1
|
Zhang T, Chen L, Kueth G, Shao E, Wang X, Ha T, Williams DL, Li C, Fan M, Yang K. Lactate's impact on immune cells in sepsis: unraveling the complex interplay. Front Immunol 2024; 15:1483400. [PMID: 39372401 PMCID: PMC11449721 DOI: 10.3389/fimmu.2024.1483400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 09/05/2024] [Indexed: 10/08/2024] Open
Abstract
Lactate significantly impacts immune cell function in sepsis and septic shock, transcending its traditional view as just a metabolic byproduct. This review summarizes the role of lactate as a biomarker and its influence on immune cell dynamics, emphasizing its critical role in modulating immune responses during sepsis. Mechanistically, key lactate transporters like MCT1, MCT4, and the receptor GPR81 are crucial in mediating these effects. HIF-1α also plays a significant role in lactate-driven immune modulation. Additionally, lactate affects immune cell function through post-translational modifications such as lactylation, acetylation, and phosphorylation, which alter enzyme activities and protein functions. These interactions between lactate and immune cells are central to understanding sepsis-associated immune dysregulation, offering insights that can guide future research and improve therapeutic strategies to enhance patient outcomes.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Linjian Chen
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Gatkek Kueth
- James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Emily Shao
- Program in Neuroscience, College of Arts and Science, Vanderbilt University, Nashville, TN, United States
| | - Xiaohui Wang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Tuanzhu Ha
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - David L. Williams
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Chuanfu Li
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Min Fan
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Kun Yang
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
2
|
Santacroce E, D’Angerio M, Ciobanu AL, Masini L, Lo Tartaro D, Coloretti I, Busani S, Rubio I, Meschiari M, Franceschini E, Mussini C, Girardis M, Gibellini L, Cossarizza A, De Biasi S. Advances and Challenges in Sepsis Management: Modern Tools and Future Directions. Cells 2024; 13:439. [PMID: 38474403 PMCID: PMC10931424 DOI: 10.3390/cells13050439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Sepsis, a critical condition marked by systemic inflammation, profoundly impacts both innate and adaptive immunity, often resulting in lymphopenia. This immune alteration can spare regulatory T cells (Tregs) but significantly affects other lymphocyte subsets, leading to diminished effector functions, altered cytokine profiles, and metabolic changes. The complexity of sepsis stems not only from its pathophysiology but also from the heterogeneity of patient responses, posing significant challenges in developing universally effective therapies. This review emphasizes the importance of phenotyping in sepsis to enhance patient-specific diagnostic and therapeutic strategies. Phenotyping immune cells, which categorizes patients based on clinical and immunological characteristics, is pivotal for tailoring treatment approaches. Flow cytometry emerges as a crucial tool in this endeavor, offering rapid, low cost and detailed analysis of immune cell populations and their functional states. Indeed, this technology facilitates the understanding of immune dysfunctions in sepsis and contributes to the identification of novel biomarkers. Our review underscores the potential of integrating flow cytometry with omics data, machine learning and clinical observations to refine sepsis management, highlighting the shift towards personalized medicine in critical care. This approach could lead to more precise interventions, improving outcomes in this heterogeneously affected patient population.
Collapse
Affiliation(s)
- Elena Santacroce
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Miriam D’Angerio
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Alin Liviu Ciobanu
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Linda Masini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Irene Coloretti
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Stefano Busani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Ignacio Rubio
- Department of Anesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany;
| | - Marianna Meschiari
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Erica Franceschini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Cristina Mussini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Massimo Girardis
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| |
Collapse
|
3
|
Marques A, Torre C, Pinto R, Sepodes B, Rocha J. Treatment Advances in Sepsis and Septic Shock: Modulating Pro- and Anti-Inflammatory Mechanisms. J Clin Med 2023; 12:2892. [PMID: 37109229 PMCID: PMC10142733 DOI: 10.3390/jcm12082892] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/10/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Sepsis is currently defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection, and it affects over 25 million people every year. Even more severe, septic shock is a subset of sepsis defined by persistent hypotension, and hospital mortality rates are higher than 40%. Although early sepsis mortality has greatly improved in the past few years, sepsis patients who survive the hyperinflammation and subsequent organ damage often die from long-term complications, such as secondary infection, and despite decades of clinical trials targeting this stage of the disease, currently, no sepsis-specific therapies exist. As new pathophysiological mechanisms have been uncovered, immunostimulatory therapy has emerged as a promising path forward. Highly investigated treatment strategies include cytokines and growth factors, immune checkpoint inhibitors, and even cellular therapies. There is much to be learned from related illnesses, and immunotherapy trials in oncology, as well as the recent COVID-19 pandemic, have greatly informed sepsis research. Although the journey ahead is a long one, the stratification of patients according to their immune status and the employment of combination therapies represent a hopeful way forward.
Collapse
Affiliation(s)
- Adriana Marques
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.M.); (C.T.); (R.P.); (B.S.)
| | - Carla Torre
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.M.); (C.T.); (R.P.); (B.S.)
| | - Rui Pinto
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.M.); (C.T.); (R.P.); (B.S.)
- Joaquim Chaves Saúde, Joaquim Chaves Laboratório de Análises Clínicas, Miraflores, 1495-069 Algés, Portugal
| | - Bruno Sepodes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.M.); (C.T.); (R.P.); (B.S.)
| | - João Rocha
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.M.); (C.T.); (R.P.); (B.S.)
| |
Collapse
|
4
|
Amanati A, Shahriari M, Bordbar MR, Hedayati SB, Ziyaeyan M, Jamalidoust M, Kalani M, Heydari Marandi N. Severe acute respiratory syndrome coronavirus-2 Alpha variant (B.1.1.7), original wild-type severe acute respiratory syndrome coronavirus 2, and cytomegalovirus co-infection in a young adult with acute lymphoblastic leukemia, case report, and review of the possible cytomegalovirus reactivation mechanisms. J Med Case Rep 2023; 17:66. [PMID: 36765433 PMCID: PMC9913040 DOI: 10.1186/s13256-022-03750-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 12/29/2022] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND Like other viral infections, severe acute respiratory syndrome coronavirus-2 infection could affect different human body systems, including host immune responses. Three years after its pandemic, we learn more about this novel coronavirus. As we expected, different co-infections with various organisms, such as viruses, bacteria, and even fungi, have been reported. However, concurrent infection with two severe acute respiratory syndrome coronavirus-2 strains and cytomegalovirus is extremely unusual. We have only a rudimentary understanding of such co-infections and their long-term consequences for patients with cancer. CASE PRESENTATION An 18-year-old young Iranian adult with acute lymphoblastic leukemia presented with abdominal pain, diarrhea, nausea, and vomiting following a recent history of severe acute respiratory syndrome coronavirus-2 infection. The patient never experienced respiratory symptoms, and the chest imaging study was normal on admission. His primary laboratory investigation revealed prerenal azotemia and severe abnormal liver function tests (blood urea nitrogen 32 mg/dL, creatinine 1.75 mg/dL, prothrombin time 66 s, partial thromboplastin time 44.5 s, international normalized ratio 5.14, total bilirubin 2.9 mg/dL, and direct bilirubin 2.59 mg/dL). Cytomegalovirus disease was diagnosed by polymerase chain reaction in his blood and stool samples. The patient's gastrointestinal signs and symptoms improved shortly after receiving intravenous ganciclovir treatment. His gastrointestinal symptoms continued intermittently for weeks despite maintenance valganciclovir prescription, necessitating frequent hospitalizations. The patient was complicated by the recurrence of gastrointestinal symptoms during the sixth hospitalization, even though he had no respiratory symptoms, and the nasopharyngeal test revealed severe acute respiratory syndrome coronavirus-2 Wuhan strain for the first time. Remdesivir and valganciclovir were administrated due to persistent enteritis and evidence of intestinal tissue invasion by severe acute respiratory syndrome coronavirus 2 and cytomegalovirus on multiple intestinal biopsies, which led to partial clinical responses. Cytomegalovirus and severe acute respiratory syndrome coronavirus-2 fecal shedding continued for more than 6 months despite repeated antiviral therapy, and the Wuhan and Alpha strains were also detected in his nasopharyngeal samples through repeated sampling (confirmed by four nasopharyngeal sampling and multiple stool specimens and several intestinal biopsies). Finally, during the Delta-variant (B.1.617.2) outbreak in Iran, the patient was admitted again with febrile neutropenia and decreased level of consciousness, necessitating respiratory support and mechanical ventilation. During the Delta-variant peak, the patient's nasopharyngeal sample once more tested positive for severe acute respiratory syndrome coronavirus 2. The patient died a few days later from cardiopulmonary arrest. CONCLUSION The coronavirus disease 2019 pandemic has encountered patients with cancer with critical diagnostic and treatment challenges. Patients who are immunocompromised may co-infect with multiple severe acute respiratory syndrome coronavirus-2 strains and cytomegalovirus, and even with timely diagnosis and treatment, the prognosis may be poor.
Collapse
Affiliation(s)
- Ali Amanati
- Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Departments of Pediatrics, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mahdi Shahriari
- The Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Mazyar Ziyaeyan
- Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Jamalidoust
- Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Kalani
- Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nahid Heydari Marandi
- Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
5
|
Pei F, Yao RQ, Ren C, Bahrami S, Billiar TR, Chaudry IH, Chen DC, Chen XL, Cui N, Fang XM, Kang Y, Li WQ, Li WX, Liang HP, Lin HY, Liu KX, Lu B, Lu ZQ, Maegele M, Peng TQ, Shang Y, Su L, Sun BW, Wang CS, Wang J, Wang JH, Wang P, Xie JF, Xie LX, Zhang LN, Zingarelli B, Guan XD, Wu JF, Yao YM. Expert consensus on the monitoring and treatment of sepsis-induced immunosuppression. Mil Med Res 2022; 9:74. [PMID: 36567402 PMCID: PMC9790819 DOI: 10.1186/s40779-022-00430-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/14/2022] [Indexed: 12/27/2022] Open
Abstract
Emerged evidence has indicated that immunosuppression is involved in the occurrence and development of sepsis. To provide clinical practice recommendations on the immune function in sepsis, an expert consensus focusing on the monitoring and treatment of sepsis-induced immunosuppression was developed. Literature related to the immune monitoring and treatment of sepsis were retrieved from PubMed, Web of Science, and Chinese National Knowledge Infrastructure to design items and expert opinions were collected through an online questionnaire. Then, the Delphi method was used to form consensus opinions, and RAND appropriateness method was developed to provide consistency evaluation and recommendation levels for consensus opinions. This consensus achieved satisfactory results through two rounds of questionnaire survey, with 2 statements rated as perfect consistency, 13 as very good consistency, and 9 as good consistency. After summarizing the results, a total of 14 strong recommended opinions, 8 weak recommended opinions and 2 non-recommended opinions were produced. Finally, a face-to-face discussion of the consensus opinions was performed through an online meeting, and all judges unanimously agreed on the content of this consensus. In summary, this expert consensus provides a preliminary guidance for the monitoring and treatment of immunosuppression in patients with sepsis.
Collapse
Affiliation(s)
- Fei Pei
- Department of Critical Care Medicine, the First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Er Road, Yuexiu District, Guangzhou, 510080, Guangdong, China
| | - Ren-Qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Chao Ren
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Soheyl Bahrami
- Ludwig-Boltzmann Institute for Experimental and Clinical Traumatology, 1200, Vienna, Austria
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Irshad H Chaudry
- Center for Surgical Research and Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - De-Chang Chen
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, 200025, China
| | - Xu-Lin Chen
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Na Cui
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Xiang-Ming Fang
- Department of Anesthesiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 31003, China
| | - Yan Kang
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Wei-Qin Li
- Department of Critical Care Medicine, General Hospital of Eastern Theater Command of Chinese PLA, Nanjing, 210002, China
| | - Wen-Xiong Li
- Department of Surgical Intensive Critical Unit, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Hua-Ping Liang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Hong-Yuan Lin
- Department of Critical Care Medicine, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, 100048, China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ben Lu
- Department of Critical Care Medicine and Hematology, the Third Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Zhong-Qiu Lu
- Emergency Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Marc Maegele
- Department of Traumatology and Orthopedic Surgery, University Witten-Herdecke, 51109, Cologne, Germany
| | - Tian-Qing Peng
- Critical Illness Research, Lawson Health Research Institute, London Health Sciences Centre, London, ON, N6A 4G4, Canada
| | - You Shang
- Department of Critical Care Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lei Su
- Department of Intensive Care Unit, General Hospital of Southern Theater Command of Chinese PLA, Guangzhou, 510030, China
| | - Bing-Wei Sun
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China
| | - Chang-Song Wang
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Jian Wang
- Children's Hospital of Soochow University, Pediatric Research Institute of Soochow University, Suzhou, 215123, China
| | - Jiang-Huai Wang
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork, T12 E8YV, Ireland
| | - Ping Wang
- Center for Immunology and Inflammation, the Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Jian-Feng Xie
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Li-Xin Xie
- Department of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, 100853, China
| | - Li-Na Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Basilia Zingarelli
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 41073, USA
| | - Xiang-Dong Guan
- Department of Critical Care Medicine, the First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Er Road, Yuexiu District, Guangzhou, 510080, Guangdong, China.
| | - Jian-Feng Wu
- Department of Critical Care Medicine, the First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Er Road, Yuexiu District, Guangzhou, 510080, Guangdong, China. .,Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China.
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | | | | | | |
Collapse
|
6
|
Electroacupuncture at Zusanli (ST36), Guanyuan (CV4), and Qihai (CV6) Acupoints Regulates Immune Function in Patients with Sepsis via the PD-1 Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7037497. [PMID: 35860804 PMCID: PMC9293513 DOI: 10.1155/2022/7037497] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 06/12/2022] [Accepted: 06/23/2022] [Indexed: 11/17/2022]
Abstract
Objective The present study is aimed at investigating the biochemical and clinical effects of electroacupuncture in patients with sepsis. Methods Patients with sepsis treated at Guangdong Provincial Hospital of Chinese Medicine from July 2019 to December 2020 were included. Patients were randomly assigned to treatment with routine Western medicine (WM group) or treatment with Western medicine plus electroacupuncture based on Western medicine (EA group). Indices associated with immune function and clinical efficacy were determined before and at 3 and 5 days after treatment. Indicators of immune function included the percentage of T lymphocyte subsets, natural killer (NK) cells, and soluble programmed death protein 1 (sPD-1) levels. Indicators of clinical efficacy included infection-related indicators in whole blood; levels of tumor necrosis factor-α (TNF-α), C-reactive protein (CRP), and interferon-γ (INF-γ); and assessments using acute physiology and chronic health evaluation-II (APACHE-II) and sequential organ failure assessment (SOFA) scores. Results Baseline data were not different between WM (N = 30) and EA groups (N = 30). At day 5 following treatment, the level of sPD-1 in the EA group was lower than that in the WM group. Proportions of CD3 + T lymphocytes, CD4 + T lymphocytes, and NK cells, the percentage of lymphocytes, and INF-γ levels in the EA group were significantly higher than those in the WM group. Compared with the WM group, the white blood cell count (WBC), percentage and count of neutrophils, ratio of neutrophils to lymphocytes, and levels of CRP and TNF-α were significantly decreased in the EA group 5 days after treatment. The APACHE-II score of the EA group was significantly lower than that of the WM group 5 days after treatment. Conclusion Electroacupuncture may regulate the immune function of patients with sepsis through the PD-1 pathway to achieve an anti-inflammatory state and improve clinical symptoms.
Collapse
|
7
|
Ong DSY, Chong GLM, Chemaly RF, Cremer OL. Comparative clinical manifestations and immune effects of cytomegalovirus infections following distinct types of immunosuppression. Clin Microbiol Infect 2022; 28:1335-1344. [PMID: 35709902 DOI: 10.1016/j.cmi.2022.05.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cytomegalovirus (CMV) infection is a well-recognized complication of solid organ and hematopoietic cell transplantation. However, CMV infection also occurs in patients with human immunodeficiency virus (HIV) infection, previously immunocompetent intensive care unit (ICU) patients, and individuals on immunosuppressive medications for various underlying diseases. OBJECTIVES This review describes the comparative effects of CMV infection in distinct types of acquired immunosuppression. SOURCES Selected peer-reviewed publications on CMV infections published until December 2021. CONTENT CMV infection affects various organ systems through direct cytolytic mechanisms, but may also exert indirect effects by promoting pro-inflammatory and immunosuppressive responses. This has been well studied in transplant recipients, for whom antiviral prophylaxis and pre-emptive therapy have now become standard practice. These strategies not only prevent direct CMV disease manifestations, but also mitigate various immunopathological processes to reduce graft-versus-host disease, graft rejection, and the occurrence of secondary bacterial and fungal infections. The efficacy of neither prophylactic nor pre-emptive treatment of CMV infection has been demonstrated for patients with critical illness- or medication-induced immunosuppression. Many observational studies have shown an independent association between CMV reactivation and a prolonged duration of mechanical ventilation or increased mortality in the ICU. Furthermore, data suggest that CMV reactivation may increase pulmonary inflammation and prolong the duration of mechanical ventilation. IMPLICATIONS A large number of observational and experimental studies suggest attributable morbidity and mortality related to CMV infection, not only in transplant recipients and patients with HIV infection but also in patients with critically illness- or medication-induced immunosuppression. Adequately powered randomized controlled trials investigating the efficacy of prophylaxis or pre-emptive treatment of CMV infection in these patients are lacking, with a notable exception for transplant recipients.
Collapse
Affiliation(s)
- David S Y Ong
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands; Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| | - Ga-Lai M Chong
- Erasmus University Medical Center, Department of Medical Microbiology & Infectious Diseases, Rotterdam, the Netherlands
| | - Roy F Chemaly
- Department of Infectious Diseases, Infection Control, & Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Olaf L Cremer
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
8
|
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: 24] [Impact Index Per Article: 12.0] [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
|
9
|
Taherifard E, Movahed H, Kiani Salmi S, Taherifard A, Abdollahifard S, Taherifard E. Cytomegalovirus coinfection in patients with severe acute respiratory syndrome coronavirus 2 infection: a systematic review of reported cases. Infect Dis (Lond) 2022; 54:543-557. [PMID: 35522073 DOI: 10.1080/23744235.2022.2070273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Dysfunction of both the innate and the adaptive immune systems is observed in severe coronavirus disease 2019 which, together with administration of immunosuppressive drugs, could lead to cytomegalovirus coinfection or reactivation associated with a poorer outcome. The current study aimed to systematically review the pattern, presentations, clinical course and outcome of patients with severe acute respiratory syndrome coronavirus 2 and cytomegalovirus coinfection. METHODS Three online databases, PubMed, Scopus and Web of Science, were searched, and after excluding duplicates and irrelevant reports, eligible articles were identified. Information about patients' age and gender, comorbidities, presentations of coronavirus disease 2019 and cytomegalovirus, treatment courses and outcomes were extracted. RESULTS A total of 34 reports with 59 patients with coinfection were considered to be eligible for data extraction. A majority of patients were middle-aged or elderly (84.5%). More than three-fourths (79.2%) had at least one comorbidity. Cytomegalovirus viremia was documented in 43 patients. The most common end organ involved was the gastrointestinal tract in 13 patients (48.1% of 27 patients with end organ involvement), mostly as cytomegalovirus colitis, followed by the respiratory tract in 12 patients. There was a significant association between intubation and fatal outcome (p = .011). CONCLUSION We comprehensively reviewed published cases with coronavirus disease 2019 and cytomegalovirus reactivation. The findings may assist in appraising signs and symptoms for early suspicion, detection and treatment in patients with unusual clinical courses or with severe, prolonged or unexplained deterioration of end organ function.
Collapse
Affiliation(s)
| | - Hamed Movahed
- Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Sima Kiani Salmi
- Radiology Department, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Ali Taherifard
- Radiology Department, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Saeed Abdollahifard
- Research Center for Neuromodulation and Pain, Shiraz, Fars, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | | |
Collapse
|
10
|
Lan J, Zhang H, Zhao H, Liu L, Shi Q, Li D, Ju X. Cord Blood Natural Killer Cells Inhibit Sepsis Caused by Feces-Induced Acute Peritonitis via Increasing Endothelium Integrity. Cell Transplant 2022; 31:9636897221090257. [PMID: 35438589 PMCID: PMC9021520 DOI: 10.1177/09636897221090257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Sepsis is associated with acute peritonitis, which can be induced by lipopolysaccharide exposure and feces. Generally, lipopolysaccharide induces mono-microbial peritonitis, whereas feces cause poly-microbial peritonitis; the latter is a more complicated and closer to the clinical diseases. Although several reports have discussed the mechanism of immune response in peritonitis-induced sepsis, however, the role of natural killer (NK) cells in sepsis, especially the relationship between NK cells and stabilization of the vascular endothelial barrier, is still unclear. Accordingly, in this study, we assessed the roles of NK cells in an acute sepsis model in mice. NK cells were injected via the tail vein into mice with acute sepsis, and nitric oxide (NO), anti-inflammatory cytokine, and angiogenic factors were tested to explore the effects of NK cells on sepsis. The survival rate of septic model mice infused with NK cells was significantly improved compared with the control group. Interestingly, the levels of NO, interleukin-10, and vascular endothelial growth factor (VEGF) decreased in NK cells therapy group. After the injection of NK cells, CD31 positive endothelial cells significantly increased in the kidneys and liver, although the expression of VEGF, ANGPT-1, and ET-1 was downregulated. Consistent with our hypothesis, the transfusion of NK cells into mice with sepsis blocked inflammation and increased endothelium integrity. Overall, these findings suggest that NK cells may block sepsis by modulating the VEGF pathway.
Collapse
Affiliation(s)
- Jing Lan
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hong Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Hui Zhao
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Linghong Liu
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China.,Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan, China
| | - Qing Shi
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China.,Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan, China
| | - Dong Li
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China.,Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan, China
| | - Xiuli Ju
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China.,Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
11
|
Abstract
Reactivation of herpsviruses, mainly HSV, CMV and EBV, are frequent among critically ill patients. Although they are not immunocompromised from a classical point of view, these patients often present an alteration of their immune system favoring viral reactivation. Seropositive patients with sepsis and under mechanical ventilation are particularly at risk. Herpesviruses have a pulmonary tropism and can be responsible for non-resolving forms of acute respiratory distress syndrome with high mortality. However, the direct causality between herpesviruses reactivation and impaired outcomes among severely ill patients remains under debate.
Collapse
|
12
|
Nedeva C. Inflammation and Cell Death of the Innate and Adaptive Immune System during Sepsis. Biomolecules 2021; 11:1011. [PMID: 34356636 PMCID: PMC8301842 DOI: 10.3390/biom11071011] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/05/2021] [Accepted: 07/09/2021] [Indexed: 12/23/2022] Open
Abstract
Sepsis is a life-threatening medical condition that occurs when the host has an uncontrolled or abnormal immune response to overwhelming infection. It is now widely accepted that sepsis occurs in two concurrent phases, which consist of an initial immune activation phase followed by a chronic immunosuppressive phase, leading to immune cell death. Depending on the severity of the disease and the pathogen involved, the hosts immune system may not fully recover, leading to ongoing complications proceeding the initial infection. As such, sepsis remains one of the leading causes of morbidity and mortality world-wide, with treatment options limited to general treatment in intensive care units (ICU). Lack of specific treatments available for sepsis is mostly due to our limited knowledge of the immuno-physiology associated with the disease. This review will provide a comprehensive overview of the mechanisms and cell types involved in eliciting infection-induced immune activation from both the innate and adaptive immune system during sepsis. In addition, the mechanisms leading to immune cell death following hyperactivation of immune cells will be explored. The evaluation and better understanding of the cellular and systemic responses leading to disease onset could eventuate into the development of much needed therapies to combat this unrelenting disease.
Collapse
Affiliation(s)
- Christina Nedeva
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
| |
Collapse
|
13
|
Downregulation of miR-497-5p Improves Sepsis-Induced Acute Lung Injury by Targeting IL2RB. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6624702. [PMID: 33954185 PMCID: PMC8057895 DOI: 10.1155/2021/6624702] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/29/2021] [Accepted: 03/13/2021] [Indexed: 01/04/2023]
Abstract
Introduction Acute lung injury (ALI) induced by sepsis is a process related to inflammatory reactions, which involves lung cell apoptosis and production of inflammatory cytokine. Here, lipopolysaccharide (LPS) was applied to stimulate the mouse or human normal lung epithelial cell line (BEAS-2B) to construct a sepsis model in vivo and in vitro, and we also investigated the effect of miR-497-5p on sepsis-induced ALI. Material and Methods. Before LPS treatment, miR-497-5p antagomir was injected intravenously into mice to inhibit miR-497-5p expression in vivo. Similarly, miR-497-5p was knocked down in BEAS-2B cells. Luciferase reporter assay was applied to predict and confirm the miR-497-5p target gene. Cell viability, apoptosis, the levels of miR-497-5p, IL2RB, SP1, inflammatory cytokine, and lung injury were assessed. Results In BEAS-2B cells, a significant increase of apoptosis and inflammatory cytokine was shown after LPS stimulation. In septic mice, increased inflammatory cytokine production and apoptosis in lung cells and pulmonary morphological abnormalities were shown. The miR-497-5p inhibitor transfection showed antiapoptotic and anti-inflammatory effects on BEAS-2B cells upon LPS stimulation. In septic mice, the miR-497-5p antagomir injection also alleviated ALI, apoptosis, and inflammation caused by sepsis. The downregulation of IL2RB in BEAS-2B cells reversed the protective effects of the miR-497-5p inhibitor against ALI. Conclusion In conclusion, downregulation of miR-497-5p reduced ALI caused by sepsis through targeting IL2RB, indicating the potential effect of miR-497-5p for improving ALI caused by sepsis.
Collapse
|
14
|
Imlay H, Dasgupta S, Boeckh M, Stapleton RD, Rubenfeld GD, Chen Y, Limaye AP. Risk Factors for Cytomegalovirus Reactivation and Association With Outcomes in Critically Ill Adults With Sepsis: A Pooled Analysis of Prospective Studies. J Infect Dis 2020; 223:2108-2112. [PMID: 33159200 DOI: 10.1093/infdis/jiaa697] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 11/03/2020] [Indexed: 12/26/2022] Open
Abstract
We performed multivariable analysis of potential risk factors (including cytomegalovirus [CMV] reactivation) for clinical outcomes by day 28 (death or continued hospitalization, ventilator-free days, intensive care unit (ICU)-free days, hospital-free days) from pooled cohorts of 2 previous prospective studies of CMV-seropositive adults with sepsis. CMV reactivation at any level, >100 IU/mL, >1000 IU/mL, peak viral load, and area under the curve were independently associated with the clinical outcomes. We identified the potential effect size of CMV on outcomes that could be used as end points for future interventional trials of CMV prevention using antiviral prophylaxis in ICU patients with sepsis.
Collapse
Affiliation(s)
- Hannah Imlay
- Department of Internal Medicine, University of Washington, Seattle, Washington, USA.,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Sayan Dasgupta
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Michael Boeckh
- Department of Internal Medicine, University of Washington, Seattle, Washington, USA.,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Renee D Stapleton
- Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | | | - Ying Chen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Ajit P Limaye
- Department of Internal Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
15
|
Identification of Potential Biomarkers and Immune Features of Sepsis Using Bioinformatics Analysis. Mediators Inflamm 2020; 2020:3432587. [PMID: 33132754 PMCID: PMC7568774 DOI: 10.1155/2020/3432587] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/26/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
Sepsis remains a major global concern and is associated with high mortality and morbidity despite improvements in its management. Markers currently in use have shortcomings such as a lack of specificity and failures in the early detection of sepsis. In this study, we aimed to identify key genes involved in the molecular mechanisms of sepsis and search for potential new biomarkers and treatment targets for sepsis using bioinformatics analyses. Three datasets (GSE95233, GSE57065, and GSE28750) associated with sepsis were downloaded from the public functional genomics data repository Gene Expression Omnibus. Differentially expressed genes (DEGs) were identified using R packages (Affy and limma). Functional enrichment of the DEGs was analyzed with the DAVID database. Protein-protein interaction networks were derived using the STRING database and visualized using Cytoscape software. Potential biomarker genes were analyzed using receiver operating characteristic (ROC) curves in the R package (pROC). The three datasets included 156 whole blood RNA samples from 89 sepsis patients and 67 healthy controls. Between the two groups, 568 DEGs were identified, among which 315 were upregulated and 253 were downregulated in the septic group. These genes were enriched for pathways mainly involved in the innate immune response, T-cell biology, antigen presentation, and natural killer cell function. ROC analyses identified nine genes—LRG1, ELANE, TP53, LCK, TBX21, ZAP70, CD247, ITK, and FYN—as potential new biomarkers for sepsis. Real-time PCR confirmed that the expression of seven of these genes was in accordance with the microarray results. This study revealed imbalanced immune responses at the transcriptomic level during early sepsis and identified nine genes as potential biomarkers for sepsis.
Collapse
|
16
|
Lymphocyte Immunosuppression and Dysfunction Contributing to Persistent Inflammation, Immunosuppression, and Catabolism Syndrome (PICS). Shock 2020; 55:723-741. [PMID: 33021569 DOI: 10.1097/shk.0000000000001675] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
ABSTRACT Persistent Inflammation, Immune Suppression, and Catabolism Syndrome (PICS) is a disease state affecting patients who have a prolonged recovery after the acute phase of a large inflammatory insult. Trauma and sepsis are two pathologies after which such an insult evolves. In this review, we will focus on the key clinical determinants of PICS: Immunosuppression and cellular dysfunction. Currently, relevant immunosuppressive functions have been attributed to both innate and adaptive immune cells. However, there are significant gaps in our knowledge, as for trauma and sepsis the immunosuppressive functions of these cells have mostly been described in acute phase of inflammation so far, and their clinical relevance for the development of prolonged immunosuppression is mostly unknown. It is suggested that the initial immune imbalance determines the development of PCIS. Additionally, it remains unclear what distinguishes the onset of immune dysfunction in trauma and sepsis and how this drives immunosuppression in these cells. In this review, we will discuss how regulatory T cells (Tregs), innate lymphoid cells, natural killer T cells (NKT cells), TCR-a CD4- CD8- double-negative T cells (DN T cells), and B cells can contribute to the development of post-traumatic and septic immunosuppression. Altogether, we seek to fill a gap in the understanding of the contribution of lymphocyte immunosuppression and dysfunction to the development of chronic immune disbalance. Further, we will provide an overview of promising diagnostic and therapeutic interventions, whose potential to overcome the detrimental immunosuppression after trauma and sepsis is currently being tested.
Collapse
|
17
|
Rasid O, Chevalier C, Camarasa TMN, Fitting C, Cavaillon JM, Hamon MA. H3K4me1 Supports Memory-like NK Cells Induced by Systemic Inflammation. Cell Rep 2020; 29:3933-3945.e3. [PMID: 31851924 DOI: 10.1016/j.celrep.2019.11.043] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 09/06/2019] [Accepted: 11/08/2019] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells are unique players in innate immunity and, as such, an attractive target for immunotherapy. NK cells display immune memory properties in certain models, but the long-term status of NK cells following systemic inflammation is unknown. Here we show that following LPS-induced endotoxemia in mice, NK cells acquire cell-intrinsic memory-like properties, showing increased production of IFNγ upon specific secondary stimulation. The NK cell memory response is detectable for at least 9 weeks and contributes to protection from E. coli infection upon adoptive transfer. Importantly, we reveal a mechanism essential for NK cell memory, whereby an H3K4me1-marked latent enhancer is uncovered at the ifng locus. Chemical inhibition of histone methyltransferase activity erases the enhancer and abolishes NK cell memory. Thus, NK cell memory develops after endotoxemia in a histone methylation-dependent manner, ensuring a heightened response to secondary stimulation.
Collapse
Affiliation(s)
- Orhan Rasid
- G5 Chromatine et Infection, Institut Pasteur, Paris, France; Unité Cytokines & Inflammation, Institut Pasteur, Paris, France.
| | | | - Tiphaine Marie-Noelle Camarasa
- G5 Chromatine et Infection, Institut Pasteur, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | | | | |
Collapse
|
18
|
Schildermans J, De Vlieger G. Cytomegalovirus: A Troll in the ICU? Overview of the Literature and Perspectives for the Future. Front Med (Lausanne) 2020; 7:188. [PMID: 32500076 PMCID: PMC7243473 DOI: 10.3389/fmed.2020.00188] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/20/2020] [Indexed: 12/12/2022] Open
Abstract
Cytomegalovirus (CMV) is one of the most pathogenic viruses in human. After a primary infection, CMV resides in the host for life as a latent infection. When immunity is reduced, CMV can escape the suppressive effects of the immune system and lead to viremia and antigenemia. This reactivation, first seen in transplant patients, has also been documented in non-immunocompromised CMV-seropositive critically ill patients and is associated with higher morbidity and mortality. In the latter, it is not clear whether CMV reactivation is an innocent bystander or the cause of this observed worse outcome. Two studies showed no difference in the outcome of CMV-seropositive and seronegative patients. In addition, proof-of-concept studies investigating prophylactic antiviral treatment to prevent CMV reactivation during critical illness, failed to show a beneficial effect on interleukin levels or clinical outcome. Further research is necessary to resolve the question whether CMV replication impairs the prognosis in non-immunocompromised critically ill patients. We here give a concise overview on the available data and propose strategies to further unravel this question. First, post-mortem investigation may be useful to evaluate the effect of viral replication on organ inflammation and function. Second, further research should focus on the question whether the level of viremia needs to exceed a threshold to be associated with worse outcome. Third, clinical and biochemical assessments may help to identify patients at high risk for reactivation. Fourth, preemptive treatment based upon early detection of the virus is currently under investigation. Finally, immune-stimulating biologicals may be beneficial in high-risk groups.
Collapse
Affiliation(s)
- Jolien Schildermans
- Clinical Division of Intensive Care Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Greet De Vlieger
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Cao C, Yu M, Chai Y. Pathological alteration and therapeutic implications of sepsis-induced immune cell apoptosis. Cell Death Dis 2019; 10:782. [PMID: 31611560 PMCID: PMC6791888 DOI: 10.1038/s41419-019-2015-1] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 08/28/2019] [Accepted: 09/02/2019] [Indexed: 02/07/2023]
Abstract
Sepsis is a life-threatening organ dysfunction syndrome caused by dysregulated host response to infection that leads to uncontrolled inflammatory response followed by immunosuppression. However, despite the high mortality rate, no specific treatment modality or drugs with high efficacy is available for sepsis to date. Although improved treatment strategies have increased the survival rate during the initial state of excessive inflammatory response, recent trends in sepsis show that mortality occurs at a period of continuous immunosuppressive state in which patients succumb to secondary infections within a few weeks or months due to post-sepsis “immune paralysis.” Immune cell alteration induced by uncontrolled apoptosis has been considered a major cause of significant immunosuppression. Particularly, apoptosis of lymphocytes, including innate immune cells and adaptive immune cells, is associated with a higher risk of secondary infections and poor outcomes. Multiple postmortem studies have confirmed that sepsis-induced immune cell apoptosis occurs in all age groups, including neonates, pediatric, and adult patients, and it is considered to be a primary contributing factor to the immunosuppressive pathophysiology of sepsis. Therapeutic perspectives targeting apoptosis through various strategies could improve survival in sepsis. In this review article, we will focus on describing the major apoptosis process of immune cells with respect to physiologic and molecular mechanisms. Further, advances in apoptosis-targeted treatment modalities for sepsis will also be discussed.
Collapse
Affiliation(s)
- Chao Cao
- Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Medical University, Tianjin, China.,Department of Internal Medicine, The University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Muming Yu
- Tianjin Medical University General Hospital, Tianjin, China
| | - Yanfen Chai
- Tianjin Medical University General Hospital, Tianjin, China. .,Tianjin Medical University, Tianjin, China.
| |
Collapse
|
20
|
Carvelli J, Piperoglou C, Bourenne J, Farnarier C, Banzet N, Demerlé C, Gainnier M, Vély F. Imbalance of Circulating Innate Lymphoid Cell Subpopulations in Patients With Septic Shock. Front Immunol 2019; 10:2179. [PMID: 31616411 PMCID: PMC6763762 DOI: 10.3389/fimmu.2019.02179] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 08/29/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Septic shock, a major cause of death in critical care, is the clinical translation of a cytokine storm in response to infection. It can be complicated by sepsis-induced immunosuppression, exemplified by blood lymphopenia, an excess of circulating Treg lymphocytes, and decreased HLA-DR expression on circulating monocytes. Such immunosuppression is associated with secondary infections, and higher mortality. The effect of these biological modifications on circulating innate lymphoid cells (ILCs) has been little studied. Methods: We prospectively enrolled patients with septic shock (Sepsis-3 definition) in the intensive care unit (ICU) of Timone CHU Hospital. ICU controls (trauma, cardiac arrest, neurological dysfunction) were recruited at the same time (NCT03297203). We performed immunophenotyping of adaptive lymphocytes (CD3+ T cells, CD19+ B cells, CD4+CD25+FoxP3+ Treg lymphocytes), ILCs (CD3−CD56+ NK cells and helper ILCs – ILC1, ILC2, and ILC3), and monocytes by flow cytometry on fresh blood samples collected between 24 and 72 h after admission. Results: We investigated adaptive and innate circulating lymphoid cells in the peripheral blood of 18 patients in septic shock, 15 ICU controls, and 30 healthy subjects. As expected, the peripheral blood lymphocytes of all ICU patients showed lymphopenia, which was not specific to sepsis, whereas those of the healthy volunteers did not. Circulating CD3+ T cells and CD3−CD56+ NK cells were mainly concerned. There was a tendency toward fewer Treg lymphocytes and lower HLA-DR expression on monocytes in ICU patients with sepsis. Although the ILC1 count was higher in septic patients than healthy subjects, ILC2, and ILC3 counts were lower in both ICU groups. However, ILC3s within the total ILCs were overrepresented in patients with septic shock. The depression of immune responses has been correlated with the occurrence of secondary infections. We did not find any differences in ILC distribution according to this criterion. Conclusion: All ICU patients exhibit lymphopenia, regardless of the nature (septic or sterile) of the initial medical condition. Specific distribution of circulating ILCs, with an excess of ILC1, and a lack of ILC3, may characterize septic shock during the first 3 days of the disease.
Collapse
Affiliation(s)
- Julien Carvelli
- APHM, Service de Médecine Intensive et Réanimation, Réanimation Des Urgences, Hôpital la Timone, Marseille, France.,CEReSS - Center for Studies and Research on Health Services and Quality of Life EA3279, Aix-Marseille University, Marseille, France
| | - Christelle Piperoglou
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille Immunopôle, Marseille, France.,Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Jeremy Bourenne
- APHM, Service de Médecine Intensive et Réanimation, Réanimation Des Urgences, Hôpital la Timone, Marseille, France.,CEReSS - Center for Studies and Research on Health Services and Quality of Life EA3279, Aix-Marseille University, Marseille, France
| | - Catherine Farnarier
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille Immunopôle, Marseille, France
| | - Nathalie Banzet
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille Immunopôle, Marseille, France
| | - Clemence Demerlé
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille Immunopôle, Marseille, France
| | - Marc Gainnier
- APHM, Service de Médecine Intensive et Réanimation, Réanimation Des Urgences, Hôpital la Timone, Marseille, France.,CEReSS - Center for Studies and Research on Health Services and Quality of Life EA3279, Aix-Marseille University, Marseille, France
| | - Frédéric Vély
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille Immunopôle, Marseille, France.,Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| |
Collapse
|
21
|
van de Groep K, Nierkens S, Cremer OL, Peelen LM, Klein Klouwenberg PMC, Schultz MJ, Hack CE, van der Poll T, Bonten MJM, Ong DSY. Effect of cytomegalovirus reactivation on the time course of systemic host response biomarkers in previously immunocompetent critically ill patients with sepsis: a matched cohort study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2018; 22:348. [PMID: 30563551 PMCID: PMC6299562 DOI: 10.1186/s13054-018-2261-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cytomegalovirus (CMV) reactivation in previously immunocompetent critically ill patients is associated with increased mortality, which has been hypothesized to result from virus-induced immunomodulation. Therefore, we studied the effects of CMV reactivation on the temporal course of host response biomarkers in patients with sepsis. METHODS In this matched cohort study, each sepsis patient developing CMV reactivation between day 3 and 17 (CMV+) was compared with one CMV seropositive patient without reactivation (CMVs+) and one CMV seronegative patient (CMVs-). CMV serostatus and plasma loads were determined by enzyme-linked immunoassays and real-time polymerase chain reaction, respectively. Systemic interleukin-6 (IL-6), IL-8, IL-18, interferon-gamma-induced protein-10 (IP-10), neutrophilic elastase, IL-1 receptor antagonist (RA), and IL-10 were measured at five time points by multiplex immunoassay. The effects of CMV reactivation on sequential concentrations of these biomarkers were assessed in multivariable mixed models. RESULTS Among 64 CMV+ patients, 45 could be matched to CMVs+ or CMVs- controls or both. The two baseline characteristics and host response biomarker levels at viremia onset were similar between groups. CMV+ patients had increased IP-10 on day 7 after viremia onset (symmetric percentage difference +44% versus -15% when compared with CMVs+ and +37% versus +4% when compared with CMVs-) and decreased IL-1RA (-41% versus 0% and -49% versus +10%, respectively). However, multivariable analyses did not show an independent association between CMV reactivation and time trends of IL-6, IP-10, IL-10, or IL-1RA. CONCLUSION CMV reactivation was not independently associated with changes in the temporal trends of host response biomarkers in comparison with non-reactivating patients. Therefore, these markers should not be used as surrogate clinical endpoints for interventional studies evaluating anti-CMV therapy.
Collapse
Affiliation(s)
- Kirsten van de Groep
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, P.O. Box 85500, 3508 GA, Utrecht, the Netherlands. .,Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands.
| | - Stefan Nierkens
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Olaf L Cremer
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Linda M Peelen
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, P.O. Box 85500, 3508 GA, Utrecht, the Netherlands.,Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Peter M C Klein Klouwenberg
- Division of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Marcus J Schultz
- Department of Intensive Care, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ , Amsterdam, the Netherlands
| | - C Erik Hack
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Tom van der Poll
- Center of Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands.,Division of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Marc J M Bonten
- Division of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - David S Y Ong
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, P.O. Box 85500, 3508 GA, Utrecht, the Netherlands.,Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Kleiweg 500, 3045 PM, Rotterdam, the Netherlands
| | | |
Collapse
|
22
|
Denstaedt SJ, Singer BH, Standiford TJ. Sepsis and Nosocomial Infection: Patient Characteristics, Mechanisms, and Modulation. Front Immunol 2018; 9:2446. [PMID: 30459764 PMCID: PMC6232897 DOI: 10.3389/fimmu.2018.02446] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/03/2018] [Indexed: 12/16/2022] Open
Abstract
Sepsis is a leading cause of death worldwide. After initial trials modulating the hyperinflammatory phase of sepsis failed, generations of researchers have focused on evaluating hypo-inflammatory immune phenotypes. The main goal has been to develop prognostic biomarkers and therapies to reduce organ dysfunction, nosocomial infection, and death. The depressed host defense in sepsis has been characterized by broad cellular reprogramming including lymphocyte exhaustion, apoptosis, and depressed cytokine responses. Despite major advances in this field, our understanding of the dynamics of the septic host response and the balance of inflammatory and anti-inflammatory cellular programs remains limited. This review aims to summarize the epidemiology of nosocomial infections and characteristic immune responses associated with sepsis, as well as immunostimulatory therapies currently under clinical investigation.
Collapse
Affiliation(s)
| | | | - Theodore J. Standiford
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
23
|
Rasid O, Cavaillon JM. Compartment diversity in innate immune reprogramming. Microbes Infect 2018; 20:156-165. [DOI: 10.1016/j.micinf.2017.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/11/2017] [Indexed: 02/07/2023]
|
24
|
Venet F, Monneret G. Advances in the understanding and treatment of sepsis-induced immunosuppression. Nat Rev Nephrol 2017; 14:121-137. [PMID: 29225343 DOI: 10.1038/nrneph.2017.165] [Citation(s) in RCA: 505] [Impact Index Per Article: 72.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sepsis is defined as a life-threatening organ dysfunction that is caused by a dysregulated host response to infection. Sepsis can induce acute kidney injury and multiple organ failures and represents the most common cause of death in the intensive care unit. Sepsis initiates a complex immune response that varies over time, with the concomitant occurrence of both pro-inflammatory and anti-inflammatory mechanisms. As a result, most patients with sepsis rapidly display signs of profound immunosuppression, which is associated with deleterious consequences. Scientific advances have highlighted the role of metabolic failure, epigenetic reprogramming, myeloid-derived suppressor cells, immature suppressive neutrophils and immune alterations in primary lymphoid organs (the thymus and bone marrow) in sepsis. An improved understanding of the mechanisms underlying this immunosuppression as well as of the similarities between sepsis-induced immunosuppression and immune defects in cancer or immunosenescence has led to novel therapeutic strategies aimed at stimulating immune function in patients with sepsis. Trials assessing the therapeutic benefit of IL-7, granulocyte-macrophage colony-stimulating factor (GM-CSF) and antibodies against programmed cell death protein 1 (PD1) and programmed cell death 1 ligand 1 (PDL1) for the treatment of sepsis are in progress. The reappraisal of sepsis pathophysiology has also resulted in a novel approach to the design of clinical trials evaluating sepsis treatments, based on an evaluation of the immune status and biomarker-based stratification of patients.
Collapse
Affiliation(s)
- Fabienne Venet
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Immunology Department, Flow Division, 69003 Lyon, France.,Equipe d'Accueil 7426, Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon 1, Hospices Civils de Lyon - bioMérieux, Hôpital Edouard Herriot, 69003 Lyon, France
| | - Guillaume Monneret
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Immunology Department, Flow Division, 69003 Lyon, France.,Equipe d'Accueil 7426, Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon 1, Hospices Civils de Lyon - bioMérieux, Hôpital Edouard Herriot, 69003 Lyon, France
| |
Collapse
|
25
|
Frydrych LM, Fattahi F, He K, Ward PA, Delano MJ. Diabetes and Sepsis: Risk, Recurrence, and Ruination. Front Endocrinol (Lausanne) 2017; 8:271. [PMID: 29163354 PMCID: PMC5670360 DOI: 10.3389/fendo.2017.00271] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 09/27/2017] [Indexed: 12/16/2022] Open
Abstract
Sepsis develops when an infection surpasses local tissue containment. A series of dysregulated physiological responses are generated, leading to organ dysfunction and a 10% mortality risk. When patients with sepsis demonstrate elevated serum lactates and require vasopressor therapy to maintain adequate blood pressure in the absence of hypovolemia, they are in septic shock with an in-hospital mortality rate >40%. With improvements in intensive care treatment strategies, overall sepsis mortality has diminished to ~20% at 30 days; however, mortality continues to steadily climb after recovery from the acute event. Traditionally, it was thought that the complex interplay between inflammatory and anti-inflammatory responses led to sepsis-induced organ dysfunction and mortality. However, a closer examination of those who die long after sepsis subsides reveals that many initial survivors succumb to recurrent, nosocomial, and secondary infections. The comorbidly challenged, physiologically frail diabetic individuals suffer the highest infection rates. Recent reports suggest that even after clinical "recovery" from sepsis, persistent alterations in innate and adaptive immune responses exists resulting in chronic inflammation, immune suppression, and bacterial persistence. As sepsis-associated immune defects are associated with increased mortality long-term, a potential exists for immune modulatory therapy to improve patient outcomes. We propose that diabetes causes a functional immune deficiency that directly reduces immune cell function. As a result, patients display diminished bactericidal clearance, increased infectious complications, and protracted sepsis mortality. Considering the substantial expansion of the elderly and obese population, global adoption of a Western diet and lifestyle, and multidrug resistant bacterial emergence and persistence, diabetic mortality from sepsis is predicted to rise dramatically over the next two decades. A better understanding of the underlying diabetic-induced immune cell defects that persist following sepsis are crucial to identify potential therapeutic targets to bolster innate and adaptive immune function, prevent infectious complications, and provide more durable diabetic survival.
Collapse
Affiliation(s)
- Lynn M. Frydrych
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Fatemeh Fattahi
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Katherine He
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Peter A. Ward
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Matthew J. Delano
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
26
|
Delano MJ, Ward PA. The immune system's role in sepsis progression, resolution, and long-term outcome. Immunol Rev 2017; 274:330-353. [PMID: 27782333 DOI: 10.1111/imr.12499] [Citation(s) in RCA: 477] [Impact Index Per Article: 68.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sepsis occurs when an infection exceeds local tissue containment and induces a series of dysregulated physiologic responses that result in organ dysfunction. A subset of patients with sepsis progress to septic shock, defined by profound circulatory, cellular, and metabolic abnormalities, and associated with a greater mortality. Historically, sepsis-induced organ dysfunction and lethality were attributed to the complex interplay between the initial inflammatory and later anti-inflammatory responses. With advances in intensive care medicine and goal-directed interventions, early 30-day sepsis mortality has diminished, only to steadily escalate long after "recovery" from acute events. As so many sepsis survivors succumb later to persistent, recurrent, nosocomial, and secondary infections, many investigators have turned their attention to the long-term sepsis-induced alterations in cellular immune function. Sepsis clearly alters the innate and adaptive immune responses for sustained periods of time after clinical recovery, with immune suppression, chronic inflammation, and persistence of bacterial representing such alterations. Understanding that sepsis-associated immune cell defects correlate with long-term mortality, more investigations have centered on the potential for immune modulatory therapy to improve long-term patient outcomes. These efforts are focused on more clearly defining and effectively reversing the persistent immune cell dysfunction associated with long-term sepsis mortality.
Collapse
Affiliation(s)
- Matthew J Delano
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Peter A Ward
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
27
|
Lachance P, Chen J, Featherstone R, Sligl WI. Association Between Cytomegalovirus Reactivation and Clinical Outcomes in Immunocompetent Critically Ill Patients: A Systematic Review and Meta-Analysis. Open Forum Infect Dis 2017; 4:ofx029. [PMID: 29497626 PMCID: PMC5781329 DOI: 10.1093/ofid/ofx029] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 02/07/2017] [Indexed: 01/17/2023] Open
Abstract
Background The aim of our systematic review was to investigate the association between cytomegalovirus (CMV) reactivation and outcomes in immunocompetent critically ill patients. Methods We searched electronic databases and gray literature for original studies and abstracts published between 1990 and October 2016. The review was limited to studies including critically ill immunocompetent patients. Cytomegalovirus reactivation was defined as positive polymerase chain reaction, pp65 antigenemia, or viral culture from blood or bronchoalveolar lavage. Selected patient-centered outcomes included mortality, duration of mechanical ventilation, need for renal replacement therapy (RRT), and nosocomial infections. Health resource utilization outcomes included intensive care unit and hospital lengths of stay. Results Twenty-two studies were included. In our primary analysis, CMV reactivation was associated with increased ICU mortality (odds ratio [OR], 2.55; 95% confidence interval [CI], 1.87–3.47), overall mortality (OR, 2.02; 95% CI, 1.60–2.56), duration of mechanical ventilation (mean difference 6.60 days; 95% CI, 3.09–10.12), nosocomial infections (OR, 3.20; 95% CI, 2.05–4.98), need for RRT (OR, 2.37; 95% CI, 1.31–4.31), and ICU length of stay (mean difference 8.18 days; 95% CI, 6.14–10.22). In addition, numerous sensitivity analyses were performed. Conclusions In this meta-analysis, CMV reactivation was associated with worse clinical outcomes and greater health resource utilization in critically ill patients. However, it remains unclear whether CMV reactivation plays a causal role or if it is a surrogate for more severe illness.
Collapse
Affiliation(s)
| | - Justin Chen
- Division of Infectious Diseases, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada; and
| | - Robin Featherstone
- Alberta Research Centre for Health Evidence, Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - Wendy I Sligl
- Department of Critical Care Medicine and.,Division of Infectious Diseases, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada; and
| |
Collapse
|
28
|
Hraiech S, Bordes J, Mège J, de Lamballerie X, Charrel R, Bechah Y, Pastorino B, Guervilly C, Forel J, Adda M, Rolain J, Lepidi H, Raoult D, Lehingue S, Papazian L. Cytomegalovirus reactivation enhances the virulence of Staphylococcus aureus pneumonia in a mouse model. Clin Microbiol Infect 2017; 23:38-45. [DOI: 10.1016/j.cmi.2016.09.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 09/13/2016] [Accepted: 09/26/2016] [Indexed: 10/20/2022]
|
29
|
Al-Omari A, Aljamaan F, Alhazzani W, Salih S, Arabi Y. Cytomegalovirus infection in immunocompetent critically ill adults: literature review. Ann Intensive Care 2016; 6:110. [PMID: 27813024 PMCID: PMC5095093 DOI: 10.1186/s13613-016-0207-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 10/17/2016] [Indexed: 12/21/2022] Open
Abstract
Cytomegalovirus (CMV) infection is increasingly recognized in critically ill immunocompetent patients. Some studies have demonstrated an association between CMV disease and increased mortality rates, prolonged intensive care unit and hospital length of stay, prolonged mechanical ventilation, and nosocomial infections. However, there is a considerable controversy whether such association represents a causal relationship between CMV disease and unfavorable outcomes or just a marker of the severity of the critical illness. Detection of CMV using polymerase chain reaction and CMV antigenemia is the standard diagnostic approach. CMV may have variety of clinical manifestations reflecting the involvement of different organ systems. Treatment of CMV in critical care is challenging due to diagnostic challenge and drug toxicity, and building predictive model for CMV disease in critical care setting would be promising to identify patients at risk and starting prophylactic therapy. Our objective was to broadly review the current literature on the prevalence and incidence, clinical manifestations, potential limitations of different diagnostic modalities, prognosis, and therapeutic options of CMV disease in critically ill patients.
Collapse
Affiliation(s)
- Awad Al-Omari
- Critical Care and Infection Control Department, Dr. Sulaiman Al Habib Medical Group, Riyadh, Saudi Arabia. .,AlFaisal University, Riyadh, Saudi Arabia.
| | - Fadi Aljamaan
- Intensive Care Department, King Khalid University Hospital, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | | | - Samer Salih
- Department of Internal Medicine, Dr. Sulaiman Al Habib Medical Group, Riyadh, Saudi Arabia
| | - Yaseen Arabi
- Intensive Care Department, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| |
Collapse
|
30
|
Navarro D. Expanding role of cytomegalovirus as a human pathogen. J Med Virol 2016; 88:1103-12. [DOI: 10.1002/jmv.24450] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2015] [Indexed: 12/19/2022]
Affiliation(s)
- David Navarro
- Microbiology Service, Hospital Clínico Universitario; Fundación INCLIVA; Valencia Spain
- Department of Microbiology, School of Medicine; University of Valencia; Valencia Spain
| |
Collapse
|
31
|
Delano MJ, Ward PA. Sepsis-induced immune dysfunction: can immune therapies reduce mortality? J Clin Invest 2016; 126:23-31. [PMID: 26727230 DOI: 10.1172/jci82224] [Citation(s) in RCA: 432] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Sepsis is a systemic inflammatory response induced by an infection, leading to organ dysfunction and mortality. Historically, sepsis-induced organ dysfunction and lethality were attributed to the interplay between inflammatory and antiinflammatory responses. With advances in intensive care management and goal-directed interventions, early sepsis mortality has diminished, only to surge later after "recovery" from acute events, prompting a search for sepsis-induced alterations in immune function. Sepsis is well known to alter innate and adaptive immune responses for sustained periods after clinical "recovery," with immunosuppression being a prominent example of such alterations. Recent studies have centered on immune-modulatory therapy. These efforts are focused on defining and reversing the persistent immune cell dysfunction that is associated with mortality long after the acute events of sepsis have resolved.
Collapse
|
32
|
|
33
|
Castón JJ, Cantisán S, González-Gasca F, Páez-Vega A, Abdel-Hadi H, Illescas S, Alonso G, Torre-Cisneros J. Interferon-γ production by CMV-specific CD8+ T lymphocytes provides protection against cytomegalovirus reactivation in critically ill patients. Intensive Care Med 2015; 42:46-53. [PMID: 26537489 DOI: 10.1007/s00134-015-4077-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 09/16/2015] [Indexed: 01/01/2023]
Abstract
PURPOSE To evaluate the usefulness of the secretion of interferon-γ (IFNγ) by cytomegalovirus (CMV)-specific CD8+ T cells to determine the risk of CMV reactivation in critically ill non-immunosuppressed patients. METHODS Two-center prospective cohort study including critically ill non-immunosuppressed CMV-seropositive patients admitted between December 2012 and March 2013. The incidence of CMV reactivation by polymerase chain reaction (real-time PCR) in plasma was investigated. IFNγ secretion by CMV-specific CD8+ T lymphocytes was determined at the time of admission to the intensive care unit (ICU) by means of the QuantiFERON(®)-CMV (QF-CMV) test. Cox regression analyses were performed to investigate CMV reactivation risk factors. RESULTS Fifty-three patients were included, of whom 13 (24.5%) presented CMV reactivation. Twenty-six patients (49.1%) were QF-CMV "reactive" (QF-CMV(R)). Of the 26 QF-CMV(R) patients, 11.5% (3/26) had CMV reactivation, whereas 37% (10/27) of QF-CMV "non reactive" patients (QF-CMV(NR)) presented reactivation (p = 0.03). By Cox regression, the presence of QF-CMV(R) at ICU admission (HR 0.09, 95% CI 0.02-0.44; p = 0.003) was associated with a decreased risk of CMV reactivation. The sensitivity, specificity, positive predictive value, and negative predictive value of QF-CMV were 77, 57, 37, and 88%, respectively. Eleven of the 53 patients (20.7%) died during the follow-up period. Mortality was more frequent in patients with CMV reactivation (6/13, 46.1 vs. 5/40, 12.5%; p = 0.015). CONCLUSIONS In critically ill non-immunosuppressed patients, the presence of functional CMV-specific CD8+ T lymphocyte response at intensive care unit admission provides protection against CMV reactivation.
Collapse
Affiliation(s)
- Juan José Castón
- Unit of Infecious Diseases, Department of Internal Medicine, Hospital General Universitario, Universidad de Castilla La Mancha, Ciudad Real, Spain.,Spanish Network for the Research in Infectious Diseases (REIPI RD12/0015), Madrid, Spain
| | - Sara Cantisán
- Spanish Network for the Research in Infectious Diseases (REIPI RD12/0015), Madrid, Spain. .,Clinical Unit of Infectious Diseases, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Avda, Menéndez Pidal s/n, 14004, Cordoba, Spain.
| | - Francisco González-Gasca
- Unit of Infecious Diseases, Department of Internal Medicine, Hospital General Universitario, Universidad de Castilla La Mancha, Ciudad Real, Spain
| | - Aurora Páez-Vega
- Clinical Unit of Infectious Diseases, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Avda, Menéndez Pidal s/n, 14004, Cordoba, Spain
| | | | - Soledad Illescas
- Service of Microbiology, Hospital General Universitario, Ciudad Real, Spain
| | - Gema Alonso
- Intensive Care Unit, Hospital Universitario Reina Sofía, Cordoba, Spain
| | - Julián Torre-Cisneros
- Spanish Network for the Research in Infectious Diseases (REIPI RD12/0015), Madrid, Spain.,Clinical Unit of Infectious Diseases, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Avda, Menéndez Pidal s/n, 14004, Cordoba, Spain
| |
Collapse
|
34
|
Papazian L, Hraiech S, Lehingue S, Roch A, Chiche L, Wiramus S, Forel JM. Cytomegalovirus reactivation in ICU patients. Intensive Care Med 2015; 42:28-37. [PMID: 26424680 PMCID: PMC7095171 DOI: 10.1007/s00134-015-4066-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/09/2015] [Indexed: 11/23/2022]
Abstract
Introduction Approximately 20 years have passed since we reported our results of histologically proven cytomegalovirus (CMV) pneumonia in non-immunocompromised ICU patients. Even if there are more recent reports suggesting that CMV may worsen the outcomes for ICU patients, there is no definite answer to this question: is CMV a potential pathogen for ICU patients or is it simply a bystander? Methods We will describe the pathophysiology of active CMV infection and the most recent insights concerning the epidemiological aspects of these reactivations. Major findings Cytomegalovirus can be pathogenic by a direct organ insult (such as for the lung), by decreasing host defences against other microorganisms and/or by enhancing the body’s inflammatory response (as in acute respiratory distress syndrome). The incidence of active CMV infection is dependent on the diagnostic method used. Using the most sophisticated available biological tools, the incidence can reach 15–20 % of ICU patients (20–40 % in ICU patients with positive CMV serology). In adequately powered cohorts of patients, active CMV infection appears to be associated with worse outcomes for mechanically ventilated ICU patients. Discussion There is no absolute direct proof of a negative impact of active CMV infection on the health outcomes of mechanically ventilated patients. Prospective randomized trials are lacking. Future trials should examine the potential benefits for health outcomes of using antiviral treatments. Such treatments could be prophylactic, pre-emptive or used only when there is an end-organ disease. Conclusion Cytomegalovirus infection may affect health outcomes for ICU patients. Additional prospective trials are necessary to confirm this hypothesis.
Collapse
Affiliation(s)
- Laurent Papazian
- Faculté de Médecine, Aix-Marseille Université, URMITE UMR CNRS 7278, 13005, Marseille, France. .,Réanimation des Détresses Respiratoires et des Infections Sévères, Assistance Publique-Hôpitaux de Marseille, Hôpital Nord, Chemin des Bourrely, 13015, Marseille, France.
| | - Sami Hraiech
- Faculté de Médecine, Aix-Marseille Université, URMITE UMR CNRS 7278, 13005, Marseille, France.,Réanimation des Détresses Respiratoires et des Infections Sévères, Assistance Publique-Hôpitaux de Marseille, Hôpital Nord, Chemin des Bourrely, 13015, Marseille, France
| | - Samuel Lehingue
- Faculté de Médecine, Aix-Marseille Université, URMITE UMR CNRS 7278, 13005, Marseille, France.,Réanimation des Détresses Respiratoires et des Infections Sévères, Assistance Publique-Hôpitaux de Marseille, Hôpital Nord, Chemin des Bourrely, 13015, Marseille, France
| | - Antoine Roch
- Faculté de Médecine, Aix-Marseille Université, URMITE UMR CNRS 7278, 13005, Marseille, France.,Service d'accueil des Urgences, Assistance Publique-Hôpitaux de Marseille, Nord, 13015, Marseille, France
| | - Laurent Chiche
- Département de Médecine Interne, Hôpital Européen, 13003, Marseille, France
| | - Sandrine Wiramus
- Service d'Anesthésie-Réanimation, Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, 13005, Marseille, France
| | - Jean-Marie Forel
- Faculté de Médecine, Aix-Marseille Université, URMITE UMR CNRS 7278, 13005, Marseille, France.,Réanimation des Détresses Respiratoires et des Infections Sévères, Assistance Publique-Hôpitaux de Marseille, Hôpital Nord, Chemin des Bourrely, 13015, Marseille, France
| |
Collapse
|
35
|
Roquilly A, David G, Cinotti R, Vourc'h M, Morin H, Rozec B, Retière C, Asehnoune K. Role of IL-12 in overcoming the low responsiveness of NK cells to missing self after traumatic brain injury. Clin Immunol 2015; 177:87-94. [PMID: 26387630 DOI: 10.1016/j.clim.2015.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/07/2015] [Accepted: 08/18/2015] [Indexed: 01/13/2023]
Abstract
Blood samples from 32 patients with severe Traumatic brain injury (TBI) were studied and compared with 11 cardiac surgery patients, and 29 healthy controls. A dramatic decreased expression of HLA class I molecules on monocytes was associated with increased KIR+ NK cell frequency in TBI patients. Overall, the phenotype of TBI NK cells marked by KIR and CD57 expression and lower level of NKp46 and DNAM-1 reflected a differentiated state. The NK-cell response to missing self was marked by lower degranulation and lower IFN-γ production after stimulation with HLA class I deficient cell line. In contrast, the NK-cell ADCC was not altered. IL-12 was able to restore both IFN-γ production and the cytotoxicity capacities of NK cells. This study provides the first extensive description of the phenotype and functions of NK cells in TBI patients. Further evaluation of IL-12 treatment to overcome immunosuppression-induced nosocomial infections is warranted.
Collapse
Affiliation(s)
- Antoine Roquilly
- Intensive Care Unit, Anesthesia and Critical Care Department, Hôtel Dieu - HME, University Hospital of Nantes, France
| | - Gaëlle David
- Intensive Care Unit, Anesthesia and Critical Care Department, Hôtel Dieu - HME, University Hospital of Nantes, France; Université de Nantes, Faculté de Médecine, Thérapeutiques Cliniques et Expérimentales des Infections, EA 3826 Nantes, France; Etablissement Français du Sang, Nantes, France; Equipe d'Accueil 4271, ImmunoVirologie et Polymorphisme Génétique, Université de Nantes, France
| | - Raphael Cinotti
- Intensive Care Unit, Anesthesia and Critical Care Department, Hôtel Dieu - HME, University Hospital of Nantes, France; Université de Nantes, Faculté de Médecine, Thérapeutiques Cliniques et Expérimentales des Infections, EA 3826 Nantes, France
| | - Mickaël Vourc'h
- Intensive Care Unit, Anesthesia and Critical Care Department, Hôtel Dieu - HME, University Hospital of Nantes, France; Université de Nantes, Faculté de Médecine, Thérapeutiques Cliniques et Expérimentales des Infections, EA 3826 Nantes, France
| | - Helene Morin
- Intensive Care Unit, Anesthesia and Critical Care Department, Laennec, University Hospital of Nantes,Nantes, France
| | - Bertrand Rozec
- Intensive Care Unit, Anesthesia and Critical Care Department, Laennec, University Hospital of Nantes,Nantes, France
| | - Christelle Retière
- Etablissement Français du Sang, Nantes, France; Equipe d'Accueil 4271, ImmunoVirologie et Polymorphisme Génétique, Université de Nantes, France
| | - Karim Asehnoune
- Intensive Care Unit, Anesthesia and Critical Care Department, Hôtel Dieu - HME, University Hospital of Nantes, France; Université de Nantes, Faculté de Médecine, Thérapeutiques Cliniques et Expérimentales des Infections, EA 3826 Nantes, France.
| |
Collapse
|
36
|
Hydrocortisone prevents immunosuppression by interleukin-10+ natural killer cells after trauma-hemorrhage. Crit Care Med 2015; 42:e752-61. [PMID: 25289930 DOI: 10.1097/ccm.0000000000000658] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Trauma induces a state of immunosuppression, which is responsible for the development of nosocomial infections. Hydrocortisone reduces the rate of pneumonia in patients with trauma. Because alterations of dendritic cells and natural killer cells play a central role in trauma-induced immunosuppression, we investigated whether hydrocortisone modulates the dendritic cell/natural killer cell cross talk in the context of posttraumatic pneumonia. DESIGN Experimental study. SETTINGS Research laboratory from an university hospital. SUBJECTS Bagg Albino/cJ mice (weight, 20-24 g). INTERVENTIONS First, in an a priori substudy of a multicenter, randomized, double-blind, placebo-controlled trial of hydrocortisone (200 mg/d for 7 d) in patients with severe trauma, we have measured the blood levels of five cytokines (tumor necrosis factor-α, interleukin-6, interleukin-10, interleukin-12, interleukin-17) at day 1 and day 8. In a second step, the effects of hydrocortisone on dendritic cell/natural killer cell cross talk were studied in a mouse model of posttraumatic pneumonia. Hydrocortisone (0.6 mg/mice i.p.) was administered immediately after hemorrhage. Twenty-four hours later, the mice were challenged with Staphylococcus aureus (7 × 10 colony-forming units). MEASUREMENTS AND MAIN RESULTS Using sera collected during a multicenter study in patients with trauma, we found that hydrocortisone decreased the blood level of interleukin-10, a cytokine centrally involved in the regulation of dendritic cell/natural killer cell cluster. In a mouse model of trauma-hemorrhage-induced immunosuppression, splenic natural killer cells induced an interleukin-10-dependent elimination of splenic dendritic cell. Hydrocortisone treatment reduced this suppressive function of natural killer cells and increased survival of mice with posthemorrhage pneumonia. The reduction of the interleukin-10 level in natural killer cells by hydrocortisone was partially dependent on the up-regulation of glucocorticoid-induced tumor necrosis factor receptor-ligand (TNFsf18) on dendritic cell. CONCLUSIONS These data demonstrate that trauma-induced immunosuppression is characterized by an interleukin-10-dependent elimination of dendritic cell by natural killer cells and that hydrocortisone improves outcome by limiting this immunosuppressive feedback loop.
Collapse
|
37
|
Virus nosocomiaux : mythe ou réalité ? MEDECINE INTENSIVE REANIMATION 2015. [DOI: 10.1007/s13546-014-1010-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
38
|
Treating HSV and CMV reactivations in critically ill patients who are not immunocompromised: pro. Intensive Care Med 2014; 40:1945-9. [PMID: 25361591 DOI: 10.1007/s00134-014-3445-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 08/11/2014] [Indexed: 12/22/2022]
|
39
|
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: 1669] [Impact Index Per Article: 151.7] [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
|
40
|
Bravo D, Clari MA, Aguilar G, Belda J, Giménez E, Carbonell JA, Henao L, Navarro D. Looking for biological factors to predict the risk of active cytomegalovirus infection in non-immunosuppressed critically ill patients. J Med Virol 2013; 86:827-33. [DOI: 10.1002/jmv.23838] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2013] [Indexed: 11/05/2022]
Affiliation(s)
- Dayana Bravo
- Microbiology Service; Hospital Clínico Universitario; Institute for Research INCLIVA; Valencia Spain
| | - María A. Clari
- Microbiology Service; Hospital Clínico Universitario; Institute for Research INCLIVA; Valencia Spain
| | - Gerardo Aguilar
- Intensive Surgical Care Unit; Hospital Clínico Universitario; Institute for Research INCLIVA; Valencia Spain
| | - Javier Belda
- Intensive Surgical Care Unit; Hospital Clínico Universitario; Institute for Research INCLIVA; Valencia Spain
- Department of Surgery and Anesthesiology; School of Medicine; University of Valencia; Valencia Spain
| | - Estela Giménez
- Microbiology Service; Hospital Clínico Universitario; Institute for Research INCLIVA; Valencia Spain
| | - José A. Carbonell
- Intensive Surgical Care Unit; Hospital Clínico Universitario; Institute for Research INCLIVA; Valencia Spain
| | - Liliana Henao
- Intensive Surgical Care Unit; Hospital Clínico Universitario; Institute for Research INCLIVA; Valencia Spain
| | - David Navarro
- Microbiology Service; Hospital Clínico Universitario; Institute for Research INCLIVA; Valencia Spain
- Department of Microbiology; School of Medicine; University of Valencia; Valencia Spain
| |
Collapse
|
41
|
Souza-Fonseca-Guimaraes F, Cavaillon JM, Adib-Conquy M. Bench-to-bedside review: Natural killer cells in sepsis - guilty or not guilty? CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2013; 17:235. [PMID: 23998530 PMCID: PMC4057064 DOI: 10.1186/cc12700] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Bacterial sepsis and septic shock are complex inflammatory disorders associated with a systemic inflammatory response syndrome. In the most severe cases of infection, an overzealous release of pro-inflammatory cytokines and inflammatory mediators by activated leukocytes, epithelial cells and endothelial cells, known as a 'cytokine storm', leads to deleterious effects such as organ dysfunction and even death. By the end of the 20th century, natural killer (NK) cells were for the first time identified as important players during sepsis. The role of this cell type was, however, double-edged, either 'angel' or 'devil' depending upon the bacterial infection model under study. Bacterial sensors (such as Toll-like receptors) have recently been shown to be expressed at the protein level in these cells. In addition, NK cells are important sources of interferon-γ and granulocyte-macrophage colony-stimulating factor, which are pro-inflammatory cytokines necessary to fight infection but can contribute to deleterious inflammation as well. Interestingly, an adaptative response occurs aimed to silence them, similar to the well-known phenomenon of endotoxin reprogramming.
Collapse
|
42
|
Clari MA, Aguilar G, Benet I, Belda J, Giménez E, Bravo D, Carbonell JA, Henao L, Navarro D. Evaluation of cytomegalovirus (CMV)-specific t-cell immunity for the assessment of the risk of active CMV infection in non-immunosuppressed surgical and trauma intensive care unit patients. J Med Virol 2013; 85:1802-10. [DOI: 10.1002/jmv.23621] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2013] [Indexed: 11/08/2022]
Affiliation(s)
- María A. Clari
- Microbiology Service, Hospital Clínico Universitario; Institute for Research INCLIVA; Valencia; Spain
| | - Gerardo Aguilar
- Surgical Intense Care Unit, Hospital Clínico Universitario; Institute for Research INCLIVA; Valencia; Spain
| | - Isabel Benet
- Hematology and Oncology Service, Hospital Clínico Universitario; Institute for Research INCLIVA; Valencia; Spain
| | | | - Estela Giménez
- Microbiology Service, Hospital Clínico Universitario; Institute for Research INCLIVA; Valencia; Spain
| | - Dayana Bravo
- Microbiology Service, Hospital Clínico Universitario; Institute for Research INCLIVA; Valencia; Spain
| | - José A. Carbonell
- Surgical Intense Care Unit, Hospital Clínico Universitario; Institute for Research INCLIVA; Valencia; Spain
| | - Liliana Henao
- Surgical Intense Care Unit, Hospital Clínico Universitario; Institute for Research INCLIVA; Valencia; Spain
| | | |
Collapse
|
43
|
Induction of B7-H6, a ligand for the natural killer cell-activating receptor NKp30, in inflammatory conditions. Blood 2013; 122:394-404. [PMID: 23687088 DOI: 10.1182/blood-2013-01-481705] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
B7-H6, a member of the B7 family of immunoreceptors, is as a cell-surface ligand for the NKp30-activating receptor expressed on natural killer cells. B7-H6 is not detected in normal human tissues at steady state but is expressed on tumor cells. However, whether B7-H6 can be expressed in other conditions remains unknown. We analyzed here the pathways that lead to the expression of B7-H6 in nontransformed cells. In vitro, B7-H6 was induced at the surface of CD14(+)CD16(+) proinflammatory monocytes and neutrophils upon stimulation by ligands of Toll-like receptors or proinflammatory cytokines such as interleukin-1β and tumor necrosis factor α. In these conditions, a soluble form of B7-H6 (sB7-H6) was also produced by activated monocytes and neutrophils. In vivo, B7-H6 was expressed on circulating proinflammatory CD14(+)CD16(+) monocytes in a group of patients in sepsis conditions, and was linked to an increased mortality. sB7-H6 was selectively detected in the sera of patients with gram-negative sepsis and was associated with membrane vesicles that co-sedimented with the exosomal fraction. These findings reveal that B7-H6 is not only implicated in tumor immunosurveillance but also participates in the inflammatory response in infectious conditions.
Collapse
|
44
|
|