1
|
Silva EE, Skon-Hegg C, Badovinac VP, Griffith TS. The Calm after the Storm: Implications of Sepsis Immunoparalysis on Host Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:711-719. [PMID: 37603859 PMCID: PMC10449360 DOI: 10.4049/jimmunol.2300171] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/12/2023] [Indexed: 08/23/2023]
Abstract
The immunological hallmarks of sepsis include the inflammation-mediated cytokine storm, apoptosis-driven lymphopenia, and prolonged immunoparalysis. Although early clinical efforts were focused on increasing the survival of patients through the first phase, studies are now shifting attention to the long-term effects of sepsis on immune fitness in survivors. In particular, the most pertinent task is deciphering how the immune system becomes suppressed, leading to increased incidence of secondary infections. In this review, we introduce the contribution of numerical changes and functional reprogramming within innate (NK cells, dendritic cells) and adaptive (T cells, B cells) immune cells on the chronic immune dysregulation in the septic murine and human host. We briefly discuss how prior immunological experience in murine models impacts sepsis severity, immune dysfunction, and clinical relevance. Finally, we dive into how comorbidities, specifically autoimmunity and cancer, can influence host susceptibility to sepsis and the associated immune dysfunction.
Collapse
Affiliation(s)
- Elvia E Silva
- Department of Pathology, University of Iowa, Iowa City, IA
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA
| | - Cara Skon-Hegg
- Department of Urology, University of Minnesota, Minneapolis, MN
| | - Vladimir P Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA
| | - Thomas S Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
- Minneapolis VA Health Care System, Minneapolis, MN
| |
Collapse
|
2
|
Magatti M, Pischiutta F, Ortolano F, Pasotti A, Caruso E, Cargnoni A, Papait A, Capuzzi F, Zoerle T, Carbonara M, Stocchetti N, Borsa S, Locatelli M, Erba E, Prati D, Silini AR, Zanier ER, Parolini O. Systemic immune response in young and elderly patients after traumatic brain injury. Immun Ageing 2023; 20:41. [PMID: 37573338 PMCID: PMC10422735 DOI: 10.1186/s12979-023-00369-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) is a leading cause of death and long-term disability worldwide. In addition to primary brain damage, systemic immune alterations occur, with evidence for dysregulated immune responses in aggravating TBI outcome and complications. However, immune dysfunction following TBI has been only partially understood, especially in the elderly who represent a substantial proportion of TBI patients and worst outcome. Therefore, we aimed to conduct an in-depth immunological characterization of TBI patients, by evaluating both adaptive (T and B lymphocytes) and innate (NK and monocytes) immune cells of peripheral blood mononuclear cells (PBMC) collected acutely (< 48 h) after TBI in young (18-45 yo) and elderly (> 65 yo) patients, compared to age-matched controls, and also the levels of inflammatory biomarkers. RESULTS Our data show that young respond differently than elderly to TBI, highlighting the immune unfavourable status of elderly compared to young patients. While in young only CD4 T lymphocytes are activated by TBI, in elderly both CD4 and CD8 T cells are affected, and are induced to differentiate into subtypes with low cytotoxic activity, such as central memory CD4 T cells and memory precursor effector CD8 T cells. Moreover, TBI enhances the frequency of subsets that have not been previously investigated in TBI, namely the double negative CD27- IgD- and CD38-CD24- B lymphocytes, and CD56dim CD16- NK cells, both in young and elderly patients. TBI reduces the production of pro-inflammatory cytokines TNF-α and IL-6, and the expression of HLA-DM, HLA-DR, CD86/B7-2 in monocytes, suggesting a compromised ability to drive a pro-inflammatory response and to efficiently act as antigen presenting cells. CONCLUSIONS We described the acute immunological response induced by TBI and its relation with injury severity, which could contribute to pathologic evolution and possibly outcome. The focus on age-related immunological differences could help design specific therapeutic interventions based on patients' characteristics.
Collapse
Affiliation(s)
- Marta Magatti
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy.
| | - Francesca Pischiutta
- Department of Acute Brain Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Fabrizio Ortolano
- Dipartimento di Anestesia-Rianimazione e Emergenza Urgenza, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Anna Pasotti
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Enrico Caruso
- Department of Acute Brain Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
- Dipartimento di Anestesia-Rianimazione e Emergenza Urgenza, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Anna Cargnoni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Andrea Papait
- Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Roma, Italy
| | - Franco Capuzzi
- Dipartimento Medicina di Laboratorio, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Tommaso Zoerle
- Dipartimento di Anestesia-Rianimazione e Emergenza Urgenza, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milano, Italy
| | - Marco Carbonara
- Dipartimento di Anestesia-Rianimazione e Emergenza Urgenza, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Nino Stocchetti
- Dipartimento di Anestesia-Rianimazione e Emergenza Urgenza, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milano, Italy
| | - Stefano Borsa
- Unit of Neurosurgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Marco Locatelli
- Department of Pathophysiology and Transplantation, University of Milan, Milano, Italy
- Unit of Neurosurgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Elisa Erba
- Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Daniele Prati
- Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Antonietta R Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Elisa R Zanier
- Department of Acute Brain Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Ornella Parolini
- Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Roma, Italy
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Roma, Italy
| |
Collapse
|
3
|
Bouras M, Asehnoune K, Roquilly A. Immune modulation after traumatic brain injury. Front Med (Lausanne) 2022; 9:995044. [PMID: 36530909 PMCID: PMC9751027 DOI: 10.3389/fmed.2022.995044] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/14/2022] [Indexed: 07/20/2023] Open
Abstract
Traumatic brain injury (TBI) induces instant activation of innate immunity in brain tissue, followed by a systematization of the inflammatory response. The subsequent response, evolved to limit an overwhelming systemic inflammatory response and to induce healing, involves the autonomic nervous system, hormonal systems, and the regulation of immune cells. This physiological response induces an immunosuppression and tolerance state that promotes to the occurrence of secondary infections. This review describes the immunological consequences of TBI and highlights potential novel therapeutic approaches using immune modulation to restore homeostasis between the nervous system and innate immunity.
Collapse
Affiliation(s)
- Marwan Bouras
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France
| | - Karim Asehnoune
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France
| | - Antoine Roquilly
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France
| |
Collapse
|
4
|
ICU and Sepsis: Role of Myeloid and Lymphocyte Immune Cells. JOURNAL OF ONCOLOGY 2022; 2022:7340266. [PMID: 36199798 PMCID: PMC9527402 DOI: 10.1155/2022/7340266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/31/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022]
Abstract
Sepsis is a severe immune system reaction to infection and a major cause of ICU-related fatalities. Because of the high mortality, high cost of treatment, and complex aetiology of sepsis, sepsis has a huge impact on healthcare. Some of the health complications in sepsis are abnormal cardiac functions, hypoperfusion, hypotension, tissue damage, multiple organ failure, and ultimately death. Individuals with weak immune systems and chronic medical conditions are highly vulnerable to sepsis. In sepsis, a patient shows the extreme immune response in the initial stage while prolonged immunosuppression in the later stages. Sepsis-driven immunosuppression ushers in death because sepsis cases develop secondary infections postrecovery. The later immunocompromised state in sepsis is attributed myeloid-derived suppressor cell upregulation and reduced immune activity displayed by lymphocytes (lymphocyte anergy). As a result, it is currently suggested that regulating the immune response is a better therapeutic approach than focusing on inflammation to improve the immune system's capacity to fight infections. Moreover, finding novel and accurate prognostic biomarkers that can help in rapid sepsis diagnoses and deciding better therapeutic strategies will significantly lower clinical case mortality rates.
Collapse
|
5
|
Yao RQ, Ren C, Zheng LY, Xia ZF, Yao YM. Advances in Immune Monitoring Approaches for Sepsis-Induced Immunosuppression. Front Immunol 2022; 13:891024. [PMID: 35619710 PMCID: PMC9127053 DOI: 10.3389/fimmu.2022.891024] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/29/2022] [Indexed: 12/29/2022] Open
Abstract
Sepsis represents a life-threatening organ dysfunction due to an aberrant host response. Of note is that majority of patients have experienced a severe immune depression during and after sepsis, which is significantly correlated with the occurrence of nosocomial infection and higher risk of in-hospital death. Nevertheless, the clinical sign of sepsis-induced immune paralysis remains highly indetectable and ambiguous. Given that, specific yet robust biomarkers for monitoring the immune functional status of septic patients are of prominent significance in clinical practice. In turn, the stratification of a subgroup of septic patients with an immunosuppressive state will greatly contribute to the implementation of personalized adjuvant immunotherapy. In this review, we comprehensively summarize the mechanism of sepsis-associated immunosuppression at the cellular level and highlight the recent advances in immune monitoring approaches targeting the functional status of both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Ren-Qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chao Ren
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Pulmonary and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Li-Yu Zheng
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhao-Fan Xia
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
6
|
Cytokine producing ability of peripheral blood cells from COVID-19 patients after unspecific in vitro stimulation. Inflamm Res 2022; 71:331-341. [PMID: 35157090 PMCID: PMC8853300 DOI: 10.1007/s00011-022-01543-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/29/2021] [Accepted: 01/29/2022] [Indexed: 11/05/2022] Open
Abstract
Objective and design Subjects and methods Results Conclusion
Collapse
|
7
|
Bao W, Lin Y, Chen Z. The Peripheral Immune System and Traumatic Brain Injury: Insight into the role of T-helper cells. Int J Med Sci 2021; 18:3644-3651. [PMID: 34790036 PMCID: PMC8579286 DOI: 10.7150/ijms.46834] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/17/2021] [Indexed: 12/15/2022] Open
Abstract
Emerging evidence suggests that immune-inflammatory processes are key elements in the physiopathological events associated with traumatic brain injury (TBI). TBI is followed by T-cell-specific immunological changes involving several subsets of T-helper cells and the cytokines they produce; these processes can have opposite effects depending on the disease course and cytokine concentrations. Efforts are underway to identify the T-helper cells and cytokine profiles associated with prognosis. These predictors may eventually serve as effective treatment targets to decrease morbidity and mortality and to improve the management of TBI patients. Here, we review the immunological response to TBI, the possible molecular mechanisms of this response, and therapeutic strategies to address it.
Collapse
Affiliation(s)
| | | | - Zuobing Chen
- Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
8
|
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
|
9
|
Gaborit BJ, Chaumette T, Chauveau M, Asquier-Khati A, Roquilly A, Boutoille D, Josien R, Salomon BL, Asehnoune K. Circulating Treg cells expressing TNF receptor type 2 contributes to sepsis-induced immunosuppression in patients during sepsis shock. J Infect Dis 2021; 224:2160-2169. [PMID: 34019653 DOI: 10.1093/infdis/jiab276] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 05/17/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Septic shock remains a major cause of death that can be complicated by a long-term impairment in immune function defining immunosuppression induced by sepsis (IS). Among Treg cells, the tumor necrosis factor receptor 2 positive (TNFR2 pos) Treg cell subset endorses significant immunosuppressive functions in human tumors and in a sepsis mouse model but have not been investigated during septic shock in humans. METHODS We prospectively enrolled patients with septic shock hospitalized in Intensive Care Unit (ICU). We performed immunophenotyping and functional tests of CD4+T cells, Treg cells and TNFR2 posTregcells, on blood samples collected at 1, 4 and 7 days after admission in ICU. RESULTS We investigated 10 patients with septic shock and compared to 10 healthy controls. Although the proportions of circulating Tregcells and TNFR2 posTregcells subsets were not increased, their CTLA-4 expression and suppressive functions in vitro were increased at 4 days of septic shock. Also, PBMC from healthy donors cultured with serum from septic shock patients had increased CTLA4 expression in TNFR2 pos Treg cells compared to TNFR2 neg Treg cells. CONCLUSION In patients with septic shock, CTLA-4 expression and suppressive function were increased in circulating TNFR2 posTreg cells. We identify TNFR2 posTreg cells as a potential attractive target for therapeutic intervention.
Collapse
Affiliation(s)
- Benjamin Jean Gaborit
- Nantes Université, Thérapeutiques Anti-Infectieuses, Nantes, France.,CHU Nantes, Department of Infectious Diseases.,CHU Nantes, INSERM, CIC, Nantes, France
| | - Tanguy Chaumette
- Nantes Université, Thérapeutiques Anti-Infectieuses, Nantes, France
| | - Marie Chauveau
- Nantes Université, Thérapeutiques Anti-Infectieuses, Nantes, France.,CHU Nantes, Department of Infectious Diseases.,CHU Nantes, INSERM, CIC, Nantes, France
| | - Antoine Asquier-Khati
- Nantes Université, Thérapeutiques Anti-Infectieuses, Nantes, France.,CHU Nantes, Department of Infectious Diseases.,CHU Nantes, INSERM, CIC, Nantes, France
| | - Antoine Roquilly
- Nantes Université, Thérapeutiques Anti-Infectieuses, Nantes, France.,CHU Nantes, Surgical Intensive Care Unit, Nantes, France
| | - David Boutoille
- Nantes Université, Thérapeutiques Anti-Infectieuses, Nantes, France.,CHU Nantes, Department of Infectious Diseases.,CHU Nantes, INSERM, CIC, Nantes, France
| | - Régis Josien
- Nantes Université, INSERM, Centre de Recherche en Transplantation et Immunologie UMR 1064, ITUN, Nantes, France.,CHU Nantes, Laboratoire d'Immunologie, CIMNA, Nantes, France
| | - Benoit L Salomon
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Karim Asehnoune
- Nantes Université, Thérapeutiques Anti-Infectieuses, Nantes, France.,CHU Nantes, Surgical Intensive Care Unit, Nantes, France
| |
Collapse
|
10
|
Brady J, Horie S, Laffey JG. Role of the adaptive immune response in sepsis. Intensive Care Med Exp 2020; 8:20. [PMID: 33336293 PMCID: PMC7746432 DOI: 10.1186/s40635-020-00309-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023] Open
Abstract
Sepsis is a syndrome of shock and dysfunction of multiple vital organs that is caused by an uncontrolled immune response to infection and has a high mortality rate. There are no therapies for sepsis, and it has become a global cause for concern. Advances in patient care and management now mean that most patients survive the initial hyper-inflammatory phase of sepsis but progress to a later immunosuppressed phase, where 30% of patients die due to secondary infection. Deficits in the adaptive immune response may play a major role in sepsis patient mortality. The adaptive immune response involves a number of cell types including T cells, B cells and dendritic cells, all with immunoregulatory roles aimed at limiting damage and returning immune homeostasis after infection or insult. However, in sepsis, adaptive immune cells experience cell death or exhaustion, meaning that they have defective effector and memory responses ultimately resulting in an ineffective or suppressed immune defence. CD4+ T cells seem to be the most susceptible to cell death during sepsis and have ensuing defective secretory profiles and functions. Regulatory T cells seem to evade apoptosis and contribute to the immune suppression observed with sepsis. Preclinical studies have identified a number of new targets for therapy in sepsis including anti-apoptotic agents and monoclonal antibodies aimed at reducing cell death, exhaustion and maintaining/restoring adaptive immune cell functions. While early phase clinical trials have demonstrated safety and encouraging signals for biologic effect, larger scale clinical trial testing is required to determine whether these strategies will prove effective in improving outcomes from sepsis.
Collapse
Affiliation(s)
- Jack Brady
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| | - Shahd Horie
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| | - John G Laffey
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland. .,Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland. .,Department of Anaesthesia, Galway University Hospitals, SAOLTA University Health Group, Galway, Ireland.
| |
Collapse
|
11
|
Martin MD, Badovinac VP, Griffith TS. CD4 T Cell Responses and the Sepsis-Induced Immunoparalysis State. Front Immunol 2020; 11:1364. [PMID: 32733454 PMCID: PMC7358556 DOI: 10.3389/fimmu.2020.01364] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022] Open
Abstract
Sepsis remains a major cause of death in the United States and worldwide, and costs associated with treating septic patients place a large burden on the healthcare industry. Patients who survive the acute phase of sepsis display long-term impairments in immune function due to reductions in numbers and function of many immune cell populations. This state of chronic immunoparalysis renders sepsis survivors increasingly susceptible to infection with newly or previously encountered infections. CD4 T cells play important roles in the development of cellular and humoral immune responses following infection. Understanding how sepsis impacts the CD4 T cell compartment is critical for informing efforts to develop treatments intended to restore immune system homeostasis following sepsis. This review will focus on the current understanding of how sepsis impacts the CD4 T cell responses, including numerical representation, repertoire diversity, phenotype and effector functionality, subset representation (e.g., Th1 and Treg frequency), and therapeutic efforts to restore CD4 T cell numbers and function following sepsis. Additionally, we will discuss recent efforts to model the acute sepsis phase and resulting immune dysfunction using mice that have previously encountered infection, which more accurately reflects the immune system of humans with a history of repeated infection throughout life. A thorough understanding of how sepsis impacts CD4 T cells based on previous studies and new models that accurately reflect the human immune system may improve translational value of research aimed at restoring CD4 T cell-mediated immunity, and overall immune fitness following sepsis.
Collapse
Affiliation(s)
- Matthew D. Martin
- Department of Urology, University of Minnesota, Minneapolis, MN, United States
| | - Vladimir P. Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA, United States
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Thomas S. Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN, United States
- Microbiology, Immunology, and Cancer Biology PhD Program, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Minneapolis VA Healthcare System, Minneapolis, MN, United States
| |
Collapse
|
12
|
Cavaillon JM, Giamarellos-Bourboulis EJ. Immunosuppression is Inappropriately Qualifying the Immune Status of Septic and SIRS Patients. Shock 2019; 52:307-317. [PMID: 30239420 DOI: 10.1097/shk.0000000000001266] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Immunosuppression is the most commonly used concept to qualify the immune status of patients with either sterile systemic inflammatory response syndrome (SIRS) or sepsis. In this review we attempt to demonstrate that the concept of immunosuppression is an oversimplification of the complex anti-inflammatory response that occurs in patients dealing with a severe sterile or infectious insult. Particularly, the immune status of leukocytes varies greatly depending on the compartment from where they are derived from. Furthermore, although certain functions of immune cells present in the blood stream or in the hematopoietic organs can be significantly diminished, other functions are either unchanged or even enhanced. This juxtaposition illustrates that there is no global defect. The mechanisms called reprogramming or trained innate immunity are probably aimed at preventing a generalized deleterious inflammatory reaction, and work to maintain the defense mechanisms at their due levels.
Collapse
|
13
|
Sjaastad FV, Condotta SA, Kotov JA, Pape KA, Dail C, Danahy DB, Kucaba TA, Tygrett LT, Murphy KA, Cabrera-Perez J, Waldschmidt TJ, Badovinac VP, Griffith TS. Polymicrobial Sepsis Chronic Immunoparalysis Is Defined by Diminished Ag-Specific T Cell-Dependent B Cell Responses. Front Immunol 2018; 9:2532. [PMID: 30429857 PMCID: PMC6220049 DOI: 10.3389/fimmu.2018.02532] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/15/2018] [Indexed: 12/30/2022] Open
Abstract
Immunosuppression is one hallmark of sepsis, decreasing the host response to the primary septic pathogens and/or secondary nosocomial infections. CD4 T cells and B cells are among the array of immune cells that experience reductions in number and function during sepsis. “Help” from follicular helper (Tfh) CD4 T cells to B cells is needed for productive and protective humoral immunity, but there is a paucity of data defining the effect of sepsis on a primary CD4 T cell-dependent B cell response. Using the cecal ligation and puncture (CLP) mouse model of sepsis induction, we observed reduced antibody production in mice challenged with influenza A virus or TNP-KLH in alum early (2 days) and late (30 days) after CLP surgery compared to mice subjected to sham surgery. To better understand how these CD4 T cell-dependent B cell responses were altered by a septic event, we immunized mice with a Complete Freund's Adjuvant emulsion containing the MHC II-restricted peptide 2W1S56−68 coupled to the fluorochrome phycoerythrin (PE). Immunization with 2W1S-PE/CFA results in T cell-dependent B cell activation, giving us the ability to track defined populations of antigen-specific CD4 T cells and B cells responding to the same immunogen in the same mouse. Compared to sham mice, differentiation and class switching in PE-specific B cells were blunted in mice subjected to CLP surgery. Similarly, mice subjected to CLP had reduced expansion of 2W1S-specific T cells and Tfh differentiation after immunization. Our data suggest CLP-induced sepsis impacts humoral immunity by affecting the number and function of both antigen-specific B cells and CD4 Tfh cells, further defining the period of chronic immunoparalysis after sepsis induction.
Collapse
Affiliation(s)
- Frances V Sjaastad
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN, United States
| | | | - Jessica A Kotov
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN, United States
| | - Kathryn A Pape
- Department of Microbiology and Immunology, 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
| | - Derek B Danahy
- Department of Pathology, University of Iowa, Iowa City, IA, United States.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| | - Tamara A Kucaba
- Department of Urology, University of Minnesota, Minneapolis, MN, United States
| | - Lorraine T Tygrett
- Department of Pathology, University of Iowa, Iowa City, IA, United States
| | - Katherine A Murphy
- Department of Urology, 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
| | | | - 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
- 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
|
14
|
Bakos O, Lawson C, Rouleau S, Tai LH. Combining surgery and immunotherapy: turning an immunosuppressive effect into a therapeutic opportunity. J Immunother Cancer 2018; 6:86. [PMID: 30176921 PMCID: PMC6122574 DOI: 10.1186/s40425-018-0398-7] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 08/16/2018] [Indexed: 12/27/2022] Open
Abstract
Background Cancer surgery is necessary and life-saving. However, the majority of patients develop postoperative recurrence and metastasis, which are the main causes of cancer-related deaths. The postoperative stress response encompasses a broad set of physiological changes that have evolved to safeguard the host following major tissue trauma. These stress responses, however, intersect with cellular mediators and signaling pathways that contribute to cancer proliferation. Main Previous descriptive and emerging mechanistic studies suggest that the surgery-induced prometastatic effect is linked to impairment of both innate and adaptive immunity. Existing studies that combine surgery and immunotherapies have revealed that this combination strategy is not straightforward and patients have experienced both therapeutic benefit and drawbacks. This review will specifically assess the immunological pathways that are disrupted by oncologic surgical stress and provide suggestions for rationally combining cancer surgery with immunotherapies to improve immune and treatment outcomes. Short conclusion Given the prevalence of surgery as frontline therapy for solid cancers, the emerging data on postoperative immunosuppression and the rapid development of immunotherapy for oncologic treatment, we believe that future targeted studies of perioperative immunotherapy are warranted.
Collapse
Affiliation(s)
- Orneala Bakos
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Christine Lawson
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Samuel Rouleau
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Lee-Hwa Tai
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada. .,Centre de Recherche Clinique de Centre Hospitalier de l'Université de Sherbrooke (CHUS), Room 4853, 3001, 12e Avenue Nord, Sherbrooke, Québec, J1H 5N4, Canada.
| |
Collapse
|
15
|
Cavaillon JM, Adib-Conquy M, Cloëz-Tayarani I, Fitting C. Review: Immunodepression in sepsis and SIRS assessed by ex vivo cytokine production is not a generalized phenomenon: a review. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519010070020201] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Sepsis and non-infectious systemic inflammatory response syndrome (SIRS) are paradoxically associated with an exacerbated production of cytokines, as assessed by their presence in biological fluids, and a diminished ability of circulating leukocytes to produce cytokine upon in vitro activation. In this review, we depict that the observed cellular hyporeactivity is not a global phenomenon and that some signalling pathways are unaltered and allow the cells to respond normally to certain stimuli. Furthermore, we illustrate that during sepsis and SIRS, cells derived from tissues are either fully responsive to ex vivo stimuli or even primed, in contrast to cells derived from hematopoietic compartments (blood, spleen, etc.) which are hyporeactive. In addition to cytokine production, nuclear factor-κB (NF-κB) status within leukocytes can be used as a useful marker of hypo- or hyper-reactivity. We illustrate that the immune-depression reported in sepsis and SIRS patients, often revealed by a diminished capacity of leukocytes to respond to lipopolysaccharide, is not a generalized phenomenon and that SIRS is associated with a compartmentalized responsiveness which involves either anergic or primed cells.
Collapse
|
16
|
Role of cellular events in the pathophysiology of sepsis. Inflamm Res 2016; 65:853-868. [PMID: 27392441 DOI: 10.1007/s00011-016-0970-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 05/11/2016] [Accepted: 06/25/2016] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Sepsis is a dysregulated host immune response due to an uncontrolled infection. It is a leading cause of mortality in adult intensive care units globally. When the host immune response induced against a local infection fails to contain it locally, it progresses to sepsis, severe sepsis, septic shock and death. METHOD Literature survey was performed on the roles of different innate and adaptive immune cells in the development and progression of sepsis. Additionally, the effects of septic changes on reprogramming of different immune cells were also summarized to prepare the manuscript. FINDINGS Scientific evidences to date suggest that the loss of balance between inflammatory and anti-inflammatory responses results in reprogramming of immune cell activities that lead to irreversible tissue damaging events and multi-organ failure during sepsis. Many surface receptors expressed on immune cells at various stages of sepsis have been suggested as biomarkers for sepsis diagnosis. Various immunomodulatory therapeutics, which could improve the functions of immune cells during sepsis, were shown to restore immunological homeostasis and improve survival in animal models of sepsis. CONCLUSION In-depth and comprehensive knowledge on the immune cell activities and their correlation with severity of sepsis will help clinicians and scientists to design effective immunomodulatory therapeutics for treating sepsis.
Collapse
|
17
|
Abstract
Traumatic injury remains one of the most prevalent reasons for patients to be hospitalized. Burn injury accounts for 40,000 hospitalizations in the United States annually, resulting in a large burden on both the health and economic system and costing millions of dollars every year. The complications associated with postburn care can quickly cause life-threatening conditions including sepsis and multiple organ dysfunction and failure. In addition, alcohol intoxication at the time of burn injury has been shown to exacerbate these problems. One of the biggest reasons for the onset of these complications is the global suppression of the host immune system and increased susceptibility to infection. It has been hypothesized that infections after burn and other traumatic injury may stem from pathogenic bacteria from within the host's gastrointestinal tract. The intestine is the major reservoir of bacteria within the host, and many studies have demonstrated perturbations of the intestinal barrier after burn injury. This article reviews the findings of these studies as they pertain to changes in the intestinal immune system after alcohol and burn injury.
Collapse
|
18
|
Namas RA, Mi Q, Namas R, Almahmoud K, Zaaqoq AM, Abdul-Malak O, Azhar N, Day J, Abboud A, Zamora R, Billiar TR, Vodovotz Y. Insights into the Role of Chemokines, Damage-Associated Molecular Patterns, and Lymphocyte-Derived Mediators from Computational Models of Trauma-Induced Inflammation. Antioxid Redox Signal 2015; 23:1370-87. [PMID: 26560096 PMCID: PMC4685502 DOI: 10.1089/ars.2015.6398] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
SIGNIFICANCE Traumatic injury elicits a complex, dynamic, multidimensional inflammatory response that is intertwined with complications such as multiple organ dysfunction and nosocomial infection. The complex interplay between inflammation and physiology in critical illness remains a challenge for translational research, including the extrapolation to human disease from animal models. RECENT ADVANCES Over the past decade, we and others have attempted to decipher the biocomplexity of inflammation in these settings of acute illness, using computational models to improve clinical translation. In silico modeling has been suggested as a computationally based framework for integrating data derived from basic biology experiments as well as preclinical and clinical studies. CRITICAL ISSUES Extensive studies in cells, mice, and human blunt trauma patients have led us to suggest (i) that while an adequate level of inflammation is required for healing post-trauma, inflammation can be harmful when it becomes self-sustaining via a damage-associated molecular pattern/Toll-like receptor-driven feed-forward circuit; (ii) that chemokines play a central regulatory role in driving either self-resolving or self-maintaining inflammation that drives the early activation of both classical innate and more recently recognized lymphoid pathways; and (iii) the presence of multiple thresholds and feedback loops, which could significantly affect the propagation of inflammation across multiple body compartments. FUTURE DIRECTIONS These insights from data-driven models into the primary drivers and interconnected networks of inflammation have been used to generate mechanistic computational models. Together, these models may be used to gain basic insights as well as serving to help define novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Rami A. Namas
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Qi Mi
- Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rajaie Namas
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, Michigan
| | - Khalid Almahmoud
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Akram M. Zaaqoq
- Department of Critical Care Medicine, University of Pittsburgh, Pennsylvania
| | - Othman Abdul-Malak
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nabil Azhar
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Judy Day
- Department of Mathematics, University of Tennessee, Knoxville, Tennessee
| | - Andrew Abboud
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ruben Zamora
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
19
|
Palomero Rodríguez MÁ, García Navas R, Laporta Báez Y, de Vicente Sánchez J, Moyano Maza JC, Rodríguez López JM, Sánchez Conde MP, Mollinedo F. Arginase activity and CD3ζ expression after major surgery. Intensive Care Med 2015; 41:939-40. [PMID: 25739821 DOI: 10.1007/s00134-015-3714-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2015] [Indexed: 10/23/2022]
|
20
|
Cabrera-Perez J, Condotta SA, Badovinac VP, Griffith TS. Impact of sepsis on CD4 T cell immunity. J Leukoc Biol 2014; 96:767-77. [PMID: 24791959 PMCID: PMC4197564 DOI: 10.1189/jlb.5mr0114-067r] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 03/08/2014] [Accepted: 03/19/2014] [Indexed: 12/13/2022] Open
Abstract
Sepsis remains the primary cause of death from infection in hospital patients, despite improvements in antibiotics and intensive-care practices. Patients who survive severe sepsis can display suppressed immune function, often manifested as an increased susceptibility to (and mortality from) nosocomial infections. Not only is there a significant reduction in the number of various immune cell populations during sepsis, but there is also decreased function in the remaining lymphocytes. Within the immune system, CD4 T cells are important players in the proper development of numerous cellular and humoral immune responses. Despite sufficient clinical evidence of CD4 T cell loss in septic patients of all ages, the impact of sepsis on CD4 T cell responses is not well understood. Recent findings suggest that CD4 T cell impairment is a multipronged problem that results from initial sepsis-induced cell loss. However, the subsequent lymphopenia-induced numerical recovery of the CD4 T cell compartment leads to intrinsic alterations in phenotype and effector function, reduced repertoire diversity, changes in the composition of naive antigen-specific CD4 T cell pools, and changes in the representation of different CD4 T cell subpopulations (e.g., increases in Treg frequency). This review focuses on sepsis-induced alterations within the CD4 T cell compartment that influence the ability of the immune system to control secondary heterologous infections. The understanding of how sepsis affects CD4 T cells through their numerical loss and recovery, as well as function, is important in the development of future treatments designed to restore CD4 T cells to their presepsis state.
Collapse
Affiliation(s)
- Javier Cabrera-Perez
- Microbiology, Immunology, and Cancer Biology Graduate Program Medical Scientist Training Program
| | | | - Vladimir P Badovinac
- Department of Pathology and Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Thomas S Griffith
- Microbiology, Immunology, and Cancer Biology Graduate Program Center for Immunology, and Department of Urology, University of Minnesota Medical School, Minneapolis, Minnesota, USA; Minneapolis Veterans Administration Health Care System, Minneapolis, Minnesota, USA; and
| |
Collapse
|
21
|
Abstract
Major burn triggers immune dysfunction, which is associated with wound healing complications. Gamma-δ T-cells have been shown to be important in postburn inflammation and wound healing; however, their cytokine phenotype at the burn wound site is unknown. C57BL/6 male mice were subjected to a major burn (25% TBSA, third degree) or sham treatment. At 3 hours, 3 days, and 7 days thereafter, skin samples were collected and subjected to dispase and trypsin digestion to isolate single cells. The cells were phenotyped and evaluated for cytokine profiles by flow cytometry. Th1 cells were defined as interferon (IFN)γ positive, Th2 cells were defined as interleukin (IL)-10 positive, and Th17 cells were defined as IL-17 positive. At 7 days after burn a shift toward Th2 and Th17 positive T-cells at the wound site was observed. Further analysis revealed that at 3-hour postinjury the percentage of γδ T-cells positive for IFNγ, IL-10, and IL-17 were comparable between sham and burn skin samples. At 3 days and 7 days postinjury the percentage of cells positive for each cytokine increased; however, the increase was significantly greater for IL-10 and IL-17, as compared with IFNγ (ie, 9-20-fold vs 3-fold). Skin αβ T-cells preferentially produced IFNγ (~20%), which was unaffected by burn injury. These data demonstrate that burn wound γδ T-cells are activated for enhanced cytokine production and display a shift toward a Th2 and/or Th17 phenotype. In contrast, burn wound αβ T-cells were not activated for enhanced cytokine production.
Collapse
|
22
|
Abstract
Altered immune status of blood leukocytes is a general phenomenon observed in adult patients with sepsis or septic shock. This is also the case in children with septic shock for both T helper 1 and T helper 2 lymphocytes, as demonstrated by their reduced ex vivo cytokine production upon activation by phytohemagglutinin.
Collapse
Affiliation(s)
- Jean-Marc Cavaillon
- Unité Cytokines & Inflammation, Institut Pasteur, 28 Rue Dr. Roux, 75014, Paris, France.
| |
Collapse
|
23
|
The central role of arginine catabolism in T-cell dysfunction and increased susceptibility to infection after physical injury. Ann Surg 2014; 259:171-8. [PMID: 23470573 DOI: 10.1097/sla.0b013e31828611f8] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVE To explore the hypothesis that decreased arginine availability by myeloid-derived suppressor cells (MDSCs) is a cause of T-cell dysfunction after physical injury (PI). BACKGROUND Arginine is an essential amino acid for normal T-cell function whose availability becomes limited after PI. MDSCs expressing arginase 1 are induced by PI. T-cell dysfunction after PI seems to increase the risk of infection but the mechanisms that cause it are unclear. METHODS PI was created using a standard laparotomy model. Phenotypical and functional alterations in T cells were evaluated in vivo. MDSCs expressing arginase 1 were measured by flow cytometry. Infection after PI was created by intraperitoneal injection of Listeria monocytogenes. N-Hydroxy-Nor-L-arginine (Nor-NOHA) was used as an arginase inhibitor. The effect of arginine depletion on T-cell function and susceptibility to infection was assessed through adoptive transfer of MDSC or injection of arginase into noninjured mice. RESULTS PI caused a decrease in intracellular arginine in T cells, loss of the T-cell receptor (TCR) CD3-ζ chain, inhibition of in vivo T-cell proliferation, memory, and cytotoxicity. PI exponentially increased bacterial growth and mortality to L. monocytogenes. T-cell dysfunction and increased infection were reversed by arginase inhibitor Nor-NOHA but were reproduced by adoptively transferring MDSC or injecting arginase 1 to noninjured mice. CONCLUSIONS Arginine availability is decreased after PI coinciding with an induction of MDSC expressing arginase 1. Decreased arginine may inhibit T-cell function and increase susceptibility to infection after injury.
Collapse
|
24
|
Osuka A, Ogura H, Ueyama M, Shimazu T, Lederer JA. Immune response to traumatic injury: harmony and discordance of immune system homeostasis. Acute Med Surg 2014; 1:63-69. [PMID: 29930824 DOI: 10.1002/ams2.17] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 11/19/2013] [Indexed: 01/27/2023] Open
Abstract
Trauma remains one of the leading causes of death worldwide. Traumatic injury disrupts immune system homeostasis and may predispose patients to opportunistic infections and inflammatory complications. Prevention of multiple organ dysfunction syndrome due to septic complications following severe trauma is a challenging problem. Following severe injury, the immune system usually tends toward a pro-inflammatory phenotype and then changes to a counter-inflammatory phenotype. This immune system homeostasis is believed to be a protective response based on the balance between the innate and adaptive immune systems. We reported that injury activates inflammasomes and primes Toll-like receptors. The primed innate immune system is prepared for a rapid and strong antimicrobial immune defense. However, trauma can also develop the "two-hit" response phenotype. We also reported that injury augments regulatory T cell activity, which can control the "two-hit" response phenotype in trauma. We discuss the current idea that traumatic injury induces a unique type of innate and adaptive immune response that may be triggered by damage-associated molecular pattern molecules, which are a combination of endogenous danger signal molecules that include alarmins and pathogen-associated molecular pattern molecules.
Collapse
Affiliation(s)
- Akinori Osuka
- Department of Trauma, Critical Care Medicine and Burn Center Social Insurance Chukyo Hospital Nagoya Japan.,Department of Traumatology and Acute Critical Medicine Osaka University Graduate School of Medicine Suita Japan.,Department of Surgery (Immunology) Brigham and Women's Hospital/Harvard Medical School Boston Massachusetts
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine Osaka University Graduate School of Medicine Suita Japan
| | - Masashi Ueyama
- Department of Trauma, Critical Care Medicine and Burn Center Social Insurance Chukyo Hospital Nagoya Japan
| | - Takeshi Shimazu
- Department of Traumatology and Acute Critical Medicine Osaka University Graduate School of Medicine Suita Japan
| | - James A Lederer
- Department of Surgery (Immunology) Brigham and Women's Hospital/Harvard Medical School Boston Massachusetts
| |
Collapse
|
25
|
Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol 2013; 13:862-74. [PMID: 24232462 PMCID: PMC4077177 DOI: 10.1038/nri3552] [Citation(s) in RCA: 1781] [Impact Index Per Article: 148.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Sepsis - which is a severe life-threatening infection with organ dysfunction - initiates a complex interplay of host pro-inflammatory and anti-inflammatory processes. Sepsis can be considered a race to the death between the pathogens and the host immune system, and it is the proper balance between the often competing pro- and anti-inflammatory pathways that determines the fate of the individual. Although the field of sepsis research has witnessed the failure of many highly touted clinical trials, a better understanding of the pathophysiological basis of the disorder and the mechanisms responsible for the associated pro- and anti-inflammatory responses provides a novel approach for treating this highly lethal condition. Biomarker-guided immunotherapy that is administered to patients at the proper immune phase of sepsis is potentially a major advance in the treatment of sepsis and in the field of infectious disease.
Collapse
Affiliation(s)
- Richard S Hotchkiss
- Department of Anesthesiology, Medicine, and Surgery, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | |
Collapse
|
26
|
Heffernan DS, Monaghan SF, Chung CS, Cioffi WG, Gravenstein S, Ayala A. A divergent response of innate regulatory T-cells to sepsis in humans: circulating invariant natural killer T-cells are preserved. Hum Immunol 2013; 75:277-82. [PMID: 24269692 DOI: 10.1016/j.humimm.2013.11.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 11/05/2013] [Accepted: 11/11/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND Sepsis is associated with severe immunosuppression, evidenced by loss and dysfunction of CD3(+) lymphocytes and γδ-TCR(+) T-cells. There is limited data addressing changes in the invariant natural killer T-(iNKT) cell population with sepsis, and whether such changes correlate with clinical outcomes. Specifically, septic geriatric patients have marked mortality. How γδ-TCR(+) T-cells and iNKT-cells are altered in the settings of sepsis and advanced age, and how these changes correlate with mortality are unknown. METHODS 49 young (18-50years) and 55 geriatric (>65years) ICU patients with confirmed sepsis were enrolled. Blood was stained with antibodies to detect the percentage and absolute number of CD3(+) (T-cells), γδ-TCR(+) T-cell, TCR-Vα-24(+) (iNKT-cells), and CD69(+) (marker of cell activation). Blood from 10 healthy controls was also collected. RESULTS Septic patients displayed marked leukocytosis, decreased CD3(+) lymphocytes, and γδ-TCR(+) T-cells, and increased percentage and number of iNKT-cells. Young and geriatric patients had similar degree of leukocytosis, along with percentage, number, and %CD69(+) CD3(+) T-cell and γδ-TCR(+) T-cells; however, percentage, number, and %CD69(+)iNKT-cells were most markedly elevated in geriatric patients. Geriatric non-survivors had higher percentage and number of, but decreased %CD69(+), iNKT-cells vs survivors. CONCLUSIONS iNKT-cells are increased in sepsis, suggesting that they typify an evolving morbid state. This is most pronounced in geriatric non-survivors, a group demonstrating dysfunctional regulatory iNKT-cell phenotype.
Collapse
Affiliation(s)
- Daithi S Heffernan
- Division of Surgical Research, Department of Surgery, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, United States.
| | - Sean F Monaghan
- Division of Surgical Research, Department of Surgery, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, United States
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, United States
| | - William G Cioffi
- Division of Surgical Research, Department of Surgery, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, United States
| | - Stefan Gravenstein
- Division of Geriatrics, Department of Medicine, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, United States
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, United States
| |
Collapse
|
27
|
Trauma patients' elevated tumor necrosis related apoptosis inducing ligand (TRAIL) contributes to increased T cell apoptosis. Clin Immunol 2012; 145:44-54. [PMID: 22926077 DOI: 10.1016/j.clim.2012.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 07/23/2012] [Accepted: 07/24/2012] [Indexed: 11/22/2022]
Abstract
Immunosuppression resulting from excessive post-trauma apoptosis of hyperactivated T cells is controversial. TRAIL mediated T cell apoptosis decreases highly activated T cells' responses. Caspase-10, a particular TRAIL target, was increased in trauma patients' T cells with concomitantly elevated plasma TRAIL levels. These patients' T cells developed anergy, implicating increased TRAIL-mediated T cell apoptosis in post-trauma T cell anergy. Control T cells cultured with patients' sera containing high TRAIL levels increased their caspase-10 activity and apoptosis. Stimulated primary T cells are TRAIL apoptosis resistant. Increased plasma thrombospondin-1 and T cell expression of CD47, a thrombospondin-1 receptor, preceded patients' T cell anergy. CD47 triggering of T cells increased their sensitivity to TRAIL-induced apoptosis. Augmentation of T cell TRAIL-induced apoptosis was secondary to CD47 triggered activation of the Src homology-containing phosphatase-1 (SHP-1) and was partially blocked by a SHP-1 inhibitor. We suggest that combined post-trauma CD47 triggering, SHP-1 mediated NFκB suppression, and elevated TRAIL levels increase patients' CD47 expressing T cell apoptosis, thus contributing to subsequent T cell anergy.
Collapse
|
28
|
Stoecklein VM, Osuka A, Lederer JA. Trauma equals danger--damage control by the immune system. J Leukoc Biol 2012; 92:539-51. [PMID: 22654121 DOI: 10.1189/jlb.0212072] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Traumatic injuries induce a complex host response that disrupts immune system homeostasis and predisposes patients to opportunistic infections and inflammatory complications. The response to injuries varies considerably by type and severity, as well as by individual variables, such as age, sex, and genetics. These variables make studying the impact of trauma on the immune system challenging. Nevertheless, advances have been made in understanding how injuries influence immune system function as well as the immune cells and pathways involved in regulating the response to injuries. This review provides an overview of current knowledge about how traumatic injuries affect immune system phenotype and function. We discuss the current ideas that traumatic injuries induce a unique type of a response that may be triggered by a combination of endogenous danger signals, including alarmins, DAMPs, self-antigens, and cytokines. Additionally, we review and propose strategies for redirecting injury responses to help restore immune system homeostasis.
Collapse
Affiliation(s)
- Veit M Stoecklein
- Department of Surgery, Brigham and Women’s Hospital/Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
29
|
Hawksworth JS, Graybill JC, Brown TS, Wallace SM, Davis TA, Tadaki DK, Elster EA. Lymphocyte modulation with FTY720 improves hemorrhagic shock survival in swine. PLoS One 2012; 7:e34224. [PMID: 22558085 PMCID: PMC3340389 DOI: 10.1371/journal.pone.0034224] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 02/25/2012] [Indexed: 01/08/2023] Open
Abstract
The inflammatory response to severe traumatic injury results in significant morbidity and mortality. Lymphocytes have recently been identified as critical mediators of the early innate immune response to ischemia-reperfusion injury. Experimental manipulation of lymphocytes following hemorrhagic shock may prevent secondary immunologic injury in surgical and trauma patients. The objective of this study is to evaluate the lymphocyte sequestration agent FTY720 as an immunomodulator following experimental hemorrhagic shock in a swine liver injury model. Yorkshire swine were anesthetized and underwent a grade III liver injury with uncontrolled hemorrhage to induce hemorrhagic shock. Experimental groups were treated with a lymphocyte sequestration agent, FTY720, (n = 9) and compared to a vehicle control group (n = 9). Animals were observed over a 3 day survival period after hemorrhage. Circulating total leukocyte and neutrophil counts were measured. Central lymphocytes were evaluated with mesenteric lymph node and spleen immunohistochemistry (IHC) staining for CD3. Lung tissue infiltrating neutrophils were analyzed with myeloperoxidase (MPO) IHC staining. Relevant immune-related gene expression from liver tissue was quantified using RT-PCR. The overall survival was 22.2% in the vehicle control and 66.7% in the FTY720 groups (p = 0.081), and reperfusion survival (period after hemorrhage) was 25% in the vehicle control and 75% in the FTY720 groups (p = 0.047). CD3+ lymphocytes were significantly increased in mesenteric lymph nodes and spleen in the FTY720 group compared to vehicle control, indicating central lymphocyte sequestration. Lymphocyte disruption significantly decreased circulating and lung tissue infiltrating neutrophils, and decreased expression of liver immune-related gene expression in the FTY720 treated group. There were no observed infectious or wound healing complications. Lymphocyte sequestration with FTY720 improves survival in experimental hemorrhagic shock using a porcine liver injury model. These results support a novel and clinically relevant lymphocyte immunomodulation strategy to ameliorate secondary immune injury in hemorrhagic shock.
Collapse
Affiliation(s)
- Jason S. Hawksworth
- Regenerative Medicine Department, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Department of Surgery, Walter Reed National Military Medical Center, Bethesda, Maryland, United States of America
| | - J. Christopher Graybill
- Regenerative Medicine Department, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Department of Surgery, Walter Reed National Military Medical Center, Bethesda, Maryland, United States of America
| | - Trevor S. Brown
- Regenerative Medicine Department, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, Maryland, United States of America
| | - Shannon M. Wallace
- Department of Diagnostic Pathology, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Thomas A. Davis
- Regenerative Medicine Department, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, Maryland, United States of America
| | - Doug K. Tadaki
- Regenerative Medicine Department, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Uniformed Services University of Health Sciences, Bethesda, Maryland, United States of America
- * E-mail: (EE); (DT)
| | - Eric A. Elster
- Regenerative Medicine Department, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Department of Surgery, Walter Reed National Military Medical Center, Bethesda, Maryland, United States of America
- Uniformed Services University of Health Sciences, Bethesda, Maryland, United States of America
- * E-mail: (EE); (DT)
| |
Collapse
|
30
|
Lye AD, Hayslip JW. Immunonutrition: does it have a role in improving recovery in patients receiving a stem cell transplant? Nutr Cancer 2012; 64:503-7. [PMID: 22519362 DOI: 10.1080/01635581.2012.675621] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Numerous clinical trials have demonstrated that immunomodulating diets (IMDs) reduce treatment complications such as the risk of acquired infections, length of hospital stay, and wound complications in patients receiving planned surgery. These complications are possibly exacerbated by malnutrition at the time of surgery, resulting in decreased cell-mediated and humoral immune responses, which can be improved with the utilization of IMDs both prior to and following surgery. Although numerous randomized studies have investigated IMDs in the surgical setting, IMDs have not been well studied to evaluate whether their use improves outcomes for other patient groups with high incidence of malnutrition and acquired infections. Patients receiving stem cell transplantation following preparative myeloablative chemotherapy for treatment of a hematologic malignancy would be a prime example of another patient group who would share these characteristics. Given the proposed mechanism of action by which IMDs have aided recovery after surgery, it is reasonable to expect that IMDs may aid recovery after stem cell transplantation, and current preclinical and clinical data support the need for further clinical studies.
Collapse
Affiliation(s)
- Adam D Lye
- University of Kentucky Markey Cancer Center, Hematology and Blood & Marrow Transplantation, Lexington, Kentucky 40536, USA.
| | | |
Collapse
|
31
|
Jackman RP, Utter GH, Muench MO, Heitman JW, Munz MM, Jackman RW, Biswas HH, Rivers RM, Tobler LH, Busch MP, Norris PJ. Distinct roles of trauma and transfusion in induction of immune modulation after injury. Transfusion 2012; 52:2533-50. [PMID: 22452342 DOI: 10.1111/j.1537-2995.2012.03618.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Trauma and transfusion can both alter immunity, and while transfusions are common among traumatically injured patients, few studies have examined their combined effects on immunity. STUDY DESIGN AND METHODS We tracked the plasma levels of 41 immunomodulatory proteins in 56 trauma patients from time of injury up to 1 year later. In addition, a murine model was developed to distinguish between the effects of transfusion and underlying injury and blood loss. RESULTS Thirty-one of the proteins had a significant change over time after traumatic injury, with a mixed early response that was predominantly anti-inflammatory followed by a later increase in proteins involved in wound healing and homeostasis. Results from the murine model revealed similar cytokine responses to humans. In mice, trauma and hemorrhage caused early perturbations in a number of the pro- and anti-inflammatory mediators measured, and transfusion blunted early elevations in interleukin (IL)-6, IL-10, matrix metalloproteinase-9, and interferon-γ. Transfusion caused or exacerbated changes in monocyte chemotactic protein-1, IL-1α, IL-5, IL-15, and soluble E-selectin. Finally, trauma and hemorrhage alone increased CXCL1 and IL-13. CONCLUSIONS This work provides a detailed characterization of the major shift in the immunologic environment in response to trauma and transfusion and clarifies which immune mediators are affected by trauma and hemorrhage and which by transfusion.
Collapse
Affiliation(s)
- Rachael P Jackman
- Blood Systems Research Institute, San Francisco, California 94118, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Guignant C, Lepape A, Huang X, Kherouf H, Denis L, Poitevin F, Malcus C, Chéron A, Allaouchiche B, Gueyffier F, Ayala A, Monneret G, Venet F. Programmed death-1 levels correlate with increased mortality, nosocomial infection and immune dysfunctions in septic shock patients. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2011; 15:R99. [PMID: 21418617 PMCID: PMC3219369 DOI: 10.1186/cc10112] [Citation(s) in RCA: 237] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 03/03/2011] [Accepted: 03/21/2011] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Septic shock remains a major health care problem worldwide. Sepsis-induced immune alterations are thought to play a major role in patients' mortality and susceptibility to nosocomial infections. Programmed death-1 (PD-1) receptor system constitutes a newly described immunoregulatory pathway that negatively controls immune responses. It has recently been shown that PD-1 knock-out mice exhibited a lower mortality in response to experimental sepsis. The objective of the present study was to investigate PD-1-related molecule expressions in septic shock patients. METHODS This prospective and observational study included 64 septic shock patients, 13 trauma patients and 49 healthy individuals. PD-1-related-molecule expressions were measured by flow cytometry on circulating leukocytes. Plasmatic interleukin (IL)-10 concentration as well as ex vivo mitogen-induced lymphocyte proliferation were assessed. RESULTS We observed that septic shock patients displayed increased PD-1, PD-Ligand1 (PD-L1) and PD-L2 monocyte expressions and enhanced PD-1 and PD-L1 CD4+ T lymphocyte expressions at day 1-2 and 3-5 after the onset of shock in comparison with patients with trauma and healthy volunteers. Importantly, increased expressions were associated with increased occurrence of secondary nosocomial infections and mortality after septic shock as well as with decreased mitogen-induced lymphocyte proliferation and increased circulating IL-10 concentration. CONCLUSIONS These findings indicate that PD-1-related molecules may constitute a novel immunoregulatory system involved in sepsis-induced immune alterations. Results should be confirmed in a larger cohort of patients. This may offer innovative therapeutic perspectives on the treatment of this hitherto deadly disease.
Collapse
Affiliation(s)
- Caroline Guignant
- Hospices Civils de Lyon, Hôpital E, Herriot, Laboratoire d'Immunologie, 5 Place d'Arsonval, 69003 Lyon, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
BACKGROUND Myeloid cells that express arginase 1 are upregulated by different stimuli, including trauma, and are capable of depleting arginine from the surrounding environment. Through arginine depletion, myeloid cells are capable of regulating T-cell function. We have previously reported increased arginase 1 expression in the peripheral blood mononuclear cells (PBMCs) after injury. The nature of the cells expressing arginase in humans after trauma is unknown and is the focus of this article. METHODS PBMCs were isolated using a Ficoll-Hypaque gradient. Arginase activity was measured by conversion of arginine to ornithine, and arginase 1 protein expression was measured by Western blot. The percent CD16 granulocytes and phenotypical analysis of the cells present in PBMCs were determined by flow cytometry. Magnetic microbeads were used for isolation and exclusion of specific cell subpopulations. RESULTS Trauma patients exhibited a dramatic increase in arginase activity (p < 0.05) and an increased percentage of CD16 granulocytes in the PBMC layer (p < 0.05) compared with control volunteers. Increased arginase activity in the PBMC layer was due to the contamination of this layer by granulocytes, as their exclusion decreased arginase activity back to baseline (p < 0.05). Granulocytes isolated from the PBMC layer expressed increased CD11b (p < 0.05) and CD66b (p < 0.05), markers of granulocyte activation. Furthermore, these granulocytes were significantly more swollen and degranulated compared with noncontaminating granulocytes. CONCLUSION In humans, increased arginase 1 expression after trauma observed in the PBMC layer seems to be exclusively the result of an increased number of activated granulocytes.
Collapse
|
34
|
Bohannon JK, Cui W, Toliver-Kinsky T. Endogenous Fms-like tyrosine kinase-3 ligand levels are not altered in mice after a severe burn and infection. BMC Immunol 2009; 10:47. [PMID: 19715582 PMCID: PMC2744912 DOI: 10.1186/1471-2172-10-47] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Accepted: 08/28/2009] [Indexed: 01/14/2023] Open
Abstract
Background Fms-like tyrosine kinase-3 ligand (Flt3L) is a hemopoietic cytokine and dendritic cell (DC) growth factor that promotes the proliferation and differentiation of progenitor cells into DCs. We have previously found that treatment of severely burned mice with recombinant Flt3L significantly enhances DC production and bacterial clearance from infected burn wounds, and increases global immune cell activation and survival in response to a burn wound infection. These significant benefits of Flt3L treatment after burn injury have prompted the question of whether or not severe burn injury induces deficits in endogenous Flt3L levels that could affect DCs and subsequent responses to infection. Results To address this, male BALB/c mice received a 30% total body surface area scald burn. Blood, spleens, and wound-draining lymph nodes were harvested at various time-points after injury. Some mice received a wound inoculation with P. aeruginosa. Murine Flt3L and G-CSF levels were measured by ELISA. Burn injury had no significant effect on Flt3L levels at any post-burn time-point examined compared to normal Flt3L levels in the sera, spleen, or lymph nodes. Additionally, Flt3L levels in the sera, spleen, and lymph nodes were not significantly altered when wounds were inoculated on the day of burn injury or at post-burn time points examined. Alternatively, levels of G-CSF were increased in response to burn injury and burn wound infection. Additionally, DC numbers and functions were not altered following burn injury alone. There was no significant difference between the number of DCs in the spleens of sham-injured mice and mice at 5 days after burn injury. When naïve T cells from sham-injured mice were co-cultured with DCs from either sham- or burn-injured mice, IFN-γ production was similar, however, IFN-γ levels produced by T cells harvested from burn-injured mice were significantly lower than those produced by T cells from sham mice, regardless of which DC group, sham or burn, was used in the coculture. Conclusion These data suggest that the beneficial effects of Flt3L treatments after burn injury are not due to correction of a burn-associated Flt3L deficiency but rather, are likely due to supplementary stimulation of DC production and immune responses to infection.
Collapse
Affiliation(s)
- Julia K Bohannon
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| | | | | |
Collapse
|
35
|
Burn injury induces the expression of cystine/glutamate transporter (x(c)(-)) in mouse T cells. Immunol Lett 2009; 125:137-44. [PMID: 19576933 DOI: 10.1016/j.imlet.2009.06.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Revised: 06/12/2009] [Accepted: 06/23/2009] [Indexed: 01/18/2023]
Abstract
System x(c)(-) transporter, formed by the association of CD98 and xCT proteins, regulates the import of cystine into cells and is poorly expressed in T lymphocytes. Thermal injury is associated with high oxidative stress, decreased levels of glutathione (GSH) and protein deficiency, all described as promoters of xCT expression and system x(c)(-) activity. T cell dysfunction is a consequence of thermal injury and has been related to oxidative stress. In order to evaluate if thermal injury induced system x(c)(-) expression in splenic T lymphocytes, cells were isolated from sham- and burn-injured mice at day 10 post-burn and cultured in 2-mercaptoethanol (2-ME)-rich and -free media. Isolated splenic T cells were stimulated and cell proliferation, system x(c)(-) expression and cystine transport activity were measured. Our results demonstrate that only burn-injured T cells express xCT and proliferate in (2-ME)-free media. In these cells, viability and CD25 expression was higher than control T cells. x(c)(-) system expression was responsible for significantly higher (14)C-cystine uptake by burn-injured T cells and its inhibition by sulfasalazine (SASP) decreased significantly their proliferation. Overall, these results demonstrate that xCT expression is induced by thermal injury in T lymphocytes and that cystine import by x(c)(-) leads to T cell dysfunction.
Collapse
|
36
|
Resuscitation with hydroxyethyl starch solution prevents CD4+ T-lymphocyte apoptosis and modulates the balance of T helper type 1 and T helper type 2 responses in the rat with traumatic virgule/shill hemorrhagic shock. Shock 2009; 30:692-8. [PMID: 18461021 DOI: 10.1097/shk.0b013e31816f260d] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Trauma/hemorrhagic shock (TH/S) has been associated with inflammation and immunodisorders, leading to immunosuppression, multiorgan dysfunction, and death. However, little is known about the effect of resuscitation with different solutions on the immunological function. To address this issue, groups of male Sprague-Dawley rats were induced with TH/S by fracture in the left femur and continual bleeding to keep the MAP of 30 +/- 5 mmHg for 30 min, followed by resuscitation with 6% hydroxyethyl starch solution (HES), Ringer's lactate solution (RS), or 5% albumin (ALB), and the impact of resuscitation on the activation, differentiation, and survival of CD4 T cells was longitudinally examined after TH/S and resuscitation. After resuscitation, the MAP, as expected, gradually increased regardless of the type of fluids transfused. The percentage of CD4+ T cells decreased to 20% to 25%, and the ratio of T helper type 1 (TH1)/TH2 responses was significantly reduced in all TH/S rats, however, resuscitation with HES alone reversed the trends (49.4% +/- 9.7% vs. 55.2% +/- 2.6% in sham for CD4 T cells; 0.64 +/- 0.23 vs. 0.71 +/- 0.16 in sham for the ratio of TH1/TH2, P > 0.05 for both). Treatment with HES or ALB, but not RS, prevented CD4 T-cell apoptosis (sham, 7.23% +/- 3.4%; HES, 10.2% +/- 4.1%; RS, 15.2% +/- 5.4%; ALB, 10.6% +/- 4.3%; 48 h) and nuclear factor-kappaB p65 activation (sham, 0.17 +/- 0.04; HES, 0.34 +/- 0.05; RS, 0.41 +/- 0.09; ALB, 0.25 +/- 0.09; 48 h) induced by TH/S early after resuscitation. These data demonstrated that HES resuscitation modulated the balance of TH1 and TH2 responses and inhibited TH/S-related nuclear factor-kappaB activation and CD4 T-cell apoptosis in TH/S rats. Our findings provide new insights into understanding the TH/S-related immunodisorders and may aid in the design of new therapy for intervention of TH/S.
Collapse
|
37
|
Fujimi S, Lapchak PH, Zang Y, MacConmara MP, Maung AA, Delisle AJ, Mannick JA, Lederer JA. Murine dendritic cell antigen-presenting cell function is not altered by burn injury. J Leukoc Biol 2009; 85:862-70. [PMID: 19228816 DOI: 10.1189/jlb.0408257] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Severe injury disrupts normal immune regulation causing a transient hyperinflammatory reaction and suppressed adaptive immune function. This report addresses the potential contribution of dendritic cells (DC) to changes in adaptive immune function after injury by specifically measuring injury-induced changes in splenic DC numbers and subsets, cell-surface markers, TLR responses, and APC function. Using a mouse burn injury model, we found that injury did not markedly alter the relative percentage of lymphoid, myeloid, or plasmacytoid DC in the spleens of burn-injured mice. Moreover, we did not observe a significant reduction in cell-surface expression of several major costimulatory molecules, CD40, CD80, CD86, programmed death 1 ligand, ICOS ligand, and B7-H3, on DC. Instead, we observed increased cell-surface expression of CD86 at 1 day after injury with no significant changes in costimulatory molecule expression at 7 days after injury, suggesting that burn injury causes an early activation of DC. In addition, injury did not suppress DC reactivity to TLR2, TLR4, or TLR9 agonists. Most important, DC prepared from injured mice were able to present peptide antigen to naive OTII TCR transgenic CD4+ T cells as efficiently and effectively as DC from sham-injured mice. We also found that CD4 T cells stimulated with antigen presented by DC from sham or burn mice showed similar levels of IL-2, IFN-gamma, IL-10, and IL-13 production. Taken together, these findings support the conclusion that DC do not acquire a suppressive phenotype following severe injury in mice.
Collapse
Affiliation(s)
- Satoshi Fujimi
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
BACKGROUND Major trauma induces a dysregulation of immune response supported in parts by lymphocyte dysfunction. Controversial data about a shift within the T-helper cell subsets Th1/Th2 are reported. METHODS To prove whether Th1/Th2-type cytokine plasma levels reflect the postulated Th2 shift after trauma, we investigated in a retrospective study 195 severely injured patients (47 women, 148 men; mean age 39.7 +/- 15.8 years; Injury Severity Score 32.0 +/- 11.3 points; overall 1,887 samples) during their ICU stay posttrauma. Mortality rate was 19%. Th1-type cytokines interleukin 2 (IL-2), interferon gamma, IL-12 (p70), and IL-18 and Th2-type cytokines IL-4, IL-10, and IL-11 were determined using the enzyme-linked immunosorbant assay technique in patients and in healthy controls. RESULTS IL-2 and interferon gamma were seldom detectable. All other mediators were significantly increased matched to controls (p < 0.05). All cytokines were elevated most prominent during weeks 1 and 2 posttrauma and declined thereafter. A trend toward lower levels in nonsurvivors was seen for both groups of cytokines. However, significant differences were only seen for Injury Severity Score, age, white blood cells, and C-reactive protein. All mediators correlated positively with each other (p < 0.01), a Th2-type shift was not observed. Two groups of patients were identified: one group with generally high plasma levels of all cytokines investigated and a second group of nonresponders who presented with low or diminished plasma levels in which most nonsurvivors were found. CONCLUSION We conclude that in plasma no Th1/Th2 shift can be observed after major trauma.
Collapse
|
39
|
Venet F, Chung CS, Kherouf H, Geeraert A, Malcus C, Poitevin F, Bohé J, Lepape A, Ayala A, Monneret G. Increased circulating regulatory T cells (CD4(+)CD25 (+)CD127 (-)) contribute to lymphocyte anergy in septic shock patients. Intensive Care Med 2008; 35:678-86. [PMID: 18946659 DOI: 10.1007/s00134-008-1337-8] [Citation(s) in RCA: 234] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2008] [Accepted: 10/04/2008] [Indexed: 12/16/2022]
Abstract
PURPOSE Sepsis syndrome represents the leading cause of death in intensive care unit. Patients present features consistent with a decline in immune responsiveness potentially contributing to mortality. We investigated whether CD4(+)CD25(+) regulatory T cells (Treg) participate in the induction of lymphocyte anergy after sepsis. METHOD Observational study in septic shock patients and experimental study in mice. RESULTS We took advantage of the recently described flow cytometric gating strategy using the measurement of CD25 and CD127 expressions for monitoring Treg (CD4(+)CD25(+)CD127(-)Foxp3(+)). In patients the increased circulating Treg percentage significantly correlated with a decreased lympho-proliferative response. In a murine model of sepsis mimicking these observations, the ex vivo downregulation of Foxp3 expression using siRNA was associated with a restoration of this response. CONCLUSION The relative increase in circulating Treg might play a role in lymphocyte anergy described after septic shock and represent a standardizable surrogate marker of declining proliferative capacity after sepsis.
Collapse
Affiliation(s)
- Fabienne Venet
- Division of Surgical Research, Rhode Island Hospital, Brown University, Providence, RI, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Miller AC, Rashid RM, Elamin EM. The "T" in trauma: the helper T-cell response and the role of immunomodulation in trauma and burn patients. ACTA ACUST UNITED AC 2008; 63:1407-17. [PMID: 18212667 DOI: 10.1097/ta.0b013e31815b839e] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND The immune system undergoes numerous changes after traumatic and burn injuries, including a down-regulation of the TH1 response and up-regulation of the TH2 response. METHODS The PubMed, Medline, and Ovid search engines were queried for relevant human studies. Bibliographies were also meticulously reviewed. RESULTS An interesting and potentially clinically relevant pattern of immune dysregulation exists after injury. The type 1 T-helper cell (TH1) response is suppressed as illustrated by diminished interleukin (IL)-2, interferon (IFN)-gamma, and IL-12 levels after major injury. The enhancement of the TH2 response is marked by elevated IL-10 and IL-4. Certain cytokine profiles, ratios, and polymorphisms may help identify patients at increased risk of systemic inflammatory response syndrome (SIRS), sepsis, multiple organ failure (MOF), and deep venous thrombosis. The most promising prognostic indicators to date are decreased production of the IL-12, elevated IL-10, soluble IL-2 receptor-a (sIL-2Ra), and IL-18 levels, IL-18 promoter polymorphisms, the IL-6:IL-10 ratio, and the degree of TH1 suppression as measured by diminished IL-2 and IFN-gamma. The Cytokine sections are subdivided into basic science, human studies, animal studies (when necessary), and directed therapeutics. The outcomes of sepsis, SIRS, or MOF are generally discussed together. CONCLUSIONS T-cell-specific immunologic changes occur after major injury. Identification of those TH1/TH2 cytokine profiles associated with worse prognosis may one day allow clinicians to risk stratify injured patients and identify those at increased risk of developing SIRS, sepsis, MOF, and deep venous thrombosis. Immune-targeted therapies may eventually serve as effective treatments in the acute setting to decrease morbidity and mortality, and to improve the management and prognosis of those patients at risk for developing postinjury complications.
Collapse
Affiliation(s)
- Andrew C Miller
- Department of Emergency Medicine and Internal Medicine, The State University of New York Downstate Medical Center and Kings County Hospital Center, Brooklyn, New York, USA.
| | | | | |
Collapse
|
41
|
Monneret G, Venet F, Pachot A, Lepape A. Monitoring immune dysfunctions in the septic patient: a new skin for the old ceremony. Mol Med 2008; 14:64-78. [PMID: 18026569 PMCID: PMC2078557 DOI: 10.2119/2007-00102.monneret] [Citation(s) in RCA: 250] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2007] [Accepted: 11/06/2007] [Indexed: 12/16/2022] Open
Abstract
Septic syndromes represent a major although largely underrecognized healthcare problem worldwide, accounting for thousands of deaths every year. It is now agreed that sepsis deeply perturbs immune homeostasis by inducing an initial tremendous systemic inflammatory response which is accompanied by an antiinflammatory process, acting as negative feedback. This compensatory inhibitory response secondly becomes deleterious as nearly all immune functions are compromised. These alterations might be directly responsible for worsening outcome, as they may play a major role in the decreased resistance to nosocomial infections in patients who survived initial resuscitation. Consequently, immunostimulatory therapies may now be assessed for the treatment of sepsis. This review focuses on immune dysfunctions described in septic patients and on their potential use as markers on a routine standardized basis for prediction of adverse outcome or of occurrence of secondary nosocomial infections. This constitutes a prerequisite to a staging system for individualized treatment for these hitherto deadly syndromes.
Collapse
Affiliation(s)
- Guillaume Monneret
- Hospices civils de Lyon, Immunology laboratory, Hopital E. Herriot, Lyon, France.
| | | | | | | |
Collapse
|
42
|
Bandyopadhyay G, De A, Laudanski K, Li F, Lentz C, Bankey P, Miller-Graziano C. Negative signaling contributes to T-cell anergy in trauma patients. Crit Care Med 2007; 35:794-801. [PMID: 17255857 DOI: 10.1097/01.ccm.0000256847.61085.a5] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Maintenance of postinjury T-lymphocyte immune paralysis or anergy could result from failure to activate costimulatory receptors during T-cell receptor activation and/or from chronic stimulation of a competing set of elevated corepressor receptors. Our objective was to assess whether elevated posttrauma T-lymphocyte surface expression of corepressor receptors was associated with immunodepressed lymphocyte responses and corresponded to increased inhibitory and decreased activating signal transduction molecules. DESIGN Prospective observational study. SETTING University trauma intensive care unit and research laboratory. PATIENTS Sixty-one severe thermal and mechanical trauma patients. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Isolated trauma patients' and controls' peripheral blood T cells were assayed for negative and positive costimulation receptor expression. These receptor expression levels were compared (flow cytometry) between the two groups and correlated to T-cell levels of inhibitory and activating signal transduction molecules and proliferation capacity. Patients' proliferation hyporesponsive (anergic) T cells had increased expression of novel inhibitory receptors (corepressors) PD-1 (p < .05) and CD47 (p < .05) vs. patients' T-cell proliferation competent or controls' T cells. Patients' T-cell CD152 (CTLA-4) expression was also elevated vs. controls. Only patients' anergic T cells had simultaneously increased levels of the inhibitory signal transduction proteins, c-Cbl, a ubiquitin-ligase (p < .01) and SHP-1, a phosphatase (p < .01), concomitant to depressed phosphorylation of the activating signal kinases Erk, Zap70, and CD3Euro. T-cell receptor complex phosphorylation and activation of the interleukin-2 pivotal transcriptional complex protein CREB were also simultaneously depressed as c-Cbl and SHP-1 were elevated. CONCLUSIONS Up-regulated corepressor receptor expression is novelly shown to characterize trauma patients' anergic T cells and correlate with predominance of inhibitory overactivating signal transduction molecules during T-cell stimulation. This could contribute to postinjury immunosuppression.
Collapse
Affiliation(s)
- Gautam Bandyopadhyay
- University of Rochester Medical Center, Department of Surgery, Rochester, NY, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Nakano T, Araki K, Nakatani H, Kobayashi M, Sugimoto T, Furuya Y, Matsuoka T, Jin T, Hanazaki K. Effects of geldanamycin and thalidomide on the Th1/Th2 cytokine balance in mice subjected to operative trauma. Surgery 2007; 141:490-500. [PMID: 17383526 DOI: 10.1016/j.surg.2006.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Revised: 10/12/2006] [Accepted: 10/14/2006] [Indexed: 11/18/2022]
Abstract
BACKGROUND Persistence of postoperative immune dysfunction is a critical problem because it increases the risk of serious infectious complications. The mechanisms of the immune dysfunction that occur initially after non-thermal operative injury remain to be fully elucidated. METHODS Two mouse models of operative trauma (simple laparotomy to represent minor operative injury and ileocecal resection to represent major operative injury) were used to define the characteristics of initial cytokine synthesis. Geldanamycin and thalidomide were independently added intraperitoneally before and after operative injury to examine the effect on postoperative immune dysfunction. Mice were sacrificed at scheduled times (3, 6, 12, and 24 h after operative injury) and TNF-alpha, IL-2, IL-4, and IL-10 were analyzed. Spleen was used for intracellular cytokines and RT-PCR. Sera were used for ELISA. RESULTS Major operative injury caused an initial upregulation of IL-10 synthesis with delayed synthesis of TNF-alpha and IL-2. Minor operative injury caused an early induction of IL-2 synthesis preceded by an initial induction of IL-4 synthesis. GA caused a specific early upregulation of TNF-alpha mRNA expression and intracellular TNF-alpha synthesis. The GA and THD groups showed early serum IL-2 production with reduction of IL-10 mRNA expression and intracellular IL-10 synthesis in the early post-operative phase. CONCLUSIONS Major and minor operative injury showed different Th1/Th2 cytokine patterns in the initial post-operative period. Geldanamycin and thalidomide improved the Th1/Th2 imbalance independently after major operative injury.
Collapse
Affiliation(s)
- Takumi Nakano
- Department of Tumor Surgery, Kochi Medical School, Kochi University, Nankoku, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Franke A, Lante W, Markewitz A, Weinhold C. In Vitro Restoration of Post-Operatively Decreased IFN-Gamma Levels After Cardiac Surgery and Its Effect on Pro- and Anti-Inflammatory Mediators. J Surg Res 2006; 136:266-72. [PMID: 17054992 DOI: 10.1016/j.jss.2006.06.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2006] [Revised: 06/12/2006] [Accepted: 06/13/2006] [Indexed: 11/22/2022]
Abstract
BACKGROUND A decreased synthesis of interferon gamma (IFN-gamma) by TH 1 lymphocytes after cardiac operations with cardiopulmonary bypass (CPB) is part of the inflammatory response to local operative and systemic traumas. The consequences of this mechanism on the release of pro- and anti-inflammatory cytokines remain unclear. To evaluate the role of IFN-gamma, we added recombinant IFN-gamma to peripheral blood mononuclear cells (PBMCs) on the first post-operative day in an attempt to restore pre-operative values and then measured the release of pro- and anti-inflammatory cytokines in vitro. METHODS PBMCs of 10 patients scheduled for elective coronary artery bypass grafting (CABG) were obtained pre-operatively (d0) and on the first (d1) and third (d3) post-operative days. The release of IL-6, IL-8, TNF-alpha, IFN-gamma, IL-10, IL-2, and IL-4 was studied after stimulation (48 h) with PHA (phytohemagglutinin) and LPS (lipopolysaccharide) in the absence or presence of recombinant human IFN-gamma. RESULTS Endogenous IFN-gamma synthesis was suppressed on d1. Adding exogenous IFN-gamma restored IFN-gamma levels to normal on d1 and doubled IFN-gamma levels on d0 and d3. The addition of IFN-gamma increased TNF-alpha levels up to 250% on d1 and IL-2 synthesis by 75% on d1 and d3; the IL-2 levels, however, were still significantly depressed. The addition of recombinant IFN-gamma did not affect the synthesis of IL-6, IL-8, IL-10, and IL-4. CONCLUSIONS Contrary to our expectations, the in vitro release of IL-6 and IL-8 as well as IL-10 and IL-4 was not influenced by the addition of IFN-gamma. However, TNF-alpha production in isolated PBMC cultures increased significantly on the first post-operative day. This may indicate a hyper-reactivity of PBMCs to IFN-gamma and suggests that the decrease in IFN-gamma synthesis might prevent an excessive stimulation of the non-specific immune system by high TNF-alpha levels after cardiac surgery.
Collapse
Affiliation(s)
- Axel Franke
- Department of Cardiovascular Surgery, Bundeswehr Central Hospital, Koblenz, Germany.
| | | | | | | |
Collapse
|
45
|
MacConmara MP, Maung AA, Fujimi S, McKenna AM, Delisle A, Lapchak PH, Rogers S, Lederer JA, Mannick JA. Increased CD4+ CD25+ T regulatory cell activity in trauma patients depresses protective Th1 immunity. Ann Surg 2006; 244:514-23. [PMID: 16998360 PMCID: PMC1856576 DOI: 10.1097/01.sla.0000239031.06906.1f] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVES We recently reported increased CD4 CD25 T regulatory (Treg) activity after burn injury in mice. This study sought to determine if Tregs mediate the reduction in TH1-type immunity after serious injury in man and if Treg function is altered by injury. METHODS Peripheral blood was withdrawn from 19 consenting adult patients (35.1 +/- 16.3 years of age) with Injury Severity Scores (ISS) 36.6 +/- 13.9 on days 1 and 7 after trauma and from 5 healthy individuals. CD4 T cells were purified and sorted into Treg (CD25(high)) and Treg-depleted populations. After activation of cells with anti-CD3/CD28 antibody, production of the TH1-type cytokine IFNgamma, TH2-type cytokines (IL-4 and IL-5), and the inhibitory cytokine IL-10 was measured using cytometric bead arrays. Treg activity was measured by in vitro suppression of autologous CD4 T cell proliferation. RESULTS All patients survived, 9 (47%) developed infection postinjury. IFNgamma production by patient CD4 T cells was decreased on day 1 and day 7, when compared with healthy controls. However, when Tregs were depleted from the CD4 T cells, the IFNgamma production increased to control levels. Tregs were the chief source of IL-4 and IL-5 as well as IL-10. Treg suppression of T cell proliferation increased significantly from day 1 to day 7 after injury. CONCLUSIONS We demonstrate for the first time that human Tregs are increased in potency after severe injury. Most significantly, Tregs are important mediators of the suppression of T cell activation and the reduction in TH1 cytokine production found after injury.
Collapse
Affiliation(s)
- Malcolm P MacConmara
- Department of Surgery (Immunology), Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Laudanski K, Miller-Graziano C, Xiao W, Mindrinos MN, Richards DR, De A, Moldawer LL, Maier RV, Bankey P, Baker HV, Brownstein BH, Cobb JP, Calvano SE, Davis RW, Tompkins RG. Cell-specific expression and pathway analyses reveal alterations in trauma-related human T cell and monocyte pathways. Proc Natl Acad Sci U S A 2006; 103:15564-9. [PMID: 17032758 PMCID: PMC1592643 DOI: 10.1073/pnas.0607028103] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2006] [Indexed: 01/30/2023] Open
Abstract
Monitoring genome-wide, cell-specific responses to human disease, although challenging, holds great promise for the future of medicine. Patients with injuries severe enough to develop multiple organ dysfunction syndrome have multiple immune derangements, including T cell apoptosis and anergy combined with depressed monocyte antigen presentation. Genome-wide expression analysis of highly enriched circulating leukocyte subpopulations, combined with cell-specific pathway analyses, offers an opportunity to discover leukocyte regulatory networks in critically injured patients. Severe injury induced significant changes in T cell (5,693 genes), monocyte (2,801 genes), and total leukocyte (3,437 genes) transcriptomes, with only 911 of these genes common to all three cell populations (12%). T cell-specific pathway analyses identified increased gene expression of several inhibitory receptors (PD-1, CD152, NRP-1, and Lag3) and concomitant decreases in stimulatory receptors (CD28, CD4, and IL-2Ralpha). Functional analysis of T cells and monocytes confirmed reduced T cell proliferation and increased cell surface expression of negative signaling receptors paired with decreased monocyte costimulation ligands. Thus, genome-wide expression from highly enriched cell populations combined with knowledge-based pathway analyses leads to the identification of regulatory networks differentially expressed in injured patients. Importantly, application of cell separation, genome-wide expression, and cell-specific pathway analyses can be used to discover pathway alterations in human disease.
Collapse
Affiliation(s)
- Krzysztof Laudanski
- Department of Surgery, University of Rochester School of Medicine, Rochester, NY 14642
| | - Carol Miller-Graziano
- Department of Surgery, University of Rochester School of Medicine, Rochester, NY 14642
| | - Wenzhong Xiao
- Stanford Genome Technology Center, Palo Alto, CA 94304
| | | | | | - Asit De
- Department of Surgery, University of Rochester School of Medicine, Rochester, NY 14642
| | | | - Ronald V. Maier
- Department of Surgery, Harborview Medical Center, University of Washington, Seattle, WA 98104
| | - Paul Bankey
- Department of Surgery, University of Rochester School of Medicine, Rochester, NY 14642
| | - Henry V. Baker
- Ingenuity Systems, Inc., Redwood City, CA 94063
- Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL 32610
| | | | | | - Steve E. Calvano
- Department of Surgery, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ 08903; and
| | | | - Ronald G. Tompkins
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| |
Collapse
|
47
|
Tredget EE, Yang L, Delehanty M, Shankowsky H, Scott PG. Polarized Th2 cytokine production in patients with hypertrophic scar following thermal injury. J Interferon Cytokine Res 2006; 26:179-89. [PMID: 16542140 DOI: 10.1089/jir.2006.26.179] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Following thermal injury, hypertrophic scar (HSc) is a frequent and severe form of fibrosis of the skin, which limits movement and compromises the cosmetic appearance and function of the skin. Prolonged pruritus and dysesthesia are also common problems in the previously injured, fibrotic tissues, as current understanding of the pathogenesis is limited, and few effective therapies exist, as with other fibroproliferative disorders (FPD). To investigate the role of T cells and their cytokines in the development of HSc, intracellular cytokine synthesis of circulating T cells was measured serially in burn patients using flow cytometry from the time of injury to over a 1-year period during which many patients developed HSc. Within 1 month of injury, low interferon-gamma (IFN-gamma)-positive T cells (Th1) were found in association with low interleukin-12 (IL-12) and absent IFN-gamma cytokine levels in the serum. IL-4-positive Th 2 cells, however, were significantly increased compared with normal controls by 2 months postinjury. In burn patients with HSc, serum IL-10 and transforming growth factor-beta (TGF-beta) levels were also significantly increased early after burn injury in patients who later developed HSc compared with normal volunteers and with a subset of burn patients who did not develop HSc, before returning to normal levels after 6 months. Activated peripheral blood mononuclear cells (PBMC) demonstrated that mRNA for IFN-gamma was present only in normal volunteers or patients without HSc but was undetectable in HSc patients. IL-4 mRNA levels were increased in the PBMCs of burn patients with HSc. In HSc tissues, IL-4 mRNA was increased, whereas, IFN-gamma mRNA was reduced compared with normal skin and mature scar. Increased CD3(+) and CD4(+) cells were present in HSc tissues compared with normal skin and were coexpressed with the fibrogenic cytokine TGF-beta. These longitudinal studies in human patients with HSc suggest that fibrosis in the skin is associated with a polarized Th2 systemic response to injury that leads to increased T cells and their Th2 fibrogenic cytokines in tissues and the development of fibrosis and HSc.
Collapse
Affiliation(s)
- Edward E Tredget
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery and Critical Care, University of Alberta, Edmonton, Alberta, T6G 2B7, Canada.
| | | | | | | | | |
Collapse
|
48
|
Makarenkova VP, Bansal V, Matta BM, Perez LA, Ochoa JB. CD11b+/Gr-1+ myeloid suppressor cells cause T cell dysfunction after traumatic stress. THE JOURNAL OF IMMUNOLOGY 2006; 176:2085-94. [PMID: 16455964 DOI: 10.4049/jimmunol.176.4.2085] [Citation(s) in RCA: 273] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
T cell dysfunction that occurs after surgery or trauma is associated with a poor clinical outcome. We describe that myeloid suppressor cells expressing CD11b(+)/Gr-1(+) markers invade the spleen after traumatic stress and suppress T cell function through the production of arginase 1. We created a consistent model of traumatic stress in C57BL/6 mice to perform this work. A significant number of CD11b(+)/Gr-1(+) cells expressing arginase 1 accumulated in T cell zones around the germinal centers of the white pulp of the spleen within 6 h of trauma and lasted for at least 72 h. Increased arginase activity and arginase 1 expression, along with increased [(3)H]arginine uptake, l-arginine depletion, and l-ornithine accumulation in the culture medium, were observed exclusively in CD11b(+)/Gr-1(+) cells after traumatic stress. Flow cytometry revealed CD11b(+)/Gr-1(+) as a heterogeneous myeloid suppressor cell also expressing low levels of MHC class I and II, CD80, CD86, CD31, and others. When compared with controls, trauma-induced CD11b(+)/Gr-1(+) cells significantly inhibited CD3/CD28-mediated T cell proliferation, TCR zeta-chain expression, and IL-2 production. The suppressive effects by trauma CD11b(+)/Gr-1(+) cells were overcome with the arginase antagonist N-hydroxy-nor-l-arginine or extrasupplementation of medium with l-arginine. Poor Ag-presenting capacity of control and trauma-induced CD11b(+)/Gr-1(+) cells was detected in allogeneic murine leukocyte reaction. This study demonstrates that CD11b(+)/Gr-1(+) cells invade the spleen following traumatic stress and cause T cell dysfunction by an arginase-mediated mechanism, probably that of arginine depletion. Understanding the mechanism of immune suppression by these cells has important clinical implications in the treatment of immune dysfunction after trauma or surgery.
Collapse
|
49
|
Laudanski K, De A, Pellegrini J, Miller-Graziano C. Simultaneous aberrations in MØ and T cell function adversely affect trauma patients' clinical outcome: A possible faulty IL-13 feedback loop. Clin Immunol 2006; 118:332-41. [PMID: 16303333 DOI: 10.1016/j.clim.2005.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2005] [Revised: 09/27/2005] [Accepted: 10/06/2005] [Indexed: 11/20/2022]
Abstract
Possible defective trauma patients' Mphi-T-cell feedback interactions between T cell IL-13 production and IL-1beta and IL-18 Mphi secretion were assessed. Mphi produced IL-1 and IL-18 augment T cell IL-13, which in turn limits excessive macrophage activation. Immunodepressed patients' T cells (depressed proliferation to alphaCD3 + alphaCD4) had decreased IL-13 production concomitant to aberrant Mphi activities ( upward arrow mTNFalpha, downward arrow IL-10) and consequent multiple organ failure (MOF). Decreased IL-13 levels in patients' T cell and diminished Mphi supernatant augmentation of healthy controls' T cell IL-13 production appeared concomitantly, suggesting patients' aberrant monokine levels might intensify in vivo T cell dysfunction severity. Patients' Mphi supernatants, which failed to augment controls' T cell IL-13 production, had depressed IL-1beta and lower induction of IL-18 than immunocompetent patients' Mphi, but combined addition of IL-1beta and IL-18 restored these Mphis' IL-13 enhancing activity. These data suggest that immunodepressed patients' aberrant monokine and depressed T cell IL-13 production are independent but synergistic contributors to emergence of MOF.
Collapse
Affiliation(s)
- Krzysztof Laudanski
- Department of Surgery, University of Rochester Medical Center, 601 Elmwood Ave.-SURG, Rochester, NY 14642, USA
| | | | | | | |
Collapse
|
50
|
Cavaillon JM, Adrie C, Fitting C, Adib-Conquy M. Reprogramming of circulatory cells in sepsis and SIRS. ACTA ACUST UNITED AC 2005. [PMID: 16263005 DOI: 10.1177/09680519050110050901] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Immune status is altered in patients with sepsis or non-infectious systemic inflammatory response syndrome (SIRS). Reduced ex-vivo TNF production by endotoxin-activated monocytes has been regularly reported. This observation is reminiscent of the phenomenon of endotoxin tolerance, and the term 'leukocyte reprogramming' well defines this phenomenon. This review will outline that the hyporesponsiveness of circulating leukocytes is not a generalized phenomenon in sepsis and SIRS. Indeed, the nature of the insult (i.e. infectious versus non-infectious SIRS; under anesthesia [surgery] or not [trauma, burn]), the nature of the activator used to trigger leukocytes (i.e. different Toll-like receptor ligands or whole bacteria), the nature of the cell culture (i.e. isolated monocytes versus peripheral blood mononuclear cells versus whole blood assays), and the nature of the analyzed cytokines (e.g. IL-1beta versus IL-1ra; TNF versus IL-10) have a profound influence on the outcome of the response.
Collapse
Affiliation(s)
- J-M Cavaillon
- UP Cytokines and Inflammation, Institut Pasteur, Paris, France.
| | | | | | | |
Collapse
|