC7550593 DOI: 10.1038/s41598-020-72502-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 04/15/2020] [Indexed: 11/08/2022] Open
Abstract
In this study, we investigated the effects of Formaldehyde (FA) exposure on splenic immune responses wherein helper T cells become activated and differentiate into effector T and regulatory T cells. BALB/c mice were exposed to two FA concentrations (1.38 mg/m3 and 5.36 mg/m3) for 4 h/day and 5 days/week for 2 weeks. FA-induced immune responses were examined by the production of cytokines, expression of mRNAs, and distributions of helper T cells and regulatory T cells. Moreover, expression of calcineurin and NFATs, regulatory T cell-related signalling proteins, were evaluated. FA exposure suppressed Th2-, Th1-, and Th17-related splenic cytokines in a dose-dependent manner. mRNA expression of splenic cytokines was also decreased by FA exposure, which correlated with decreased cytokine expression. In parallel, FA exposure promoted T cell differentiation into regulatory T cells in a dose-dependent manner supported by the expression of calcineurin and NFAT1. Taken together, our results indicated that FA exposure increases the number of regulatory T cells via calcineurin-NFAT signalling, thereby leading to effector T cell activity suppression with decreased T cell-related cytokine secretion and mRNA expression. These findings provide insight into the mechanisms underlying the adverse effects of FA and accordingly have general implications for human health, particularly in occupational settings.
Collapse
Affiliation(s)
- Jeongsik Park
- Jeonbuk Department of inhalation Research, National Center for Efficacy Evaluation of Respiratory Disease Products, Korea Institute of Toxicology, 30 Baehak 1-gil, Jeonguep, Jeollabuk-do, 56212, Republic of Korea
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Hyo-Seon Yang
- Jeonbuk Department of inhalation Research, National Center for Efficacy Evaluation of Respiratory Disease Products, Korea Institute of Toxicology, 30 Baehak 1-gil, Jeonguep, Jeollabuk-do, 56212, Republic of Korea
| | - Mi-Kyung Song
- Jeonbuk Department of inhalation Research, National Center for Efficacy Evaluation of Respiratory Disease Products, Korea Institute of Toxicology, 30 Baehak 1-gil, Jeonguep, Jeollabuk-do, 56212, Republic of Korea
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Dong Im Kim
- Jeonbuk Department of inhalation Research, National Center for Efficacy Evaluation of Respiratory Disease Products, Korea Institute of Toxicology, 30 Baehak 1-gil, Jeonguep, Jeollabuk-do, 56212, Republic of Korea
| | - Kyuhong Lee
- Jeonbuk Department of inhalation Research, National Center for Efficacy Evaluation of Respiratory Disease Products, Korea Institute of Toxicology, 30 Baehak 1-gil, Jeonguep, Jeollabuk-do, 56212, Republic of Korea.
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
25
|
van der Slikke EC, An AY, Hancock REW, Bouma HR. Exploring the pathophysiology of post-sepsis syndrome to identify therapeutic opportunities. EBioMedicine 2020; 61:103044. [PMID: 33039713 PMCID: PMC7544455 DOI: 10.1016/j.ebiom.2020.103044] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/09/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022] Open
Abstract
Sepsis is a major health problem worldwide. As the number of sepsis cases increases, so does the number of sepsis survivors who suffer from “post-sepsis syndrome” after hospital discharge. This syndrome involves deficits in multiple systems, including the immune, cognitive, psychiatric, cardiovascular, and renal systems. Combined, these detrimental consequences lead to rehospitalizations, poorer quality of life, and increased mortality. Understanding the pathophysiology of these issues is crucial to develop new therapeutic opportunities to improve survival rate and quality of life of sepsis survivors. Such novel strategies include modulating the immune system and addressing mitochondrial dysfunction. A sepsis follow-up clinic may be useful to identify long-term health issues associated with post-sepsis syndrome and evaluate existing and novel strategies to improve the lives of sepsis survivors.
Collapse
Affiliation(s)
- Elisabeth C van der Slikke
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, , P.O. Box 30.001, EB70, 9700 RB, Groningen, The Netherlands
| | - Andy Y An
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Hjalmar R Bouma
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, , P.O. Box 30.001, EB70, 9700 RB, Groningen, The Netherlands; Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
26
|
Rasid O, Chevalier C, Camarasa TMN, Fitting C, Cavaillon JM, Hamon MA. H3K4me1 Supports Memory-like NK Cells Induced by Systemic Inflammation. Cell Rep 2020; 29:3933-3945.e3. [PMID: 31851924 DOI: 10.1016/j.celrep.2019.11.043] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 09/06/2019] [Accepted: 11/08/2019] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells are unique players in innate immunity and, as such, an attractive target for immunotherapy. NK cells display immune memory properties in certain models, but the long-term status of NK cells following systemic inflammation is unknown. Here we show that following LPS-induced endotoxemia in mice, NK cells acquire cell-intrinsic memory-like properties, showing increased production of IFNγ upon specific secondary stimulation. The NK cell memory response is detectable for at least 9 weeks and contributes to protection from E. coli infection upon adoptive transfer. Importantly, we reveal a mechanism essential for NK cell memory, whereby an H3K4me1-marked latent enhancer is uncovered at the ifng locus. Chemical inhibition of histone methyltransferase activity erases the enhancer and abolishes NK cell memory. Thus, NK cell memory develops after endotoxemia in a histone methylation-dependent manner, ensuring a heightened response to secondary stimulation.
Collapse
Affiliation(s)
- Orhan Rasid
- G5 Chromatine et Infection, Institut Pasteur, Paris, France; Unité Cytokines & Inflammation, Institut Pasteur, Paris, France.
| | | | - Tiphaine Marie-Noelle Camarasa
- G5 Chromatine et Infection, Institut Pasteur, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | | | | |
Collapse
|
27
|
Sjaastad FV, Kucaba TA, Dileepan T, Swanson W, Dail C, Cabrera-Perez J, Murphy KA, Badovinac VP, Griffith TS. Polymicrobial Sepsis Impairs Antigen-Specific Memory CD4 T Cell-Mediated Immunity. Front Immunol 2020; 11:1786. [PMID: 32903436 PMCID: PMC7435018 DOI: 10.3389/fimmu.2020.01786] [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: 02/26/2020] [Accepted: 07/03/2020] [Indexed: 11/13/2022] Open
Abstract
Patients who survive sepsis display prolonged immune dysfunction and heightened risk of secondary infection. CD4 T cells support a variety of cells required for protective immunity, and perturbations to the CD4 T cell compartment can decrease overall immune system fitness. Using the cecal ligation and puncture (CLP) mouse model of sepsis, we investigated the impact of sepsis on endogenous Ag-specific memory CD4 T cells generated in C57BL/6 (B6) mice infected with attenuated Listeria monocytogenes (Lm) expressing the I-Ab-restricted 2W1S epitope (Lm-2W). The number of 2W1S-specific memory CD4 T cells was significantly reduced on day 2 after sepsis induction, but recovered by day 14. In contrast to the transient numerical change, the 2W1S-specific memory CD4 T cells displayed prolonged functional impairment after sepsis, evidenced by a reduced recall response (proliferation and effector cytokine production) after restimulation with cognate Ag. To define the extent to which the observed functional impairments in the memory CD4 T cells impacts protection to secondary infection, B6 mice were infected with attenuated Salmonella enterica-2W (Se-2W) 30 days before sham or CLP surgery, and then challenged with virulent Se-2W after surgery. Pathogen burden was significantly higher in the CLP-treated mice compared to shams. Similar reductions in functional capacity and protection were noted for the endogenous OVA323-specific memory CD4 T cell population in sepsis survivors upon Lm-OVA challenge. Our data collectively show CLP-induced sepsis alters the number and function of Ag-specific memory CD4 T cells, which contributes (in part) to the characteristic long-lasting immunoparalysis seen after sepsis.
Collapse
Affiliation(s)
- Frances V Sjaastad
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN, United States
| | - Tamara A Kucaba
- Department of Urology, University of Minnesota, Minneapolis, MN, United States
| | - Thamotharampillai Dileepan
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, United States.,Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Whitney Swanson
- Department of Urology, University of Minnesota, Minneapolis, MN, United States
| | - Cody Dail
- Medical Student Summer Research Program in Infection and Immunity, University of Minnesota, Minneapolis, MN, United States
| | - Javier Cabrera-Perez
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN, United States.,Medical Scientist Training Program, University of Minnesota, Minneapolis, MN, United States
| | - Katherine A Murphy
- Department of Urology, University of Minnesota, Minneapolis, MN, United States
| | - Vladimir P Badovinac
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States.,Department of Pathology, University of Iowa, Iowa City, IA, United States.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Thomas S Griffith
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN, United States.,Department of Urology, 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 Health Care System, Minneapolis, MN, United States
| |
Collapse
|
28
|
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: 93] [Impact Index Per Article: 18.6] [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
|
29
|
Wu C, Wang N, Zhou H, Wang T, Mao Q, Zhang X, Zhao D. Effects of Neoadjuvant Chemotherapy Toxicity and Postoperative Complications on Short-term and Long-term Outcomes After Curative Resection of Gastric Cancer. J Gastrointest Surg 2020; 24:1278-1289. [PMID: 31140064 DOI: 10.1007/s11605-019-04257-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 04/15/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Whether neoadjuvant chemotherapy (NAC) increased the risk of postoperative morbidities for patients with locally advanced gastric cancer (GC) is unknown. Whether neoadjuvant chemotherapy toxicity (NCT) and postoperative complications (POCs) correlate with short-term and long-term outcomes also remains unclear. We aimed to evaluate the role of NAC on the development of POCs, as well as the impact of NCT and POCs on postoperative and oncologic outcomes in curatively resected GC treated with NAC. METHODS This study retrospectively reviewed 230 patients who underwent curative gastrectomy for locally advanced GC (clinically T3/4 or N+) after NAC between 2006 and 2016. Five hundred patients undergoing upfront and curative surgery were selected as a control group. After matching, the incidence of POCs was compared between two groups. In the NAC group, clinicopathological characteristics of patients who experienced POCs were compared to those who did not. Logistic and Cox multivariate regression analyses were used to examine factors associated with POCs, disease-free survival (DFS), and overall survival (OS). RESULTS Following matching, 230 and 230 patients treated with surgery plus NAC and upfront surgery remained, respectively. The incidence of POCs was 28.7% and 24.3%, respectively (p = 0.290). In the NAC group, NCT (OR [odds ratio] 22.968, 95% CI [confidence interval] 2.948-> 99, p = 0.003) and operation time (OR 1.006, 95% CI 1.001-1.011, p = 0.021) were independent predictive factors of POCs. NCT did not affect oncologic outcomes. The Cox regression model demonstrated that POCs were independently associated with worse DFS (HR [hazard ratio] 2.128, 95% CI 1.240-3.653, p = 0.006) but not OS for patients treated with NAC. CONCLUSIONS The administration of NAC is not associated with an elevated risk of POCs. For patients treated with NAC, NCT is an independent predictor of POCs, but does not affect oncologic outcomes. POCs is independently associated with worse DFS but not OS. NAC should be considered a safe approach in patients who have locally advanced GC. Strategies to minimize chemotherapy toxicity and postoperative morbidities associated with NAC are warranted.
Collapse
Affiliation(s)
- Chaorui Wu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Nianchang Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hong Zhou
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tongbo Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qikun Mao
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiaojie Zhang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dongbing Zhao
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
30
|
Nicolai O, Pötschke C, Schmoeckel K, Darisipudi MN, van der Linde J, Raafat D, Bröker BM. Antibody Production in Murine Polymicrobial Sepsis-Kinetics and Key Players. Front Immunol 2020; 11:828. [PMID: 32425951 PMCID: PMC7205023 DOI: 10.3389/fimmu.2020.00828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/14/2020] [Indexed: 12/16/2022] Open
Abstract
Although antigen-specific priming of antibody responses is impaired during sepsis, there is nevertheless a strong increase in IgM and IgG serum concentrations. Using colon ascendens stent peritonitis (CASP), a mouse model of polymicrobial abdominal sepsis, we observed substantial increases in IgM as well as IgG of all subclasses, starting at day 3 and peaking 2 weeks after sepsis induction. The dominant source of antibody-secreting cells was by far the spleen, with a minor contribution of the mesenteric lymph nodes. Remarkably, sepsis induction in splenectomized mice did not change the dynamics of the serum IgM/IgG reaction, indicating that the marginal zone B cells, which almost exclusively reside in the spleen, are dispensable in such a setting. Hence, in systemic bacterial infection, the function of the spleen as dominant niche of antibody-producing cells can be compensated by extra-splenic B cell populations as well as other lymphoid organs. Depletion of CD4+ T cells did not affect the IgM response, while it impaired IgG generation of all subclasses with the exception of IgG3. Taken together, our data demonstrate that the robust class-switched antibody response in sepsis encompasses both T cell-dependent and -independent components.
Collapse
Affiliation(s)
- Oliver Nicolai
- Immunology Department, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Christian Pötschke
- Immunology Department, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Katrin Schmoeckel
- Immunology Department, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Murthy N Darisipudi
- Immunology Department, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Julia van der Linde
- Department of General Surgery, Visceral, Thoracic and Vascular Surgery, University Medicine Greifswald, Greifswald, Germany
| | - Dina Raafat
- Immunology Department, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany.,Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Barbara M Bröker
- Immunology Department, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
31
|
Beltrán-García J, Osca-Verdegal R, Romá-Mateo C, Carbonell N, Ferreres J, Rodríguez M, Mulet S, García-López E, Pallardó FV, García-Giménez JL. Epigenetic biomarkers for human sepsis and septic shock: insights from immunosuppression. Epigenomics 2020; 12:617-646. [PMID: 32396480 DOI: 10.2217/epi-2019-0329] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Sepsis is a life-threatening condition that occurs when the body responds to an infection damaging its own tissues. Sepsis survivors sometimes suffer from immunosuppression increasing the risk of death. To our best knowledge, there is no 'gold standard' for defining immunosuppression except for a composite clinical end point. As the immune system is exposed to epigenetic changes during and after sepsis, research that focuses on identifying new biomarkers to detect septic patients with immunoparalysis could offer new epigenetic-based strategies to predict short- and long-term pathological events related to this life-threatening state. This review describes the most relevant epigenetic mechanisms underlying alterations in the innate and adaptive immune responses described in sepsis and septic shock, and their consequences for immunosuppression states, providing several candidates to become epigenetic biomarkers that could improve sepsis management and help predict immunosuppression in postseptic patients.
Collapse
Affiliation(s)
- Jesús Beltrán-García
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Valencia 46010, Spain.,Department of Physiology, Faculty of Medicine & Dentistry, University of Valencia, Valencia 46010, Spain.,INCLIVA Biomedical Research Institute, Valencia 46010, Spain.,EpiDisease S.L. (Spin-Off CIBER-ISCIII), Parc Científic de la Universitat de València, Paterna 46980, Valencia, Spain
| | - Rebeca Osca-Verdegal
- Department of Physiology, Faculty of Medicine & Dentistry, University of Valencia, Valencia 46010, Spain
| | - Carlos Romá-Mateo
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Valencia 46010, Spain.,Department of Physiology, Faculty of Medicine & Dentistry, University of Valencia, Valencia 46010, Spain.,INCLIVA Biomedical Research Institute, Valencia 46010, Spain
| | - Nieves Carbonell
- INCLIVA Biomedical Research Institute, Valencia 46010, Spain.,Intensive Care Unit, Clinical University Hospital of Valencia, Valencia 46010, Spain
| | - José Ferreres
- INCLIVA Biomedical Research Institute, Valencia 46010, Spain.,Intensive Care Unit, Clinical University Hospital of Valencia, Valencia 46010, Spain
| | - María Rodríguez
- INCLIVA Biomedical Research Institute, Valencia 46010, Spain.,Intensive Care Unit, Clinical University Hospital of Valencia, Valencia 46010, Spain
| | - Sandra Mulet
- INCLIVA Biomedical Research Institute, Valencia 46010, Spain.,Intensive Care Unit, Clinical University Hospital of Valencia, Valencia 46010, Spain
| | - Eva García-López
- EpiDisease S.L. (Spin-Off CIBER-ISCIII), Parc Científic de la Universitat de València, Paterna 46980, Valencia, Spain
| | - Federico V Pallardó
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Valencia 46010, Spain.,Department of Physiology, Faculty of Medicine & Dentistry, University of Valencia, Valencia 46010, Spain.,INCLIVA Biomedical Research Institute, Valencia 46010, Spain
| | - José Luis García-Giménez
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Valencia 46010, Spain.,Department of Physiology, Faculty of Medicine & Dentistry, University of Valencia, Valencia 46010, Spain.,INCLIVA Biomedical Research Institute, Valencia 46010, Spain.,EpiDisease S.L. (Spin-Off CIBER-ISCIII), Parc Científic de la Universitat de València, Paterna 46980, Valencia, Spain
| |
Collapse
|
32
|
Cavaillon J, Singer M, Skirecki T. Sepsis therapies: learning from 30 years of failure of translational research to propose new leads. EMBO Mol Med 2020; 12:e10128. [PMID: 32176432 PMCID: PMC7136965 DOI: 10.15252/emmm.201810128] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/12/2020] [Accepted: 02/17/2020] [Indexed: 12/13/2022] Open
Abstract
Sepsis has been identified by the World Health Organization (WHO) as a global health priority. There has been a tremendous effort to decipher underlying mechanisms responsible for organ failure and death, and to develop new treatments. Despite saving thousands of animals over the last three decades in multiple preclinical studies, no new effective drug has emerged that has clearly improved patient outcomes. In the present review, we analyze the reasons for this failure, focusing on the inclusion of inappropriate patients and the use of irrelevant animal models. We advocate against repeating the same mistakes and propose changes to the research paradigm. We discuss the long-term consequences of surviving sepsis and, finally, list some putative approaches-both old and new-that could help save lives and improve survivorship.
Collapse
Affiliation(s)
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care MedicineUniversity College LondonLondonUK
| | - Tomasz Skirecki
- Laboratory of Flow Cytometry and Department of Anesthesiology and Intensive Care MedicineCentre of Postgraduate Medical EducationWarsawPoland
| |
Collapse
|
33
|
Salminen A. Activation of immunosuppressive network in the aging process. Ageing Res Rev 2020; 57:100998. [PMID: 31838128 DOI: 10.1016/j.arr.2019.100998] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/29/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022]
Abstract
Chronic low-grade inflammation has a key role in the aging process, a state called inflammaging. It is known that the chronic inflammatory condition generates counteracting immunosuppressive state in many diseases. Inflammaging is also associated with an immune deficiency; generally termed as immunosenescence, although it is not known whether it represents the senescence of immune cells or the active remodeling of immune system. Evidence has accumulated since the 1970's indicating that immunosenescence might be caused by an increased activity of immunosuppressive cells rather than cellular senescence. Immune cells display remarkable plasticity; many of these cells can express both proinflammatory and immunosuppressive phenotypes in a context-dependent manner. The immunosuppressive network involves the regulatory subtypes of T (Treg) and B (Breg) cells as well as regulatory phenotypes of macrophages (Mreg), dendritic (DCreg), natural killer (NKreg), and type II natural killer T (NKT) cells. The immunosuppressive network also includes monocytic (M-MDSC) and polymorphonuclear (PMN-MDSC) myeloid-derived suppressor cells which are immature myeloid cells induced by inflammatory mediators. This co-operative network is stimulated in chronic inflammatory conditions preventing excessive inflammatory responses but at the same time they exert harmful effects on the immune system and tissue homeostasis. Recent studies have revealed that the aging process is associated with the activation of immunosuppressive network, especially the functions of MDSCs, Tregs, and Mregs are increased. I will briefly review the properties of the regulatory phenotypes of immune cells and examine in detail the evidences for an activation of immunosuppressive network with aging.
Collapse
|
34
|
Grimaldi D, Pène F. Short- and Long-Term ICU-Acquired Immunosuppression. LESSONS FROM THE ICU 2020. [DOI: 10.1007/978-3-030-24250-3_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
35
|
Cao C, Yu M, Chai Y. Pathological alteration and therapeutic implications of sepsis-induced immune cell apoptosis. Cell Death Dis 2019; 10:782. [PMID: 31611560 PMCID: PMC6791888 DOI: 10.1038/s41419-019-2015-1] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 08/28/2019] [Accepted: 09/02/2019] [Indexed: 02/07/2023]
Abstract
Sepsis is a life-threatening organ dysfunction syndrome caused by dysregulated host response to infection that leads to uncontrolled inflammatory response followed by immunosuppression. However, despite the high mortality rate, no specific treatment modality or drugs with high efficacy is available for sepsis to date. Although improved treatment strategies have increased the survival rate during the initial state of excessive inflammatory response, recent trends in sepsis show that mortality occurs at a period of continuous immunosuppressive state in which patients succumb to secondary infections within a few weeks or months due to post-sepsis “immune paralysis.” Immune cell alteration induced by uncontrolled apoptosis has been considered a major cause of significant immunosuppression. Particularly, apoptosis of lymphocytes, including innate immune cells and adaptive immune cells, is associated with a higher risk of secondary infections and poor outcomes. Multiple postmortem studies have confirmed that sepsis-induced immune cell apoptosis occurs in all age groups, including neonates, pediatric, and adult patients, and it is considered to be a primary contributing factor to the immunosuppressive pathophysiology of sepsis. Therapeutic perspectives targeting apoptosis through various strategies could improve survival in sepsis. In this review article, we will focus on describing the major apoptosis process of immune cells with respect to physiologic and molecular mechanisms. Further, advances in apoptosis-targeted treatment modalities for sepsis will also be discussed.
Collapse
Affiliation(s)
- Chao Cao
- Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Medical University, Tianjin, China.,Department of Internal Medicine, The University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Muming Yu
- Tianjin Medical University General Hospital, Tianjin, China
| | - Yanfen Chai
- Tianjin Medical University General Hospital, Tianjin, China. .,Tianjin Medical University, Tianjin, China.
| |
Collapse
|
36
|
Abstract
OBJECTIVE To identify research priorities in the management, epidemiology, outcome and underlying causes of sepsis and septic shock. DESIGN A consensus committee of 16 international experts representing the European Society of Intensive Care Medicine and Society of Critical Care Medicine was convened at the annual meetings of both societies. Subgroups had teleconference and electronic-based discussion. The entire committee iteratively developed the entire document and recommendations. METHODS Each committee member independently gave their top five priorities for sepsis research. A total of 88 suggestions (Supplemental Table 1, Supplemental Digital Content 2, http://links.lww.com/CCM/D636) were grouped into categories by the committee co-chairs, leading to the formation of seven subgroups: infection, fluids and vasoactive agents, adjunctive therapy, administration/epidemiology, scoring/identification, post-intensive care unit, and basic/translational science. Each subgroup had teleconferences to go over each priority followed by formal voting within each subgroup. The entire committee also voted on top priorities across all subgroups except for basic/translational science. RESULTS The Surviving Sepsis Research Committee provides 26 priorities for sepsis and septic shock. Of these, the top six clinical priorities were identified and include the following questions: 1) can targeted/personalized/precision medicine approaches determine which therapies will work for which patients at which times?; 2) what are ideal endpoints for volume resuscitation and how should volume resuscitation be titrated?; 3) should rapid diagnostic tests be implemented in clinical practice?; 4) should empiric antibiotic combination therapy be used in sepsis or septic shock?; 5) what are the predictors of sepsis long-term morbidity and mortality?; and 6) what information identifies organ dysfunction? CONCLUSIONS While the Surviving Sepsis Campaign guidelines give multiple recommendations on the treatment of sepsis, significant knowledge gaps remain, both in bedside issues directly applicable to clinicians, as well as understanding the fundamental mechanisms underlying the development and progression of sepsis. The priorities identified represent a roadmap for research in sepsis and septic shock.
Collapse
|
37
|
Vigneron C, Mirouse A, Merdji H, Rousseau C, Cousin C, Alby-Laurent F, Mira JP, Chiche JD, Llitjos JF, Pène F. Sepsis inhibits tumor growth in mice with cancer through Toll-like receptor 4-associated enhanced Natural Killer cell activity. Oncoimmunology 2019; 8:e1641391. [PMID: 31646090 DOI: 10.1080/2162402x.2019.1641391] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/26/2019] [Accepted: 07/03/2019] [Indexed: 12/12/2022] Open
Abstract
Sepsis-induced immune dysfunctions are likely to impact on malignant tumor growth. Sequential sepsis-then-cancer models of tumor transplantation in mice recovering from sepsis have shown that the post-septic immunosuppressive environment was able to promote tumor growth. We herein addressed the impact of sepsis on pre-established malignancy in a reverse cancer-then sepsis experimental model. Mice previously inoculated with MCA205 fibrosarcoma cells were subjected to septic challenges by polymicrobial peritonitis induced by cecal ligation and puncture or endotoxinic shock. The anti-tumoral immune response was assessed through the distribution of tumor-infiltrating immune cells, as well as the functions of cytotoxic cells. As compared to sham surgery, polymicrobial sepsis dampened malignant tumor growth in wild-type (WT) mice, but neither in Toll-like receptor 4 (Tlr4)-/- nor in Myd88-/- mice. Similar tumor growth inhibition was observed following a LPS challenge in WT mice, suggesting a regulatory role of Tlr4 in this setting. The low expression of MHC class 1 onto MCA205 cells suggested the involvement of Natural Killer (NK) cells in sepsis-induced tumor inhibition. Septic insults applied to mice with cancer promoted the main anti-tumoral NK functions of IFNγ production and degranulation. The anti-tumoral properties of NK cells obtained from septic mice were exacerbated when cultured with MHC1low MCA205 or YAC-1 cells. These results suggest that sepsis may harbor dual effects on tumor growth depending on the sequential experimental model. When applied in mice with cancer, sepsis prevents tumor growth in a Tlr4-dependent manner by enhancing the anti-tumoral functions of NK cells.
Collapse
Affiliation(s)
- Clara Vigneron
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Adrien Mirouse
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Hamid Merdji
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Christophe Rousseau
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Clément Cousin
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Fanny Alby-Laurent
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Jean-Paul Mira
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France.,Médecine Intensive & Réanimation, hôpital Cochin, Hôpitaux Universitaires Paris Centre, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Jean-Daniel Chiche
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France.,Médecine Intensive & Réanimation, hôpital Cochin, Hôpitaux Universitaires Paris Centre, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Jean-François Llitjos
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France.,Médecine Intensive & Réanimation, hôpital Cochin, Hôpitaux Universitaires Paris Centre, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Frédéric Pène
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France.,Médecine Intensive & Réanimation, hôpital Cochin, Hôpitaux Universitaires Paris Centre, Assistance Publique - Hôpitaux de Paris, Paris, France
| |
Collapse
|
38
|
Darkwah S, Nago N, Appiah MG, Myint PK, Kawamoto E, Shimaoka M, Park EJ. Differential Roles of Dendritic Cells in Expanding CD4 T Cells in Sepsis. Biomedicines 2019; 7:biomedicines7030052. [PMID: 31323786 PMCID: PMC6783955 DOI: 10.3390/biomedicines7030052] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 01/01/2023] Open
Abstract
Sepsis is a systemically dysregulated inflammatory syndrome, in which dendritic cells (DCs) play a critical role in coordinating aberrant immunity. The aim of this study is to shed light on the differential roles played by systemic versus mucosal DCs in regulating immune responses in sepsis. We identified a differential impact of the systemic and mucosal DCs on proliferating allogenic CD4 T cells in a mouse model of sepsis. Despite the fact that the frequency of CD4 T cells was reduced in septic mice, septic mesenteric lymph node (MLN) DCs proved superior to septic spleen (SP) DCs in expanding allogeneic CD4 T cells. Moreover, septic MLN DCs markedly augmented the surface expression of MHC class II and CD40, as well as the messaging of interleukin-1β (IL-1β). Interestingly, IL-1β-treated CD4 T cells expanded in a dose-dependent manner, suggesting that this cytokine acts as a key mediator of MLN DCs in promoting septic inflammation. Thus, mucosal and systemic DCs were found to be functionally different in the way CD4 T cells respond during sepsis. Our study provides a molecular basis for DC activity, which can be differential in nature depending on location, whereby it induces septic inflammation or immune-paralysis.
Collapse
Affiliation(s)
- Samuel Darkwah
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Nodoka Nago
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
- Department of Clinical Nutrition, Suzuka University of Medical Science, Suzuka, Mie 510-0293, Japan
| | - Michael G Appiah
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Phyoe Kyawe Myint
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Eiji Kawamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
- Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
| |
Collapse
|
39
|
Danahy DB, Kurup SP, Winborn CS, Jensen IJ, Harty JT, Griffith TS, Badovinac VP. Sepsis-Induced State of Immunoparalysis Is Defined by Diminished CD8 T Cell-Mediated Antitumor Immunity. THE JOURNAL OF IMMUNOLOGY 2019; 203:725-735. [PMID: 31189573 DOI: 10.4049/jimmunol.1900435] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 05/27/2019] [Indexed: 01/15/2023]
Abstract
Patients who survive sepsis experience long-term immunoparalysis characterized by numerical and/or functional lesions in innate and adaptive immunity that increase the host's susceptibility to secondary complications. The extent to which tumor development/growth is affected in sepsis survivors remains unknown. In this study, we show cecal ligation and puncture (CLP) surgery renders mice permissive to increased B16 melanoma growth weeks/months after sepsis induction. CD8 T cells provide partial protection in this model, and tumors from sepsis survivors had a reduced frequency of CD8 tumor-infiltrating lymphocytes (TILs) concomitant with an increased tumor burden. Interestingly, the postseptic environment reduced the number of CD8 TILs with high expression of activating/inhibitory receptors PD-1 and LAG-3 (denoted PD-1hi) that define a tumor-specific CD8 T cell subset that retain some functional capacity. Direct ex vivo analysis of CD8 TILs from CLP hosts showed decreased proliferation, IFN-γ production, and survival compared with sham counterparts. To increase the frequency and/or functional capacity of PD-1hi CD8 TILs in tumor-bearing sepsis survivors, checkpoint blockade therapy using anti-PD-L1/anti-LAG-3 mAb was administered before or after the development of sepsis-induced lesions in CD8 TILs. Checkpoint blockade did not reduce tumor growth in CLP hosts when therapy was administered after PD-1hi CD8 TILs had become reduced in frequency and/or function. However, early therapeutic intervention before lesions were observed significantly reduced tumor growth to levels seen in nonseptic hosts receiving therapy. Thus, sepsis-induced immunoparalysis is defined by diminished CD8 T cell-mediated antitumor immunity that can respond to timely checkpoint blockade, further emphasizing the importance of early cancer detection in hosts that survive sepsis.
Collapse
Affiliation(s)
- Derek B Danahy
- Department of Pathology, University of Iowa, Iowa City, IA 52242.,Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Samarchith P Kurup
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242; and
| | | | - Isaac J Jensen
- Department of Pathology, University of Iowa, Iowa City, IA 52242.,Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - John T Harty
- Department of Pathology, University of Iowa, Iowa City, IA 52242.,Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242; and
| | - Thomas S Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN 55414
| | - Vladimir P Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA 52242; .,Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242; and
| |
Collapse
|
40
|
Jensen IJ, Sjaastad FV, Griffith TS, Badovinac VP. Sepsis-Induced T Cell Immunoparalysis: The Ins and Outs of Impaired T Cell Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 200:1543-1553. [PMID: 29463691 DOI: 10.4049/jimmunol.1701618] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022]
Abstract
Sepsis results in a deluge of pro- and anti-inflammatory cytokines, leading to lymphopenia and chronic immunoparalysis. Sepsis-induced long-lasting immunoparalysis is defined, in part, by impaired CD4 and CD8 αβ T cell responses in the postseptic environment. The dysfunction in T cell immunity affects naive, effector, and memory T cells and is not restricted to classical αβ T cells. Although sepsis-induced severe and transient lymphopenia is a contributory factor to diminished T cell immunity, T cell-intrinsic and -extrinsic factors/mechanisms also contribute to impaired T cell function. In this review, we summarize the current knowledge of how sepsis quantitatively and qualitatively impairs CD4 and CD8 T cell immunity of classical and nonclassical T cell subsets and discuss current therapeutic approaches being developed to boost the recovery of T cell immunity postsepsis induction.
Collapse
Affiliation(s)
- Isaac J Jensen
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Frances V Sjaastad
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN 55455
| | - Thomas S Griffith
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN 55455.,Center for Immunology, University of Minnesota, Minneapolis, MN 55455.,Department of Urology, University of Minnesota, Minneapolis, MN 55455.,Minneapolis VA Health Care System, Minneapolis, MN 55455
| | - Vladimir P Badovinac
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242; .,Department of Pathology, University of Iowa, Iowa City, IA 52242; and.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
41
|
Vicente D, Patino M, Marcus R, Lillmoe H, Limani P, Newhook T, Lee A, Tzeng CW, Segraves-Chun Y, Tweardy D, Gottumukkala V, Vauthey JN, Aloia T, Cata JP. Impact of epidural analgesia on the systemic biomarker response after hepatic resection. Oncotarget 2019; 10:584-594. [PMID: 30728909 PMCID: PMC6355178 DOI: 10.18632/oncotarget.26549] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/22/2018] [Indexed: 12/30/2022] Open
Abstract
Background Perioperative inflammation is associated with poor oncologic outcomes. Regional analgesia has been shown mitigate some of these inflammatory changes and be associated with better oncologic outcomes in patients with hepatic malignancies. The mechanism for this effect, however, remains unclear. The authors sought to compare systemic biomarker concentrations in a comprehensive and oncologically relevant panel in the perioperative setting between patients undergoing thoracic epidural analgesia (TEA) and intra-venous patient- controlled analgesia (IV-PCA) for resection of hepatic metastatic disease. Results Clinicopathologic variables and baseline biomarkers were similar between TEA (n = 46) and IV-PCA (n = 16) groups. Of the biomarkers which were significantly changed from baseline, there was a lower fold change from baseline in the TEA patients compared to IV-PCA including IL-6 (13.5vs19.1), MCP-1 (1.9vs3.0), IL-8 (2.4vs3.0), and Pentraxin-3 (10.8vs15.6). Overall decreased systemic concentrations of TGFb signaling were noted in TEA patients on POD1 TGFb3 (243.2 vs. 86.0, p = 0.005), POD3 TGFb1 (6558.0 vs. 2063.3, p = 0.004), POD3 TGFb2 (468.3 vs. 368.9, p = 0.036), POD3 TGFb3 (132.2 vs. 77.8, p = 0.028), and POD5 TGFb3 (306.5 vs. 92.2, p = 0.032). POD1 IL-12p70 concentrations were significantly higher in TEA patients (8.3 vs. 1.6, p = 0.024). Conclusion Epidural analgesia damped the postoperative inflammatory response and systemic immunosuppressive signaling, as well as promoted Th1 systemic signaling early in the post-operative period after hepatic resection for metastatic disease. These differences elaborate on known mechanisms for improved oncologic outcomes with regional anesthesia, and may be considered for biomarker monitoring of effective regional anesthesia in oncologic surgery. Materials and Methods Patient data, including clinicopathologic variables were collected for this study from the database of a randomized controlled trial comparing perioperative outcomes in patients undergoing hepatic resection with TEA vs. IV-PCA. Patients undergoing resection for metastatic disease were selected for this study. Plasma concentrations (pg/mL) of well-studied biomarkers (IL-1b/2/4/5/6/7/8/10/12p70/13/17, MCP-1 IFNγ, TNFα, MIP-1b, GM-CSF, G-CSF, VEGF, Resistin, TGFb1, TGFb2, and TGFb3), as well as novel perioperative markers (CXCL12, CXCL10, Omentin-1, sLeptin R, Vaspin, Pentraxin-3, Galactin-3, FGF-23, PON-1, FGF-21) were measured preoperatively, and on postoperative day (POD)1, POD3, and POD5 using multiplex bead assays. Clinicopathologic variables and perioperative variations in these biomarkers were compared between TEA vs IV-PCA groups.
Collapse
Affiliation(s)
- Diego Vicente
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA
| | - Miguel Patino
- Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA.,Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rebecca Marcus
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heather Lillmoe
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Preparim Limani
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy Newhook
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andy Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ching-Wei Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yun Segraves-Chun
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Tweardy
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vijaya Gottumukkala
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jean-Nicolas Vauthey
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas Aloia
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Juan P Cata
- Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA.,Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
42
|
Tran TB, Ethun CG, Pawlik TM, Schmidt C, Beal EW, Fields RC, Krasnick B, Weber SM, Salem A, Martin RCG, Scoggins CR, Shen P, Mogal HD, Idrees K, Isom CA, Hatzaras I, Shenoy R, Maithel SK, Poultsides GA. Actual 5-Year Survivors After Surgical Resection of Hilar Cholangiocarcinoma. Ann Surg Oncol 2018; 26:611-618. [PMID: 30539494 DOI: 10.1245/s10434-018-7075-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND The prevalence and characteristics of actual 5-year survivors after surgical treatment of hilar cholangiocarcinoma (HC) have not been described previously. METHODS Patients who underwent resection for HC from 2000 to 2015 were analyzed through a multi-institutional registry from 10 U.S. academic medical centers. The clinicopathologic characteristics and both the perioperative and long-term outcomes for actual 5-year survivors were compared with those for non-survivors (patients who died within 5 years after surgery). Patients alive at last encounter who had a follow-up period shorter than 5 years were excluded from the study. RESULTS The study identified 257 patients with HC who underwent curative-intent resection with an actuarial 5-year survival of 19%. Of 194 patients with a follow-up period longer than 5 years, 23 (12%) were 5-year survivors. Compared with non-survivors, the 5-year survivors had a lower median pretreatment CA 19-9 level (116 vs. 34 U/L; P = 0.008) and a lower rate of lymph node involvement (42% vs. 15%; P = 0.027) and R1 margins (39% vs. 17%; P = 0.042). However, the sole presence of these factors did not preclude a 5-year survival after surgery. The frequencies of bile duct resection alone, major hepatectomy, caudate lobe resection, portal vein or hepatic artery resection, preoperative biliary sepsis, intraoperative blood transfusion, serious postoperative complications, and receipt of adjuvant chemotherapy were comparable between the two groups. CONCLUSIONS One in eight patients with HC reaches the 5-year survival milestone after resection. A 5-year survival can be achieved even in the presence of traditionally unfavorable clinicopathologic factors (elevated CA 19-9, nodal metastasis, and R1 margins).
Collapse
Affiliation(s)
- Thuy B Tran
- Department of Surgery, Stanford University, Stanford, CA, USA
| | - Cecilia G Ethun
- Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Timothy M Pawlik
- Department of Surgery, The Johns Hopkins Hospital, Baltimore, MD, USA.,Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Carl Schmidt
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Eliza W Beal
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Ryan C Fields
- Department of Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - Bradley Krasnick
- Department of Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - Sharon M Weber
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Ahmed Salem
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Robert C G Martin
- Department of Surgery, University of Louisville, Louisville, KY, USA
| | | | - Perry Shen
- Department of Surgery, Wake Forest University, Winston-Salem, NC, USA
| | - Harveshp D Mogal
- Department of Surgery, Wake Forest University, Winston-Salem, NC, USA
| | - Kamran Idrees
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chelsea A Isom
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Rivfka Shenoy
- Department of Surgery, New York University, New York, NY, USA
| | - Shishir K Maithel
- Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | | |
Collapse
|
43
|
Bomans K, Schenz J, Sztwiertnia I, Schaack D, Weigand MA, Uhle F. Sepsis Induces a Long-Lasting State of Trained Immunity in Bone Marrow Monocytes. Front Immunol 2018; 9:2685. [PMID: 30510555 PMCID: PMC6254543 DOI: 10.3389/fimmu.2018.02685] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/30/2018] [Indexed: 02/03/2023] Open
Abstract
Innate immune memory describes the functional reprogramming of innate immune cells after pathogen contact, leading to either a boosted (trained immunity) or a diminished (immune tolerance) response to a secondary stimulus. Immune tolerance or “sepsis-induced immunosuppression” is a typical hallmark of patients after sepsis survival, characterized by hypo-responsiveness of the host's immune system. This condition renders the host vulnerable for a persisting infection or the occurrence of secondary, often opportunistic infections, along with an increased mortality rate. The mechanisms involved in the maintenance of this long-lasting condition are not examined yet. Polymicrobial abdominal sepsis was induced in 12 week old male C57BL/6 mice by cecal ligation and puncture. Mice were euthanized 3 months after insult. Immune cell composition of the spleen and whole blood, as well as stem and progenitor cells of the bone marrow, were assessed by flow cytometry. Whole blood and bone marrow monocytes were stimulated with LPS and supernatant levels of TNF and IL-6 detected by ELISA. Furthermore, naïve bone marrow monocytes were analyzed for metabolic (Seahorse technology) and transcriptomic (RNA sequencing) changes. Flow cytometric analysis revealed an increase of inflammatory monocytes and regulatory T cells in the spleen, whereby immune composition of whole blood kept unchanged. Granulocyte-monocyte progenitor cells are increased in sepsis survivors. Systemic cytokine response was unchanged after LPS challenge. In contrast, cytokine response of post-septic naïve bone marrow monocytes was increased. Metabolic analysis revealed enhanced glycolytic activity, whereas mitochondrial indices were not affected. In addition, RNA sequencing analysis of global gene expression in monocytes revealed a sustained signature of 367 differentially expressed genes. We here demonstrate that sepsis via functional reprogramming of naïve bone marrow monocytes induces a cellular state of trained immunity, which might be counteracted depending on the compartmental localization of the cell. These findings shed new light on the complex aftermath of sepsis and open up a new pathophysiological framework in need for further research.
Collapse
Affiliation(s)
- Katharina Bomans
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Judith Schenz
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Isabella Sztwiertnia
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Dominik Schaack
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Florian Uhle
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
44
|
Denstaedt SJ, Singer BH, Standiford TJ. Sepsis and Nosocomial Infection: Patient Characteristics, Mechanisms, and Modulation. Front Immunol 2018; 9:2446. [PMID: 30459764 PMCID: PMC6232897 DOI: 10.3389/fimmu.2018.02446] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/03/2018] [Indexed: 12/16/2022] Open
Abstract
Sepsis is a leading cause of death worldwide. After initial trials modulating the hyperinflammatory phase of sepsis failed, generations of researchers have focused on evaluating hypo-inflammatory immune phenotypes. The main goal has been to develop prognostic biomarkers and therapies to reduce organ dysfunction, nosocomial infection, and death. The depressed host defense in sepsis has been characterized by broad cellular reprogramming including lymphocyte exhaustion, apoptosis, and depressed cytokine responses. Despite major advances in this field, our understanding of the dynamics of the septic host response and the balance of inflammatory and anti-inflammatory cellular programs remains limited. This review aims to summarize the epidemiology of nosocomial infections and characteristic immune responses associated with sepsis, as well as immunostimulatory therapies currently under clinical investigation.
Collapse
Affiliation(s)
| | | | - Theodore J. Standiford
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
45
|
Abstract
An estimated 14 million patients survive sepsis hospitalization each year. However, survivors commonly experience new functional disability, cognitive impairment, and a high rate of further medical setbacks, including hospital readmission and late death. One in 5 older survivors has a potentially preventable hospital admission with in 90 days, most commonly for infection. Treatment should focus on preventing the common sequelae of critical illness during the initial hospitalization, tailoring medical care to minimize the risk for common and potentially preventable causes of hospital readmission, and promoting functional recovery.
Collapse
|
46
|
Vicente D, Ikoma N, Chiang YJ, Fournier K, Tzeng CWD, Song S, Mansfield P, Ajani J, Badgwell BD. Preoperative Therapy for Gastric Adenocarcinoma is Protective for Poor Oncologic Outcomes in Patients with Complications After Gastrectomy. Ann Surg Oncol 2018; 25:2720-2730. [PMID: 29987602 DOI: 10.1245/s10434-018-6638-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Postoperative complications (POC) are associated with poor oncologic outcomes in gastric cancer. We sought to evaluate the impact of POC on survival in patients with gastric cancer treated with upfront surgery (UpSurg) versus those treated with preoperative therapy (PreT). METHODS We analyzed data from a prospectively maintained database of patients who had undergone resection of their gastric cancer at our institution. Patients with T1N0 or M1 lesions, recurrent disease, and mortality within 90 days were excluded. Survival was compared between patients with and without POC in the UpSurg and PreT groups. Cox regression analyses were used to examine factors associated with overall survival (OS) and disease-free survival (DFS). RESULTS A total of 421 patients underwent resection of gastric cancer: 30% underwent upfront surgery, and 51% had a POC. Among patients who had POCs, 71% were infectious and 53% were Clavien-Dindo grade III or IV. UpSurg patients with a POC had shorter OS (5-year, 47 vs. 85%; p < 0.001) and DFS (5-year, 46 vs. 76%; p < 0.001) than those without a POC. In contrast, there was no difference in OS (5-year, 57 vs. 63%; p = 0.77) and DFS (5-year, 52 vs. 52%; p = 0.52) between PreT patients with and without POC. Multivariable Cox regression model demonstrated that a POC in UpSurg patients had significant impact on DFS (2.6 [95% confidence interval (CI) 1.48-4.74]), whereas it did not in PreT patients (0.9 [95% CI 0.70-1.33]). CONCLUSIONS The use of preoperative therapy negated the impact of POCs on OS and DFS in patients undergoing resection for gastric cancer.
Collapse
Affiliation(s)
- Diego Vicente
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX, 77030, USA
| | - Naruhiko Ikoma
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX, 77030, USA
| | - Yi-Ju Chiang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX, 77030, USA
| | - Keith Fournier
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX, 77030, USA
| | - Ching-Wei D Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX, 77030, USA
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Mansfield
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX, 77030, USA
| | - Jaffer Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brian D Badgwell
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX, 77030, USA.
| |
Collapse
|
47
|
Coopersmith CM, De Backer D, Deutschman CS, Ferrer R, Lat I, Machado FR, Martin GS, Martin-Loeches I, Nunnally ME, Antonelli M, Evans LE, Hellman J, Jog S, Kesecioglu J, Levy MM, Rhodes A. Surviving sepsis campaign: research priorities for sepsis and septic shock. Intensive Care Med 2018; 44:1400-1426. [PMID: 29971592 PMCID: PMC7095388 DOI: 10.1007/s00134-018-5175-z] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/11/2018] [Indexed: 02/06/2023]
Abstract
Objective To identify research priorities in the management, epidemiology, outcome and underlying causes of sepsis and septic shock. Design A consensus committee of 16 international experts representing the European Society of Intensive Care Medicine and Society of Critical Care Medicine was convened at the annual meetings of both societies. Subgroups had teleconference and electronic-based discussion. The entire committee iteratively developed the entire document and recommendations. Methods Each committee member independently gave their top five priorities for sepsis research. A total of 88 suggestions (ESM 1 - supplemental table 1) were grouped into categories by the committee co-chairs, leading to the formation of seven subgroups: infection, fluids and vasoactive agents, adjunctive therapy, administration/epidemiology, scoring/identification, post-intensive care unit, and basic/translational science. Each subgroup had teleconferences to go over each priority followed by formal voting within each subgroup. The entire committee also voted on top priorities across all subgroups except for basic/translational science. Results The Surviving Sepsis Research Committee provides 26 priorities for sepsis and septic shock. Of these, the top six clinical priorities were identified and include the following questions: (1) can targeted/personalized/precision medicine approaches determine which therapies will work for which patients at which times?; (2) what are ideal endpoints for volume resuscitation and how should volume resuscitation be titrated?; (3) should rapid diagnostic tests be implemented in clinical practice?; (4) should empiric antibiotic combination therapy be used in sepsis or septic shock?; (5) what are the predictors of sepsis long-term morbidity and mortality?; and (6) what information identifies organ dysfunction? Conclusions While the Surviving Sepsis Campaign guidelines give multiple recommendations on the treatment of sepsis, significant knowledge gaps remain, both in bedside issues directly applicable to clinicians, as well as understanding the fundamental mechanisms underlying the development and progression of sepsis. The priorities identified represent a roadmap for research in sepsis and septic shock. Electronic supplementary material The online version of this article (10.1007/s00134-018-5175-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University, Atlanta, GA, USA
| | - Daniel De Backer
- Chirec Hospitals, Université Libre de Bruxelles, Brussels, Belgium.
| | - Clifford S Deutschman
- Department of Pediatrics, Cohen Children's Medical Center, Northwell Health, New Hyde Park, NY, USA.,The Feinstein Institute for Medical Research/Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA
| | - Ricard Ferrer
- Intensive Care Department, Vall d'Hebron University Hospital, Barcelona, Spain.,Shock, Organ Dysfunction and Resuscitation (SODIR) Research Group, Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Ishaq Lat
- Rush University Medical Center, Chicago, IL, USA
| | | | - Greg S Martin
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Grady Memorial Hospital and Emory Critical Care Center, Emory University, Atlanta, GA, USA
| | - Ignacio Martin-Loeches
- Multidisciplinary Intensive Care Research Organization (MICRO), Department of Intensive Care Medicine, Trinity Centre for Health Sciences, St James's University Hospital, Dublin, Ireland
| | | | - Massimo Antonelli
- Department of Anesthesiology and Intensive Care Medicine, Fondazione Policlinico Universitario A.Gemelli-Università Cattolica del Sacro Cuore, Rome, Italy
| | - Laura E Evans
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Bellevue Hospital Center and New York University School of Medicine, New York, NY, USA
| | - Judith Hellman
- University of California, San Francisco, San Francisco, CA, USA
| | - Sameer Jog
- Deenanath Mangeshkar Hospital and Research Center, Pune, India
| | - Jozef Kesecioglu
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mitchell M Levy
- Rhode Island Hospital, Alpert Medical School at Brown University, Providence, RI, USA
| | - Andrew Rhodes
- Department of Adult Critical Care, St George's University Hospitals NHS Foundation Trust and St George's University of London, London, UK
| |
Collapse
|
48
|
Kumar V. T cells and their immunometabolism: A novel way to understanding sepsis immunopathogenesis and future therapeutics. Eur J Cell Biol 2018; 97:379-392. [PMID: 29773345 DOI: 10.1016/j.ejcb.2018.05.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/03/2018] [Accepted: 05/03/2018] [Indexed: 02/08/2023] Open
Abstract
Sepsis has always been considered as a big challenge for pharmaceutical companies in terms of discovering and designing new therapeutics. The pathogenesis of sepsis involves aberrant activation of innate immune cells (i.e. macrophages, neutrophils etc.) at early stages. However, a stage of immunosuppression is also observed during sepsis even in the patients who have recovered from it. This stage of immunosuppression is observed due to the loss of conventional (i.e. CD4+, CD8+) T cells, Th17 cells and an upregulation of regulatory T cells (Tregs). This process also impacts metabolic processes controlling immune cell metabolism called immunometabolism. The present review is focused on the T cell-mediated immune response, their immunometabolism and targeting T cell immunometabolism during sepsis as future therapeutic approach. The first part of the manuscripts describes an impact of sepsis on conventional T cells, Th17 cells and Tregs along with their impact on sepsis. The subsequent section further describes the immunometabolism of these cells (CD4+, CD8+, Th17, and Tregs) under normal conditions and during sepsis-induced immunosuppression. The article ends with the therapeutic targeting of T cell immunometabolism (both conventional T cells and Tregs) during sepsis as a future immunomodulatory approach for its management.
Collapse
Affiliation(s)
- V Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Mater Research, Faculty of Medicine, University of Queensland, St. Lucia, Brisbane, Queensland 4078, Australia; School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, Brisbane, Queensland 4078, Australia.
| |
Collapse
|
49
|
Pène F, Azabou E, Jung B, Gibot S, Guillon A, Monneret G, Silva S, Taccone F, Textoris J, Uhel F, Zafrani L, de Prost N. Faire face à la menace infectieuse en réanimation : de la veille épidémiologique à l’innovation. Actes du séminaire de recherche translationnelle de la Société de réanimation de langue française (5 décembre 2017). MEDECINE INTENSIVE REANIMATION 2018. [DOI: 10.3166/rea-2018-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Le séminaire annuel de la Commission de recherche translationnelle de la SRLF a pour but de réunir des cliniciens et scientifiques autour de grandes thématiques de recherche en médecine intensive et réanimation. La quatrième édition du séminaire a porté sur l’infectiologie, thématique au centre des préoccupations des réanimateurs. Les interventions se sont ainsi focalisées sur des aspects aussi divers que les relations hôtes–pathogènes, la contribution de pathogènes dans des pathologies habituellement considérées comme non infectieuses, l’émergence de nouveaux risques infectieux, les avancées technologiques du diagnostic moléculaire des infections et le développement de stratégies antibactériennes alternatives à l’antibiothérapie classique.
Collapse
|
50
|
Tai LH, Ananth AA, Seth R, Alkayyal A, Zhang J, de Souza CT, Staibano P, Kennedy MA, Auer RC. Sepsis increases perioperative metastases in a murine model. BMC Cancer 2018. [PMID: 29530012 PMCID: PMC5848444 DOI: 10.1186/s12885-018-4173-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Cancer surgery can promote tumour metastases and worsen prognosis, however, the effect of perioperative complications on metastatic disease remains unclear. In this study we sought to evaluate the effect of common perioperative complications including perioperative blood loss, hypothermia, and sepsis on tumour metastases in a murine model. Methods Prior to surgery, pulmonary metastases were established by intravenous challenge of CT26LacZ colon cancer cells in BALB/c mice. Surgical stress was generated through partial hepatectomy (PH) or left nephrectomy (LN). Sepsis was induced by puncturing the cecum to express stool into the abdomen. Hemorrhagic shock was induced by removal of 30% of total blood volume (i.e. stage 3 hemorrhage) via the saphenous vein. Hypothermia was induced by removing the heating apparatus during surgery and lowering core body temperatures to 30 °C. Lung tumour burden was quantified 3 days following surgery. Results Surgically stressed mice subjected to stage 3 hemorrhage or hypothermia did not show an additional increase in lung tumour burden. In contrast, surgically stressed mice subjected to intraoperative sepsis demonstrated an additional 2-fold increase in the number of tumour metastases. Furthermore, natural killer (NK) cell function, as assessed by YAC-1 tumour cell lysis, was significantly attenuated in surgically stressed mice subjected to intraoperative sepsis. Both NK cell-mediated cytotoxic function and lung tumour burden were improved with perioperative administration of polyI:C, which is a toll-like receptor (TLR)-3 ligand. Conclusions Perioperative sepsis alone, but not hemorrhage or hypothermia, enhances the prometastatic effect of surgery in murine models of cancer. Understanding the cellular mechanisms underlying perioperative immune suppression will facilitate the development of immunomodulation strategies that can attenuate metastatic disease.
Collapse
Affiliation(s)
- Lee-Hwa Tai
- Deparment of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Abhirami A Ananth
- Deparment of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Rashmi Seth
- Department of Surgery, Division of General Surgery, University of Ottawa, Ottawa, Canada
| | - Almohanad Alkayyal
- Deparment of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Medical Laboratory Technology, University of Tabuk, Tabuk, Saudi Arabia
| | - Jiqing Zhang
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Neurosurgery, The Second Hospital of Shandong University, Shandong, China.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | | | - Phillip Staibano
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Michael A Kennedy
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Rebecca C Auer
- Deparment of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada. .,Center for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Canada. .,Department of Surgery, Division of General Surgery, University of Ottawa, Ottawa, Canada. .,Ottawa General Hospital, 501 Smyth Road, 1617 CCW, Box 134, Ottawa, ON, K1H8L6, Canada.
| |
Collapse
|