1
|
Jing D, Liu J, Qin D, Lin J, Li T, Li Y, Duan M. Obeticholic acid ameliorates sepsis-induced renal mitochondrial damage by inhibiting the NF-κb signaling pathway. Ren Fail 2024; 46:2368090. [PMID: 39108162 PMCID: PMC11308967 DOI: 10.1080/0886022x.2024.2368090] [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: 02/26/2024] [Revised: 05/19/2024] [Accepted: 06/10/2024] [Indexed: 08/10/2024] Open
Abstract
Acute kidney injury (AKI), a common complication of sepsis, might be caused by overactivated inflammation, mitochondrial damage, and oxidative stress. However, the mechanisms underlying sepsis-induced AKI (SAKI) have not been fully elucidated, and there is a lack of effective therapies for AKI. To this end, this study aimed to investigate whether obeticholic acid (OCA) has a renoprotective effect on SAKI and to explore its mechanism of action. Through bioinformatics analysis, our study confirmed that the mitochondria might be a critical target for the treatment of SAKI. Thus, a septic rat model was established by cecal ligation puncture (CLP) surgery. Our results showed an evoked inflammatory response via the NF-κB signaling pathway and NLRP3 inflammasome activation in septic rats, which led to mitochondrial damage and oxidative stress. OCA, an Farnesoid X Receptor (FXR) agonist, has shown anti-inflammatory effects in numerous studies. However, the effects of OCA on SAKI remain unclear. In this study, we revealed that pretreatment with OCA can inhibit the inflammatory response by reducing the synthesis of proinflammatory factors (such as IL-1β and NLRP3) via blocking NF-κB and alleviating mitochondrial damage and oxidative stress in the septic rat model. Overall, this study provides insight into the excessive inflammation-induced SAKI caused by mitochondrial damage and evidence for the potential use of OCA in SAKI treatment.
Collapse
Affiliation(s)
- Danyang Jing
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jingfeng Liu
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Da Qin
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jin Lin
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tian Li
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yu Li
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Meili Duan
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
杨 汀, 李 育, 苏 白. [Mechanism of Extracellular Histone-Induced Endothelial Dysfunction Leading to Sepsis-Induced Acute Respiratory Distress Syndrome]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:902-910. [PMID: 39170019 PMCID: PMC11334276 DOI: 10.12182/20240760508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Indexed: 08/23/2024]
Abstract
Objective Sepsis-induced acute respiratory distress syndrome (ARDS) is an independent risk factor for mortality in critically ill septic patients. However, effective therapeutic targets are still unavailable due to the lack of understanding of its unclear pathogenesis. With increasing understanding in the roles of circulating histones and endothelial dysfunction in sepsis, we aimed to investigate the mechanism of histone-induced endothelial dysfunction leading to sepsis-induced ARDS and to provide experimental support for histone-targeted treatment of sepsis-induced ARDS. Methods First of all, in vitro experiments were conducted. Human umbilical vein endothelial cells (HUVEC) were stimulated with gradient concentrations of histones to explore for the optimal stimulation concentration in vitro. Then, HUVEC were exposed to histones at an optimal concentration with or without resatorvid (TAK-242), a selective inhibitor of Toll-like receptor 4 (TLR4), for 24 hours for modeling. The cells were divided into 4 groups: 1) the blank control group, 2) the blank control+TAK-242 intervention group, 3) the histone stimulation group, and 4) the histone+TAK-242 intervention group. HUVEC apoptosis was determined by flow cytometry, VE-Cadherin expression in endothelial cells was determined by Western blot, and the integrity of adhesion connections between endothelial cells was evaluated with confocal fluorescence microscopic images. Male C57BL/6 mice aged 6-8 weeks and weighing 22-25 g were used for the in vivo experiment. Then, the mice were given cecal ligation and puncture (CLP) as well as histone injection at 50 mg/kg via the tail vein for sepsis modeling. The experimental animals were divided into 6 groups: 1) the blank control group, 2) the blank control+TAK-242 intervention group, 3) the CLP model group, 4) the CLP+TAK-242 intervention group, 5) the histone model group, and 6) the histone+TAK-242 intervention group. After 24 h, the concentrations of serum interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were determined using ELISA kits. Western blot was performed to determine the expression of vascular endothelial (VE)-cadherin in the lung tissue. Hematoxylin and eosin (HE) staining was performed to observe the pathological changes in the lung tissue of the mice. Evans Blue was injected via the tail vein 30 min before the mice were sacrificed. Lung tissue was collected after the mice were sacrificed. Then, the concentrations of Evans blue dye per unit mass in the lung tissue from mice of different groups were evaluated, the rates of pulmonary endothelial leakage were calculated, and the integrity of the pulmonary endothelial barrier was evaluated. Results The results of the in vitro experiment showed that, compared with those of the control group, HUVEC apoptosis was significantly increased under histone stimulation (P<0.05), the expression of VE-cadherin was decreased (P<0.05), and the integrity of adherens junctions between endothelial cells was damaged. TAK-242 can significantly inhibit histone-induced HUVEC apoptosis and VE-cadherin expression reduction and maintain the integrity of adherens junctions between endothelial cells. According to the findings from the in vivo experiments, in mice with CLP-induced and histone-induced sepsis, TAK-242 effectively alleviated the increase in serum concentrations of IL-6 and TNF-α, reduced the downregulation of VE-cadherin expression in the lung tissue (P<0.05), decreased endothelial permeability of the lung vessels, and improved pathological injury in the lung tissue. Conclusion By binding to TLR-4, histone decreases VE-cadherin expression on the surface of vascular endothelial cells, disrupts the integrity of intercellular adherens junctions, and triggers pathological damage to lung tissue. Using TLR-4 inhibitors can prevent sepsis-induced ARDS in histone-induced sepsis.
Collapse
Affiliation(s)
- 汀航 杨
- 四川大学华西医院 肾脏内科/肾脏病研究所 (成都 610041)Department of Nephrology/Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 育霈 李
- 四川大学华西医院 肾脏内科/肾脏病研究所 (成都 610041)Department of Nephrology/Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 白海 苏
- 四川大学华西医院 肾脏内科/肾脏病研究所 (成都 610041)Department of Nephrology/Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Ghimire L, Paudel S, Le J, Jin L, Cai S, Bhattarai D, Jeyaseelan S. NLRP6 negatively regulates host defense against polymicrobial sepsis. Front Immunol 2024; 15:1248907. [PMID: 38720893 PMCID: PMC11078015 DOI: 10.3389/fimmu.2024.1248907] [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: 06/27/2023] [Accepted: 04/04/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction Sepsis remains a major cause of death in Intensive Care Units. Sepsis is a life-threatening multi-organ dysfunction caused by a dysregulated systemic inflammatory response. Pattern recognition receptors, such as TLRs and NLRs contribute to innate immune responses. Upon activation, some NLRs form multimeric protein complexes in the cytoplasm termed "inflammasomes" which induce gasdermin d-mediated pyroptotic cell death and the release of mature forms of IL-1β and IL-18. The NLRP6 inflammasome is documented to be both a positive and a negative regulator of host defense in distinct infectious diseases. However, the role of NLRP6 in polymicrobial sepsis remains elusive. Methods We have used NLRP6 KO mice and human septic spleen samples to examine the role of NLRP6 in host defense in sepsis. Results NLRP6 KO mice display enhanced survival, reduced bacterial burden in the organs, and reduced cytokine/chemokine production. Co-housed WT and KO mice following sepsis show decreased bacterial burden in the KO mice as observed in singly housed groups. NLRP6 is upregulated in CD3, CD4, and CD8 cells of septic patients and septic mice. The KO mice showed a higher number of CD3, CD4, and CD8 positive T cell subsets and reduced T cell death in the spleen following sepsis. Furthermore, administration of recombinant IL-18, but not IL-1β, elicited excessive inflammation and reversed the survival advantages observed in NLRP6 KO mice. Conclusion These results unveil NLRP6 as a negative regulator of host defense during sepsis and offer novel insights for the development of new treatment strategies for sepsis.
Collapse
Affiliation(s)
- Laxman Ghimire
- Laboratory of Lung Biology, Department of Pathobiological Sciences and Center for Lung Biology and Disease, School of Veterinary Medicine, Louisiana State University (LSU) and Agricultural and Mechanical College, Baton Rouge, LA, United States
| | - Sagar Paudel
- Laboratory of Lung Biology, Department of Pathobiological Sciences and Center for Lung Biology and Disease, School of Veterinary Medicine, Louisiana State University (LSU) and Agricultural and Mechanical College, Baton Rouge, LA, United States
| | - John Le
- Laboratory of Lung Biology, Department of Pathobiological Sciences and Center for Lung Biology and Disease, School of Veterinary Medicine, Louisiana State University (LSU) and Agricultural and Mechanical College, Baton Rouge, LA, United States
| | - Liliang Jin
- Laboratory of Lung Biology, Department of Pathobiological Sciences and Center for Lung Biology and Disease, School of Veterinary Medicine, Louisiana State University (LSU) and Agricultural and Mechanical College, Baton Rouge, LA, United States
| | - Shanshan Cai
- Laboratory of Lung Biology, Department of Pathobiological Sciences and Center for Lung Biology and Disease, School of Veterinary Medicine, Louisiana State University (LSU) and Agricultural and Mechanical College, Baton Rouge, LA, United States
| | - Dinesh Bhattarai
- Laboratory of Lung Biology, Department of Pathobiological Sciences and Center for Lung Biology and Disease, School of Veterinary Medicine, Louisiana State University (LSU) and Agricultural and Mechanical College, Baton Rouge, LA, United States
| | - Samithamby Jeyaseelan
- Laboratory of Lung Biology, Department of Pathobiological Sciences and Center for Lung Biology and Disease, School of Veterinary Medicine, Louisiana State University (LSU) and Agricultural and Mechanical College, Baton Rouge, LA, United States
- Section of Pulmonary and Critical Care, Department of Medicine, LSU Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
4
|
Xu H, Sheng S, Luo W, Xu X, Zhang Z. Acute respiratory distress syndrome heterogeneity and the septic ARDS subgroup. Front Immunol 2023; 14:1277161. [PMID: 38035100 PMCID: PMC10682474 DOI: 10.3389/fimmu.2023.1277161] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an acute diffuse inflammatory lung injury characterized by the damage of alveolar epithelial cells and pulmonary capillary endothelial cells. It is mainly manifested by non-cardiogenic pulmonary edema, resulting from intrapulmonary and extrapulmonary risk factors. ARDS is often accompanied by immune system disturbance, both locally in the lungs and systemically. As a common heterogeneous disease in critical care medicine, researchers are often faced with the failure of clinical trials. Latent class analysis had been used to compensate for poor outcomes and found that targeted treatment after subgrouping contribute to ARDS therapy. The subphenotype of ARDS caused by sepsis has garnered attention due to its refractory nature and detrimental consequences. Sepsis stands as the most predominant extrapulmonary cause of ARDS, accounting for approximately 32% of ARDS cases. Studies indicate that sepsis-induced ARDS tends to be more severe than ARDS caused by other factors, leading to poorer prognosis and higher mortality rate. This comprehensive review delves into the immunological mechanisms of sepsis-ARDS, the heterogeneity of ARDS and existing research on targeted treatments, aiming to providing mechanism understanding and exploring ideas for accurate treatment of ARDS or sepsis-ARDS.
Collapse
Affiliation(s)
- Huikang Xu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shiying Sheng
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Weiwei Luo
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaofang Xu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhaocai Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of the Diagnosis and Treatment for Severe Trauma and Burn of Zhejiang Province, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| |
Collapse
|
5
|
Nikolaidis I, Karakasi MV, Pilalas D, Boziki MK, Tsachouridou O, Kourelis A, Skoura L, Pavlidis P, Gargalianos-Kakoliris P, Metallidis S, Daniilidis M, Trypsiannis G, Nikolaidis P. Association of cytokine gene polymorphisms with peripheral neuropathy susceptibility in people living with HIV in Greece. J Neurovirol 2023; 29:626-639. [PMID: 37695541 DOI: 10.1007/s13365-023-01169-5] [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: 02/21/2023] [Revised: 08/02/2023] [Accepted: 08/21/2023] [Indexed: 09/12/2023]
Abstract
Relatively little research has been done in recent years to understand what leads to the unceasingly high rates of HIV sensory neuropathy despite successful antiretroviral treatment. In vivo and in vitro studies demonstrate neuronal damage induced by HIV and increasingly identified ART neurotoxicity involving mitochondrial dysfunction and innate immune system activation in peripheral nerves, ultimately all pathways resulting in enhanced pro-inflammatory cytokine secretion. Furthermore, many infectious/autoimmune/malignant diseases are influenced by the production-profile of pro-inflammatory and anti-inflammatory cytokines, due to inter-individual allelic polymorphism within cytokine gene regulatory regions. Associations of cytokine gene polymorphisms are investigated with the aim of identifying potential genetic markers for susceptibility to HIV peripheral neuropathy including ART-dependent toxic neuropathy. One hundred seventy-one people living with HIV in Northern Greece, divided into two sub-groups according to the presence/absence of peripheral neuropathy, were studied over a 5-year period. Diagnosis was based on the Brief Peripheral Neuropathy Screening. Cytokine genotyping was performed by sequence-specific primer-polymerase chain reaction. Present study findings identify age as an important risk factor (p < 0.01) and support the idea that cytokine gene polymorphisms are at least involved in HIV peripheral-neuropathy pathogenesis. Specifically, carriers of IL1a-889/rs1800587 TT genotype and IL4-1098/rs2243250 GG genotype disclosed greater relative risk for developing HIV peripheral neuropathy (OR: 2.9 and 7.7 respectively), while conversely, carriers of IL2+166/rs2069763 TT genotype yielded lower probability (OR: 3.1), all however, with marginal statistical significance. The latter, if confirmed in a larger Greek population cohort, may offer in the future novel genetic markers to identify susceptibility, while it remains significant that further ethnicity-oriented studies continue to be conducted in a similar pursuit.
Collapse
Affiliation(s)
- Ioannis Nikolaidis
- Second Department of Neurology, AHEPA University General Hospital - Department of neurosciences, Aristotle University - School of Medicine, GR 54124, Thessaloniki, Greece.
| | - Maria-Valeria Karakasi
- Third Department of Psychiatry, AHEPA University General Hospital - Department of mental health, Aristotle University - School of Medicine, GR 54124, Thessaloniki, Greece
| | - Dimitrios Pilalas
- First Department of Internal Medicine, AHEPA University General Hospital, Aristotle University - School of Medicine, GR 54124, Thessaloniki, Greece
| | - Marina-Kleopatra Boziki
- Second Department of Neurology, AHEPA University General Hospital - Department of neurosciences, Aristotle University - School of Medicine, GR 54124, Thessaloniki, Greece
| | - Olga Tsachouridou
- First Department of Internal Medicine, AHEPA University General Hospital, Aristotle University - School of Medicine, GR 54124, Thessaloniki, Greece
| | - Andreas Kourelis
- Laboratory of Immunology, Department of Microbiology, Aristotle University - School of Medicine, GR 54124, Thessaloniki, Greece
| | - Lemonia Skoura
- Laboratory of Immunology, Department of Microbiology, Aristotle University - School of Medicine, GR 54124, Thessaloniki, Greece
| | - Pavlos Pavlidis
- Laboratory of Forensic Sciences, Democritus University of Thrace - School of Medicine, GR 68100, Dragana, Alexandroupolis, Greece
| | | | - Symeon Metallidis
- First Department of Internal Medicine, AHEPA University General Hospital, Aristotle University - School of Medicine, GR 54124, Thessaloniki, Greece
| | - Michail Daniilidis
- First Department of Internal Medicine, AHEPA University General Hospital, Aristotle University - School of Medicine, GR 54124, Thessaloniki, Greece
| | - Grigorios Trypsiannis
- Laboratory of Medical Statistics, Democritus University of Thrace - School of Medicine, GR 68100, Dragana, Alexandroupolis, Greece
| | - Pavlos Nikolaidis
- First Department of Internal Medicine, AHEPA University General Hospital, Aristotle University - School of Medicine, GR 54124, Thessaloniki, Greece
| |
Collapse
|
6
|
Wang Y, Wang J, Zheng W, Zhang J, Wang J, Jin T, Tao P, Wang Y, Liu C, Huang J, Lee PY, Yu X, Zhou Q. Identification of an IL-1 receptor mutation driving autoinflammation directs IL-1-targeted drug design. Immunity 2023:S1074-7613(23)00231-5. [PMID: 37315560 DOI: 10.1016/j.immuni.2023.05.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 03/27/2023] [Accepted: 05/17/2023] [Indexed: 06/16/2023]
Abstract
The interleukin 1 (IL-1) pathway signals through IL-1 receptor type 1 (IL-1R1) and emerges as a central mediator for systemic inflammation. Aberrant IL-1 signaling leads to a range of autoinflammatory diseases. Here, we identified a de novo missense variant in IL-1R1 (p.Lys131Glu) in a patient with chronic recurrent multifocal osteomyelitis (CRMO). Patient PBMCs showed strong inflammatory signatures, particularly in monocytes and neutrophils. The p.Lys131Glu substitution affected a critical positively charged amino acid, which disrupted the binding of the antagonist ligand, IL-1Ra, but not IL-1α or IL-1β. This resulted in unopposed IL-1 signaling. Mice with a homologous mutation exhibited similar hyperinflammation and greater susceptibility to collagen antibody-induced arthritis, accompanied with pathological osteoclastogenesis. Leveraging the biology of the mutation, we designed an IL-1 therapeutic, which traps IL-1β and IL-1α, but not IL-1Ra. Collectively, this work provides molecular insights and a potential drug for improved potency and specificity in treating IL-1-driven diseases.
Collapse
Affiliation(s)
- Yusha Wang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, Zhejiang, China
| | - Jun Wang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Wenjie Zheng
- Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Jiahui Zhang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Jinbo Wang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Taijie Jin
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Panfeng Tao
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, Zhejiang, China
| | - Yibo Wang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Chenlu Liu
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Jiqian Huang
- Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Pui Y Lee
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiaomin Yu
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, Zhejiang, China; Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University, Hangzhou 311121, Zhejiang, China; Kidney Disease Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China.
| | - Qing Zhou
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, Zhejiang, China.
| |
Collapse
|
7
|
Scola L, Ferraro D, Sanfilippo GL, De Grazia S, Lio D, Giammanco GM. Age and Cytokine Gene Variants Modulate the Immunogenicity and Protective Effect of SARS-CoV-2 mRNA-Based Vaccination. Vaccines (Basel) 2023; 11:vaccines11020413. [PMID: 36851291 PMCID: PMC9962548 DOI: 10.3390/vaccines11020413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/05/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
The introduction of anti-SARS-CoV-2 vaccines in late 2020 substantially changed the pandemic picture, inducing effective protection in the population. However, individual variability was observed with different levels of cellular response and neutralizing antibodies. We report data on the impact of age, gender, and 16 single nucleotide polymorphisms (SNPs) of cytokine genes on the anti-SARS-CoV-2 IgG titers measured 31 and 105 days after administration of the second dose of BNT162b2 vaccine to 122 healthy subjects from the health care staff of the Palermo University Hospital, Italy. The higher titers at 31 days were measured in the younger subjects and in subjects bearing T-positive genotypes of IL-1R1 rs2234650 or the GG homozygous genotype of IL-6 rs1800795 SNP. T-positive genotypes are also significantly more common in subjects with higher titers at day 105. In addition, in this group of subjects, the frequency of the CT genotype of IL-4 rs2243250 is higher among those vaccinated with higher titers. Moreover, these SNPs and TNFA rs1800629 are differently distributed in a group of subjects that were found infected by SARS-CoV-2 at day 105 of evaluation. Finally, subjects that were found to be infected by SARS-CoV-2 at day 105 were significantly older than the uninfected subjects. Taken together, these data seem to suggest that age and polymorphisms of key cytokines, which regulate inflammation and humoral immune response, might influence the magnitude of the antibody response to vaccination with BNT162B2, prompting speculation about the possible benefit of a genetic background-based assessment of a personalized approach to the anti-COVID vaccination schedule.
Collapse
Affiliation(s)
- Letizia Scola
- Clinical Pathology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Corso Tukory, 211, 90134 Palermo, Italy
| | - Donatella Ferraro
- Microbiology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90133 Palermo, Italy
| | - Giuseppa Luisa Sanfilippo
- Microbiology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90133 Palermo, Italy
| | - Simona De Grazia
- Microbiology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90133 Palermo, Italy
| | - Domenico Lio
- Interdepartmental Research Center “Migrate”, University of Palermo, 90133 Palermo, Italy
- Correspondence: ; Tel.: +39-91-6555913
| | - Giovanni Maurizio Giammanco
- Microbiology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90133 Palermo, Italy
| |
Collapse
|
8
|
Biomarkers for the Prediction and Judgement of Sepsis and Sepsis Complications: A Step towards precision medicine? J Clin Med 2022; 11:jcm11195782. [PMID: 36233650 PMCID: PMC9571838 DOI: 10.3390/jcm11195782] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/19/2022] [Accepted: 09/25/2022] [Indexed: 11/16/2022] Open
Abstract
Sepsis and septic shock are a major public health concern and are still associated with high rates of morbidity and mortality. Whilst there is growing understanding of different phenotypes and endotypes of sepsis, all too often treatment strategies still only employ a “one-size-fits-all” approach. Biomarkers offer a unique opportunity to close this gap to more precise treatment approaches by providing insight into clinically hidden, yet complex, pathophysiology, or by individualizing treatment pathways. Predicting and evaluating systemic inflammation, sepsis or septic shock are essential to improve outcomes for these patients. Besides opportunities to improve patient care, employing biomarkers offers a unique opportunity to improve clinical research in patients with sepsis. The high rate of negative clinical trials in this field may partly be explained by a high degree of heterogeneity in patient cohorts and a lack of understanding of specific endotypes or phenotypes. Moving forward, biomarkers can support the selection of more homogeneous cohorts, thereby potentially improving study conditions of clinical trials. This may finally pave the way to a precision medicine approach to sepsis, septic shock and complication of sepsis in the future.
Collapse
|
9
|
Vakil MK, Mansoori Y, Al‐Awsi GRL, Hosseinipour A, Ahsant S, Ahmadi S, Ekrahi M, Montaseri Z, Pezeshki B, Mohaghegh P, Sohrabpour M, Bahmanyar M, Daraei A, Dadkhah Jouybari T, Tavassoli A, Ghasemian A. Individual genetic variability mainly of Proinflammatory cytokines, cytokine receptors, and toll-like receptors dictates pathophysiology of COVID-19 disease. J Med Virol 2022; 94:4088-4096. [PMID: 35538614 PMCID: PMC9348290 DOI: 10.1002/jmv.27849] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 11/12/2022]
Abstract
Innate and acquired immunity responses are crucial for viral infection elimination. However, genetic variations in coding genes may exacerbate the inflammation or initiate devastating cytokine storms which poses severe respiratory conditions in coronavirus disease-19 (COVID-19). Host genetic variations in particular those related to the immune responses determine the patients' susceptibility and COVID-19 severity and pathophysiology. Gene polymorphisms such as single nucleotide polymorphisms (SNPs) of interferons, TNF, IL1, IL4, IL6, IL7, IL10, and IL17 predispose patients to the severe form of COVID-19 or severe acute respiratory syndrome coronavirus-2 (SARS-COV-2). These variations mainly alter the gene expression and cause a severe response by B cells, T cells, monocytes, neutrophils, and natural killer cells participating in a cytokine storm. Moreover, cytokines and chemokines SNPs are associated with the severity of COVID-19 and clinical outcomes depending on the corresponding effect. Additionally, genetic variations in genes encoding toll-like receptors (TLRs) mainly TLR3, TLR7, and TLR9 have been related to the COVID-19 severe respiratory symptoms. The specific relation of these mutations with the novel variants of concern (VOCs) infection remains to be elucidated. Genetic variations mainly within genes encoding proinflammatory cytokines, cytokine receptors, and TLRs predispose patients to COVID-19 disease severity. Understanding host immune gene variations associated with the SARS-COV-2 infection opens insights to control the pathophysiology of emerging viral infections.
Collapse
Affiliation(s)
- Mohammad Kazem Vakil
- Noncommunicable Diseases Research CenterFasa University of Medical SciencesFasaIran
| | - Yaser Mansoori
- Noncommunicable Diseases Research CenterFasa University of Medical SciencesFasaIran
| | - Ghaidaa Raheem Lateef Al‐Awsi
- University of Al‐QadisiyahCollege of ScienceAl DiwaniyahIraq
- Department of Radiological TechniquesAl‐Mustaqbal University CollegeBabylonIraq
| | - Ali Hosseinipour
- Department of Internal MedicineFasa University of Medical SciencesFasaIran
| | - Samaneh Ahsant
- Noncommunicable Diseases Research CenterFasa University of Medical SciencesFasaIran
| | - Sedigheh Ahmadi
- Noncommunicable Diseases Research CenterFasa University of Medical SciencesFasaIran
| | - Mohammad Ekrahi
- Noncommunicable Diseases Research CenterFasa University of Medical SciencesFasaIran
| | - Zahra Montaseri
- Department of Infectious DiseasesFasa University of Medical SciencesFasaIran
| | - Babak Pezeshki
- Noncommunicable Diseases Research CenterFasa University of Medical SciencesFasaIran
| | - Poopak Mohaghegh
- Pediatrics Department, School of MedicineFasa University of Medical SciencesFasaIran
| | - Mojtaba Sohrabpour
- Noncommunicable Diseases Research CenterFasa University of Medical SciencesFasaIran
| | - Maryam Bahmanyar
- Pediatrics Department, School of MedicineFasa University of Medical SciencesFasaIran
| | - Abdolreza Daraei
- Department of Medical Genetics, School of MedicineBabol University of Medical SciencesBabolIran
| | | | | | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research CenterFasa University of Medical SciencesFasaIran
| |
Collapse
|
10
|
Martin TR, Zemans RL, Ware LB, Schmidt EP, Riches DWH, Bastarache L, Calfee CS, Desai TJ, Herold S, Hough CL, Looney MR, Matthay MA, Meyer N, Parikh SM, Stevens T, Thompson BT. New Insights into Clinical and Mechanistic Heterogeneity of the Acute Respiratory Distress Syndrome: Summary of the Aspen Lung Conference 2021. Am J Respir Cell Mol Biol 2022; 67:284-308. [PMID: 35679511 PMCID: PMC9447141 DOI: 10.1165/rcmb.2022-0089ws] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/09/2022] [Indexed: 12/15/2022] Open
Abstract
Clinical and molecular heterogeneity are common features of human disease. Understanding the basis for heterogeneity has led to major advances in therapy for many cancers and pulmonary diseases such as cystic fibrosis and asthma. Although heterogeneity of risk factors, disease severity, and outcomes in survivors are common features of the acute respiratory distress syndrome (ARDS), many challenges exist in understanding the clinical and molecular basis for disease heterogeneity and using heterogeneity to tailor therapy for individual patients. This report summarizes the proceedings of the 2021 Aspen Lung Conference, which was organized to review key issues related to understanding clinical and molecular heterogeneity in ARDS. The goals were to review new information about ARDS phenotypes, to explore multicellular and multisystem mechanisms responsible for heterogeneity, and to review how best to account for clinical and molecular heterogeneity in clinical trial design and assessment of outcomes. The report concludes with recommendations for future research to understand the clinical and basic mechanisms underlying heterogeneity in ARDS to advance the development of new treatments for this life-threatening critical illness.
Collapse
Affiliation(s)
- Thomas R. Martin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Rachel L. Zemans
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine and Program in Cellular and Molecular Biology, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Lorraine B. Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine and
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Eric P. Schmidt
- Division of Pulmonary Sciences and Critical Care, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - David W. H. Riches
- Division of Pulmonary Sciences and Critical Care, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Carolyn S. Calfee
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Anesthesia
| | - Tushar J. Desai
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Stem Cell Institute, Stanford University School of Medicine, Stanford, California
| | - Susanne Herold
- Department of Internal Medicine VI and Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Catherine L. Hough
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | | | - Michael A. Matthay
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California
| | - Nuala Meyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Samir M. Parikh
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Division of Nephrology, University of Texas Southwestern, Dallas, Texas
| | - Troy Stevens
- Department of Physiology and Cell Biology, College of Medicine, Center for Lung Biology, University of South Alabama, Mobile, Alabama; and
| | - B. Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
11
|
Brinkworth JF, Shaw JG. On race, human variation, and who gets and dies of sepsis. AMERICAN JOURNAL OF BIOLOGICAL ANTHROPOLOGY 2022. [PMCID: PMC9544695 DOI: 10.1002/ajpa.24527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Jessica F. Brinkworth
- Department of Anthropology University of Illinois Urbana‐Champaign Urbana Illinois USA
- Carl R. Woese Institute for Genomic Biology University of Illinois at Urbana‐Champaign Urbana Illinois USA
- Department of Evolution, Ecology and Behavior University of Illinois Urbana‐Champaign Urbana Illinois USA
| | - J. Grace Shaw
- Department of Anthropology University of Illinois Urbana‐Champaign Urbana Illinois USA
- Carl R. Woese Institute for Genomic Biology University of Illinois at Urbana‐Champaign Urbana Illinois USA
| |
Collapse
|
12
|
Autophagy Regulation on Pyroptosis: Mechanism and Medical Implication in Sepsis. Mediators Inflamm 2021; 2021:9925059. [PMID: 34257519 PMCID: PMC8253640 DOI: 10.1155/2021/9925059] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
Sepsis is defined as a life-threatening disease involving multiple organ dysfunction caused by dysregulated host responses to infection. To date, sepsis remains a dominant cause of death among critically ill patients. Pyroptosis is a unique form of programmed cell death mediated by the gasdermin family of proteins and causes lytic cell death and release of proinflammatory cytokines. Although there might be some positive aspects to pyroptosis, it is regarded as harmful during sepsis and needs to be restricted. Autophagy was originally characterized as a homeostasis-maintaining mechanism in living cells. In the past decade, its function in negatively modulating pyroptosis and inflammation during sepsis has attracted increased attention. Here, we present a comprehensive review of the regulatory effect of autophagy on pyroptosis during sepsis, including the latest advances in our understanding of the mechanism and signaling pathways involved, as well as the potential therapeutic application in sepsis.
Collapse
|
13
|
Homeostatic regulation of T follicular helper and antibody response to particle antigens by IL-1Ra of medullary sinus macrophage origin. Proc Natl Acad Sci U S A 2021; 118:2019798118. [PMID: 33875594 DOI: 10.1073/pnas.2019798118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hepatitis B virus (HBV) vaccines are composed of surface antigen HBsAg that spontaneously assembles into subviral particles. Factors that impede its humoral immunity in 5% to 10% of vaccinees remain elusive. Here, we showed that the low-level interleukin-1 receptor antagonist (IL-1Ra) can predict antibody protection both in mice and humans. Mechanistically, murine IL-1Ra-inhibited T follicular helper (Tfh) cell expansion and subsequent germinal center (GC)-dependent humoral immunity, resulting in significantly weakened protection against the HBV challenge. Compared to soluble antigens, HBsAg particle antigen displayed a unique capture/uptake and innate immune activation, including IL-1Ra expression, preferably of medullary sinus macrophages. In humans, a unique polymorphism in the RelA/p65 binding site of IL-1Ra enhancer associated IL-1Ra levels with ethnicity-dependent vaccination outcome. Therefore, the differential IL-1Ra response to particle antigens probably creates a suppressive milieu for Tfh/GC development, and neutralization of IL-1Ra would resurrect antibody response in HBV vaccine nonresponders.
Collapse
|
14
|
Abobaker A, Nagib T, Alsoufi A. The impact of certain genetic variants (single nucleotide polymorphisms) on incidence and severity of COVID-19. J Gene Med 2021; 23:e3310. [PMID: 33438249 PMCID: PMC7995221 DOI: 10.1002/jgm.3310] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/31/2020] [Accepted: 01/04/2021] [Indexed: 01/08/2023] Open
Affiliation(s)
| | - Taha Nagib
- Faculty of MedicineUniversity of TripoliTripoliLibya
| | - Ahmed Alsoufi
- Faculty of MedicineUniversity of TripoliTripoliLibya
| |
Collapse
|
15
|
NLRC4 gene silencing-dependent blockade of NOD-like receptor pathway inhibits inflammation, reduces proliferation and increases apoptosis of dendritic cells in mice with septic shock. Aging (Albany NY) 2021; 13:1440-1457. [PMID: 33406504 PMCID: PMC7835030 DOI: 10.18632/aging.202379] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022]
Abstract
Septic shock is one of the most significant health concerns across the world, involving hypo-perfusion and defects in tissue energy. The current study investigates the role of NLR family CARD domain containing protein 4 (NLRC4) in septic shock-induced inflammatory reactions, lung tissue injuries, and dendritic cell (DC) apoptosis. Septic shock mice models were established by modified cecal ligation and puncture and injected with retroviral vector expressing siRNA-NLRC4. DCs were then isolated and transfected with siRNA-NLRC4. The degree of lung tissue injury, cell cycle distribution, cell apoptosis and cell viability of DCs were assessed. NLRC4 was found to be expressed at high levels in mice with septic shock. NLRC4 silencing inhibited the activation of the NOD-like receptor (NLR) pathway as evidenced by the decreased levels of NOD1, NOD2, RIP2, and NF-κB. In addition, NLRC4 silencing reduced the inflammatory reaction as attributed by reduced levels of IL-1β, TNF-α and IL-6. Suppressed NLRC4 levels inhibited cell viability and promoted cell apoptosis evidenced by inhibited induction of DC surface markers (CD80, CD86, and MHC II), along with alleviated lung tissue injury. In conclusion, NLRC4 silencing ameliorates lung injury and inflammation induced by septic shock by negatively regulating the NLR pathway.
Collapse
|
16
|
Xiong S, Hong Z, Huang LS, Tsukasaki Y, Nepal S, Di A, Zhong M, Wu W, Ye Z, Gao X, Rao GN, Mehta D, Rehman J, Malik AB. IL-1β suppression of VE-cadherin transcription underlies sepsis-induced inflammatory lung injury. J Clin Invest 2020; 130:3684-3698. [PMID: 32298238 PMCID: PMC7324198 DOI: 10.1172/jci136908] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/09/2020] [Indexed: 12/18/2022] Open
Abstract
Unchecked inflammation is a hallmark of inflammatory tissue injury in diseases such as acute respiratory distress syndrome (ARDS). Yet the mechanisms of inflammatory lung injury remain largely unknown. Here we showed that bacterial endotoxin lipopolysaccharide (LPS) and cecal ligation and puncture-induced (CLP-induced) polymicrobial sepsis decreased the expression of transcription factor cAMP response element binding (CREB) in lung endothelial cells. We demonstrated that endothelial CREB was crucial for VE-cadherin transcription and the formation of the normal restrictive endothelial adherens junctions. The inflammatory cytokine IL-1β reduced cAMP generation and CREB-mediated transcription of VE-cadherin. Furthermore, endothelial cell-specific deletion of CREB induced lung vascular injury whereas ectopic expression of CREB in the endothelium prevented the injury. We also observed that rolipram, which inhibits type 4 cyclic nucleotide phosphodiesterase-mediated (PDE4-mediated) hydrolysis of cAMP, prevented endotoxemia-induced lung vascular injury since it preserved CREB-mediated VE-cadherin expression. These data demonstrate the fundamental role of the endothelial cAMP-CREB axis in promoting lung vascular integrity and suppressing inflammatory injury. Therefore, strategies aimed at enhancing endothelial CREB-mediated VE-cadherin transcription are potentially useful in preventing sepsis-induced lung vascular injury in ARDS.
Collapse
Affiliation(s)
- Shiqin Xiong
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Zhigang Hong
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Long Shuang Huang
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Yoshikazu Tsukasaki
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Saroj Nepal
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Anke Di
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Ming Zhong
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Wei Wu
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Zhiming Ye
- Department of Nephrology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaopei Gao
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Gadiparthi N. Rao
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Dolly Mehta
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Jalees Rehman
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Asrar B. Malik
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| |
Collapse
|
17
|
Al-Naseri MAS, Salman ED, Ad'hiah AH. Relapsing-remitting multiple sclerosis: A profile of interleukine-1 gene cluster polymorphisms in Iraqi patients. J Neuroimmunol 2020; 346:577291. [PMID: 32590124 DOI: 10.1016/j.jneuroim.2020.577291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/13/2022]
Abstract
In this case-control study, 68 relapsing-remitting multiple sclerosis (RRMS) patients and 133 control were genotyped for IL1A-889 (rs1800587), IL1B-511 (rs16944), IL1B+3962 (rs1143634), IL1R1pst11970 (rs2234650) and IL1RNmspa1 11,100 (rs315952) variants. Sequence-specific-primer-polymerase-chain-reaction was the genotyping method. Logistic regression analysis demonstrated protective effects of IL1B-511, IL1R1pst11970 and IL1RNmspa1 11,100 against MS incidence, while no association was found with IL1A-889 and IL1B+3962 variants. Allele frequencies showed no significant gender, medication or expanded disability status scale-associated variation. Haplotype analysis suggested 4.31-fold increased odds for MS in subjects with C-T-C-C-T haplotype of the respective loci. In conclusion, IL-1 gene variants influence MS susceptibility in Iraqi population.
Collapse
Affiliation(s)
| | - Ehab D Salman
- Biotechnology Department, College of Science, University of Baghdad, Baghdad, Iraq
| | - Ali H Ad'hiah
- Tropical-Biological Research Unit, College of Science, University of Baghdad, Baghdad, Iraq.
| |
Collapse
|
18
|
Gomez JL, Himes BE, Kaminski N. Precision Medicine in Critical Illness: Sepsis and Acute Respiratory Distress Syndrome. PRECISION IN PULMONARY, CRITICAL CARE, AND SLEEP MEDICINE 2019. [PMCID: PMC7120471 DOI: 10.1007/978-3-030-31507-8_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Sepsis and the acute respiratory distress syndrome (ARDS) each cause substantial morbidity and mortality. In contrast to other lung diseases, the entire course of disease in these syndromes is measured in days to weeks rather than months to years, which raises unique challenges in achieving precision medicine. We review advances in sepsis and ARDS resulting from omics studies, including those involving genome-wide association, gene expression, targeted proteomics, and metabolomics approaches. We focus on promising evidence of biological subtypes in both sepsis and ARDS that consistently display high risk for death. In sepsis, a gene expression signature with dysregulated adaptive immune signaling has evidence for a differential response to systemic steroid therapy, whereas in ARDS, a hyperinflammatory pattern identified in plasma using targeted proteomics responded more favorably to randomized interventions including high positive end-expiratory pressure, volume conservative fluid therapy, and simvastatin therapy. These early examples suggest heterogeneous biology that may be challenging to detect by clinical factors alone and speak to the promise of a precision approach that targets the right treatment at the right time to the right patient.
Collapse
Affiliation(s)
- Jose L. Gomez
- Assistant Professor Pulmonary, Critical Care and Sleep Medicine Section, Department of Medicine, Yale University School of Medicine, New Haven, CT USA
| | - Blanca E. Himes
- Assistant Professor of Informatics, Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA USA
| | - Naftali Kaminski
- Boehringer-Ingelheim Endowed, Professor of Internal Medicine, Chief of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT USA
| |
Collapse
|
19
|
Abstract
Sepsis is a heterogeneous disease state that is both common and consequential in critically ill patients. Unfortunately, the heterogeneity of sepsis at the individual patient level has hindered advances in the field beyond the current therapeutic standards, which consist of supportive care and antibiotics. This complexity has prompted attempts to develop a precision medicine approach, with research aimed towards stratifying patients into more homogeneous cohorts with shared biological features, potentially facilitating the identification of new therapies. Several investigators have successfully utilized leukocyte-derived mRNA and discovery-based approaches to subgroup patients on the basis of biological similarities defined by transcriptomic signatures. A critical next step is to develop a consensus sepsis subclassification system, which includes transcriptomic signatures as well as other biological and clinical data. This goal will require collaboration among various investigative groups, and validation in both existing data sets and prospective studies. Such studies are required to bring precision medicine to the bedside of critically ill patients with sepsis.
Collapse
|
20
|
Wang H, Tong Z, Li J, Xiao K, Ren F, Xie L. Genetic variants in Forkhead box O1 associated with predisposition to sepsis in a Chinese Han population. BMC Infect Dis 2019; 19:781. [PMID: 31492105 PMCID: PMC6731606 DOI: 10.1186/s12879-019-4330-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 07/29/2019] [Indexed: 12/02/2022] Open
Abstract
Background Genetic variant is one of the causes of sepsis patients’ mortality. Now, many studies have identified several SNPs related to sepsis. However, none of these studies were identified in a genome-wide way. We aimed to detect genetic polymorphisms of sepsis patients. Methods The blood samples of eight normal controls and ten sepsis patients were collected for whole exome sequencing. Then, Single Nucleotide Polymorphisms (SNPs) were selected according to quality score and number of sepsis patients who had this variants. Synonymous mutations were removed. Genes including these remaining variants were used for functional analyses. After analyses, the remaining SNPs and indels were validated in 149 normal controls and 156 sepsis patients. Finally, serum levels of proteins coded by genes including these SNPs were evaluated. Results After whole exome sequencing, 97 SNPs and one indel site were left. Then, functional screening was performed. Only seven SNPs were used for further validation. As a result, the rs2721068 in dominant model and rs17446614 in recessive model were associated with sepsis, and the ORs of these two SNPs were 3.24 (95%CI, 1.25, 8.44) and 0.47 (0.026, 0.88), respectively. These two SNPs were both located in Forkhead box O1 (FOXO1) gene. For rs2721068 (T/T, T/C-C/C) and rs17446614 (A/A-A/G, G/G), serum levels of foxo1 in sepsis patients were both significantly lower in normal controls. Conclusions We firstly reported that the rs2721068 and rs17446614 were correlated to genetic predisposition to sepsis. Electronic supplementary material The online version of this article (10.1186/s12879-019-4330-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Huijuan Wang
- Department of Respiratory Medicine, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, China.,Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
| | - Zhaohui Tong
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
| | - Jia Li
- Department of Nanlou Respiratory Medicine, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, China
| | - Kun Xiao
- Department of Respiratory Medicine, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, China
| | - Feifei Ren
- Department of Respiratory Medicine, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, China
| | - Lixin Xie
- Department of Respiratory Medicine, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
21
|
Genetic Polymorphisms in Sepsis and Cardiovascular Disease: Do Similar Risk Genes Suggest Similar Drug Targets? Chest 2019; 155:1260-1271. [PMID: 30660782 DOI: 10.1016/j.chest.2019.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/18/2018] [Accepted: 01/02/2019] [Indexed: 01/10/2023] Open
Abstract
Genetic variants are associated with altered clinical outcome of patients with sepsis and cardiovascular diseases. Common gene signaling pathways may be involved in the pathophysiology of these diseases. A better understanding of genetic commonality among these diseases may enable the discovery of important genes, signaling pathways, and therapeutic targets for these diseases. We investigated the common genetic factors by a systematic search of the literature. Twenty-four genes (ADRB2, CD14, FGB, FV, HMOX1, IL1B, IL1RN, IL6, IL10, IL17A, IRAK1, MASP2, MBL, MIR608, MIF, NOD2, PCSK9, PPARG, PROC, SERPINE1, SOD2, SVEP1, TF, TIRAP, TLR1) were extracted as reported genetic variations associated with altered outcome of both sepsis and cardiovascular diseases. Of these genes, the adverse allele (or combinations) was same in nine (ADRB2, FV, HMOX1, IL6, MBL, MIF, NOD2, PCSK9, SERPINE1), and the effect appears to be in the same direction in both sepsis and cardiovascular disease. Shared gene signaling pathways suggest that these are true biological results and could point to overlapping drug targets in sepsis and cardiovascular disease.
Collapse
|
22
|
Interleukin-1 Beta-A Friend or Foe in Malignancies? Int J Mol Sci 2018; 19:ijms19082155. [PMID: 30042333 PMCID: PMC6121377 DOI: 10.3390/ijms19082155] [Citation(s) in RCA: 299] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/14/2018] [Accepted: 07/19/2018] [Indexed: 12/19/2022] Open
Abstract
Interleukin-1 beta (IL-1β) is induced by inflammatory signals in a broad number of immune cell types. IL-1β (and IL-18) are the only cytokines which are processed by caspase-1 after inflammasome-mediated activation. This review aims to summarize current knowledge about parameters of regulation of IL-1β expression and its multi-facetted role in pathophysiological conditions. IL-1 signaling activates innate immune cells including antigen presenting cells, and drives polarization of CD4+ T cells towards T helper type (Th) 1 and Th17 cells. Therefore, IL-1β has been attributed a largely beneficial role in resolving acute inflammations, and by initiating adaptive anti-tumor responses. However, IL-1β generated in the course of chronic inflammation supports tumor development. Furthermore, IL-1β generated within the tumor microenvironment predominantly by tumor-infiltrating macrophages promotes tumor growth and metastasis via different mechanisms. These include the expression of IL-1 targets which promote neoangiogenesis and of soluble mediators in cancer-associated fibroblasts that evoke antiapoptotic signaling in tumor cells. Moreover, IL-1 promotes the propagation of myeloid-derived suppressor cells. Using genetic mouse models as well as agents for pharmacological inhibition of IL-1 signaling therapeutically applied for treatment of IL-1 associated autoimmune diseases indicate that IL-1β is a driver of tumor induction and development.
Collapse
|
23
|
Embracing Enrichment and Unknown Unknowns. Crit Care Med 2018; 46:156-158. [PMID: 29252942 DOI: 10.1097/ccm.0000000000002781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
24
|
Mortality Benefit of Recombinant Human Interleukin-1 Receptor Antagonist for Sepsis Varies by Initial Interleukin-1 Receptor Antagonist Plasma Concentration. Crit Care Med 2017; 46:21-28. [PMID: 28991823 DOI: 10.1097/ccm.0000000000002749] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Plasma interleukin-1 beta may influence sepsis mortality, yet recombinant human interleukin-1 receptor antagonist did not reduce mortality in randomized trials. We tested for heterogeneity in the treatment effect of recombinant human interleukin-1 receptor antagonist by baseline plasma interleukin-1 beta or interleukin-1 receptor antagonist concentration. DESIGN Retrospective subgroup analysis of randomized controlled trial. SETTING Multicenter North American and European clinical trial. PATIENTS Five hundred twenty-nine subjects with sepsis and hypotension or hypoperfusion, representing 59% of the original trial population. INTERVENTIONS Random assignment of placebo or recombinant human interleukin-1 receptor antagonist × 72 hours. MEASUREMENTS AND MAIN RESULTS We measured prerandomization plasma interleukin-1 beta and interleukin-1 receptor antagonist and tested for statistical interaction between recombinant human interleukin-1 receptor antagonist treatment and baseline plasma interleukin-1 receptor antagonist or interleukin-1 beta concentration on 28-day mortality. There was significant heterogeneity in the effect of recombinant human interleukin-1 receptor antagonist treatment by plasma interleukin-1 receptor antagonist concentration whether plasma interleukin-1 receptor antagonist was divided into deciles (interaction p = 0.046) or dichotomized (interaction p = 0.028). Interaction remained present across different predicted mortality levels. Among subjects with baseline plasma interleukin-1 receptor antagonist above 2,071 pg/mL (n = 283), recombinant human interleukin-1 receptor antagonist therapy reduced adjusted mortality from 45.4% to 34.3% (adjusted risk difference, -0.12; 95% CI, -0.23 to -0.01), p = 0.044. Mortality in subjects with plasma interleukin-1 receptor antagonist below 2,071 pg/mL was not reduced by recombinant human interleukin-1 receptor antagonist (adjusted risk difference, +0.07; 95% CI, -0.04 to +0.17), p = 0.230. Interaction between plasma interleukin-1 beta concentration and recombinant human interleukin-1 receptor antagonist treatment was not statistically significant. CONCLUSIONS We report a heterogeneous effect of recombinant human interleukin-1 receptor antagonist on 28-day sepsis mortality that is potentially predictable by plasma interleukin-1 receptor antagonist in one trial. A precision clinical trial of recombinant human interleukin-1 receptor antagonist targeted to septic patients with high plasma interleukin-1 receptor antagonist may be worthy of consideration.
Collapse
|
25
|
Wong HR. Intensive care medicine in 2050: precision medicine. Intensive Care Med 2017; 43:1507-1509. [PMID: 28236258 PMCID: PMC5568998 DOI: 10.1007/s00134-017-4727-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 02/12/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Hector R Wong
- Division of Critical Care Medicine, MLC 2005, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, Cincinnati Children's Research Foundation, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
26
|
Qian J, Nunez S, Kim S, Reilly MP, Foulkes AS. A score test for genetic class-level association with nonlinear biomarker trajectories. Stat Med 2017; 36:3075-3091. [PMID: 28543585 DOI: 10.1002/sim.7314] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 01/12/2017] [Accepted: 03/22/2017] [Indexed: 11/06/2022]
Abstract
Emerging data suggest that the genetic regulation of the biological response to inflammatory stress may be fundamentally different to the genetic underpinning of the homeostatic control (resting state) of the same biological measures. In this paper, we interrogate this hypothesis using a single-SNP score test and a novel class-level testing strategy to characterize protein-coding gene and regulatory element-level associations with longitudinal biomarker trajectories in response to stimulus. Using the proposed class-level association score statistic for longitudinal data, which accounts for correlations induced by linkage disequilibrium, the genetic underpinnings of evoked dynamic changes in repeatedly measured biomarkers are investigated. The proposed method is applied to data on two biomarkers arising from the Genetics of Evoked Responses to Niacin and Endotoxemia study, a National Institutes of Health-sponsored investigation of the genomics of inflammatory and metabolic responses during low-grade endotoxemia. Our results suggest that the genetic basis of evoked inflammatory response is different than the genetic contributors to resting state, and several potentially novel loci are identified. A simulation study demonstrates appropriate control of type-1 error rates, relative computational efficiency, and power. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jing Qian
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, MA, U.S.A
| | - Sara Nunez
- Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA, U.S.A
| | - Soohyun Kim
- Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA, U.S.A
| | | | - Andrea S Foulkes
- Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA, U.S.A
| |
Collapse
|
27
|
Chousterman BG, Swirski FK, Weber GF. Cytokine storm and sepsis disease pathogenesis. Semin Immunopathol 2017; 39:517-528. [PMID: 28555385 DOI: 10.1007/s00281-017-0639-8] [Citation(s) in RCA: 841] [Impact Index Per Article: 105.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 05/05/2017] [Indexed: 12/14/2022]
Abstract
Infectious diseases are a leading cause of death worldwide. Sepsis is a severe clinical syndrome related to the host response to infection. The severity of infections is due to an activation cascade that will lead to an autoamplifying cytokine production: the cytokine storm. Cytokines are a broad category of relatively small proteins (<40 kDa) that are produced and released with the aim of cell signaling. Our understanding of the processes that trigger this tremendous amount of cytokine production has made dramatic progress over the last decades, but unfortunately, these findings could not translate yet into effective treatments; so far, all clinical trials targeting cytokine production or effects failed. This review aims to summarize the pathophysiology of the cytokine storm; to describe the type, effects, and kinetics of cytokine production; and to discuss the therapeutic challenges of targeting cytokines. New promising therapeutic strategies focusing on the endothelium, as a source and a target of cytokines, are described.
Collapse
Affiliation(s)
- Benjamin G Chousterman
- Département d'Anesthésie-Réanimation, Hôpitaux Universitaires Lariboisière-Saint-Louis, AP-HP, Paris, France. .,Inserm U1160, Hôpital Saint-Louis, Paris, France.
| | - Filip K Swirski
- Center for Systems Biology, Department of Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Georg F Weber
- Department of Surgery, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
28
|
Wang L, Ko ER, Gilchrist JJ, Pittman KJ, Rautanen A, Pirinen M, Thompson JW, Dubois LG, Langley RJ, Jaslow SL, Salinas RE, Rouse DC, Moseley MA, Mwarumba S, Njuguna P, Mturi N, Williams TN, Scott JAG, Hill AVS, Woods CW, Ginsburg GS, Tsalik EL, Ko DC. Human genetic and metabolite variation reveals that methylthioadenosine is a prognostic biomarker and an inflammatory regulator in sepsis. SCIENCE ADVANCES 2017; 3:e1602096. [PMID: 28345042 PMCID: PMC5342653 DOI: 10.1126/sciadv.1602096] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 02/03/2017] [Indexed: 06/06/2023]
Abstract
Sepsis is a deleterious inflammatory response to infection with high mortality. Reliable sepsis biomarkers could improve diagnosis, prognosis, and treatment. Integration of human genetics, patient metabolite and cytokine measurements, and testing in a mouse model demonstrate that the methionine salvage pathway is a regulator of sepsis that can accurately predict prognosis in patients. Pathway-based genome-wide association analysis of nontyphoidal Salmonella bacteremia showed a strong enrichment for single-nucleotide polymorphisms near the components of the methionine salvage pathway. Measurement of the pathway's substrate, methylthioadenosine (MTA), in two cohorts of sepsis patients demonstrated increased plasma MTA in nonsurvivors. Plasma MTA was correlated with levels of inflammatory cytokines, indicating that elevated MTA marks a subset of patients with excessive inflammation. A machine-learning model combining MTA and other variables yielded approximately 80% accuracy (area under the curve) in predicting death. Furthermore, mice infected with Salmonella had prolonged survival when MTA was administered before infection, suggesting that manipulating MTA levels could regulate the severity of the inflammatory response. Our results demonstrate how combining genetic data, biomolecule measurements, and animal models can shape our understanding of disease and lead to new biomarkers for patient stratification and potential therapeutic targeting.
Collapse
Affiliation(s)
- Liuyang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Emily R. Ko
- Duke Regional Hospital, Department of Medicine, School of Medicine, Duke University, Durham, NC 27710, USA
- Duke Center for Applied Genomics & Precision Medicine, Department of Medicine, School of Medicine, Duke University, Durham, NC 27708, USA
| | - James J. Gilchrist
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, University of Oxford, Oxford OX3 7BN, U.K
- Department of Paediatrics, University of Oxford, Oxford OX3 9DU, U.K
| | - Kelly J. Pittman
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Anna Rautanen
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, University of Oxford, Oxford OX3 7BN, U.K
| | - Matti Pirinen
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, University of Oxford, Oxford OX3 7BN, U.K
| | - J. Will Thompson
- Proteomics and Metabolomics Core Facility, Duke University Medical Center, Durham, NC 27710, USA
| | - Laura G. Dubois
- Proteomics and Metabolomics Core Facility, Duke University Medical Center, Durham, NC 27710, USA
| | - Raymond J. Langley
- Department of Pharmacology and Center for Lung Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA
| | - Sarah L. Jaslow
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Raul E. Salinas
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - D. Clayburn Rouse
- Division of Laboratory Animal Resources, Duke University Medical Center, Durham, NC 27710, USA
| | - M. Arthur Moseley
- Duke Center for Applied Genomics & Precision Medicine, Department of Medicine, School of Medicine, Duke University, Durham, NC 27708, USA
- Proteomics and Metabolomics Core Facility, Duke University Medical Center, Durham, NC 27710, USA
| | - Salim Mwarumba
- Kenya Medical Research Institute–Wellcome Trust Clinical Research Programme, Kilifi 80108, Kenya
| | - Patricia Njuguna
- Kenya Medical Research Institute–Wellcome Trust Clinical Research Programme, Kilifi 80108, Kenya
| | - Neema Mturi
- Kenya Medical Research Institute–Wellcome Trust Clinical Research Programme, Kilifi 80108, Kenya
| | | | | | - Thomas N. Williams
- Kenya Medical Research Institute–Wellcome Trust Clinical Research Programme, Kilifi 80108, Kenya
- Department of Medicine, Imperial College, Norfolk Place, London W2 1PG, U.K
| | - J. Anthony G. Scott
- Kenya Medical Research Institute–Wellcome Trust Clinical Research Programme, Kilifi 80108, Kenya
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, U.K
| | - Adrian V. S. Hill
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, University of Oxford, Oxford OX3 7BN, U.K
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, U.K
| | - Christopher W. Woods
- Duke Center for Applied Genomics & Precision Medicine, Department of Medicine, School of Medicine, Duke University, Durham, NC 27708, USA
- Division of Infectious Diseases and International Health, Department of Medicine, School of Medicine, Duke University, Durham, NC 27710, USA
- Medical Service, Durham Veterans Affairs Health Care System, Durham, NC 27705, USA
| | - Geoffrey S. Ginsburg
- Duke Center for Applied Genomics & Precision Medicine, Department of Medicine, School of Medicine, Duke University, Durham, NC 27708, USA
| | - Ephraim L. Tsalik
- Duke Center for Applied Genomics & Precision Medicine, Department of Medicine, School of Medicine, Duke University, Durham, NC 27708, USA
- Division of Infectious Diseases and International Health, Department of Medicine, School of Medicine, Duke University, Durham, NC 27710, USA
- Emergency Medicine Service, Durham Veterans Affairs Health Care System, Durham, NC 27705, USA
| | - Dennis C. Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- Division of Infectious Diseases and International Health, Department of Medicine, School of Medicine, Duke University, Durham, NC 27710, USA
| |
Collapse
|
29
|
Chantratita N, Tandhavanant S, Seal S, Wikraiphat C, Wongsuvan G, Ariyaprasert P, Suntornsut P, Teerawattanasook N, Jutrakul Y, Srisurat N, Chaimanee P, Mahavanakul W, Srisamang P, Phiphitaporn S, Mokchai M, Anukunananchai J, Wongratanacheewin S, Chetchotisakd P, Emond MJ, Peacock SJ, West TE. TLR4 genetic variation is associated with inflammatory responses in Gram-positive sepsis. Clin Microbiol Infect 2017; 23:47.e1-47.e10. [PMID: 27615723 PMCID: PMC5218870 DOI: 10.1016/j.cmi.2016.08.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/06/2016] [Accepted: 08/31/2016] [Indexed: 10/21/2022]
Abstract
OBJECTIVES To identify important pathogen recognition receptor (PRR) pathways regulating innate immune responses and outcome in Staphylococcus aureus sepsis. METHODS We analysed whether candidate PRR pathway genetic variants were associated with killed S. aureus-induced cytokine responses ex vivo and performed follow-up in vitro studies. We tested the association of our top-ranked variant with cytokine responses and clinical outcomes in a prospective multicentre cohort of patients with staphylococcal sepsis. RESULTS An intronic TLR4 polymorphism and expression quantitative trait locus, rs1927907, was highly associated with cytokine release induced by stimulation of blood from healthy Thai subjects with S. aureus ex vivo. S. aureus did not induce TLR4-dependent NF-κB activation in transfected HEK293 cells. In monocytes, tumor necrosis factor (TNF)-α release induced by S. aureus was not blunted by a TLR4/MD-2 neutralizing antibody, but in a monocyte cell line, TNF-α was reduced by knockdown of TLR4. In Thai patients with staphylococcal sepsis, rs1927907 was associated with higher interleukin (IL)-6 and IL-8 levels as well as with respiratory failure. S. aureus-induced responses in blood were most highly correlated with responses to Gram-negative stimulants whole blood. CONCLUSIONS A genetic variant in TLR4 is associated with cytokine responses to S. aureus ex vivo and plasma cytokine levels and respiratory failure in staphylococcal sepsis. While S. aureus does not express lipopolysaccharide or activate TLR4 directly, the innate immune response to S. aureus does appear to be modulated by TLR4 and shares significant commonality with that induced by Gram-negative pathogens and lipopolysaccharide.
Collapse
Affiliation(s)
- N Chantratita
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - S Tandhavanant
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - S Seal
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - C Wikraiphat
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - G Wongsuvan
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - P Ariyaprasert
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - P Suntornsut
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - N Teerawattanasook
- Department of Clinical Pathology, Sunpasitthiprasong Hospital, Ubon Ratchathani, Thailand
| | - Y Jutrakul
- Department of Clinical Pathology, Udon Thani Hospital, Udon Thani, Thailand
| | - N Srisurat
- Department of Clinical Pathology, Khon Kaen Hospital, Khon Kaen, Thailand
| | - P Chaimanee
- Srinagarind Hospital, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - W Mahavanakul
- Department of Medicine, Sunpasitthiprasong Hospital, Ubon Ratchathani, Thailand
| | - P Srisamang
- Department of Pediatrics, Sunpasitthiprasong Hospital, Ubon Ratchathani, Thailand
| | - S Phiphitaporn
- Department of Medicine, Udon Thani Hospital, Udon Thani, Thailand
| | - M Mokchai
- Department of Medicine, Khon Kaen Hospital, Khon Kaen, Thailand
| | | | - S Wongratanacheewin
- Department of Microbiology and Melioidosis Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - P Chetchotisakd
- Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - M J Emond
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - S J Peacock
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - T E West
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington, Seattle, WA, USA; International Respiratory and Severe Illness Center, University of Washington, Seattle, USA
| |
Collapse
|
30
|
Reilly JP, Meyer NJ, Christie JD. Genetics in the Prevention and Treatment of Sepsis. SEPSIS 2017. [DOI: 10.1007/978-3-319-48470-9_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
31
|
Pereira S, Nogueira L, Canova F, Lopez M, Silva HC. IRAK1 variant is protective for orthodontic-induced external apical root resorption. Oral Dis 2016; 22:658-64. [PMID: 27250598 DOI: 10.1111/odi.12514] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/21/2016] [Accepted: 05/21/2016] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Interleukin-1 beta (IL1B) pathway is a key player in orthodontic-induced external apical root resorption (EARR). The aim of this work was to identify the genes related to the IL1 pathway as possible candidate genes for EARR, which might be included in an integrative predictive model of this complex phenotype. MATERIALS AND METHODS Using a stepwise multiple linear regression model, 195 patients who had undergone orthodontic treatment were assessed for clinical and genetic factors associated with %EARRmax (maximum %EARR value obtained for each patient). The four maxillary incisors and the two maxillary canines were assessed. Three functional single nucleotide polymorphisms (SNPs) were genotyped: rs1143634 in IL1B gene, rs315952 in IL1RN gene, and rs1059703 in X-linked IRAK1 gene. RESULTS The model showed that four of the nine clinical variables and one SNP explained 30% of the %EARRmax variability. The most significant unique contributions to the model were gender (P = 0.001), treatment duration (P < 0.001), premolar extractions (P = 0.003), Hyrax appliance (P < 0.001), and homozygosity/hemizygosity for variant C from IRAK1 gene (P = 0.018), which proved to be a protective factor. CONCLUSION IRAK1 polymorphism is proposed as a protective variant for EARR.
Collapse
Affiliation(s)
- S Pereira
- Department of Orthodontics, Dentistry Area, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| | - L Nogueira
- Medical Genetics Department, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - F Canova
- Department of Physics and Mathematics, Polytechnic Institute of Coimbra (IPC), Coimbra, Portugal
| | - M Lopez
- Computer Graphics Center, University of Minho, Guimarães, Portugal
| | - H C Silva
- Medical Genetics Department, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CIMAGO (Center of Investigation on Environmental, Genetics and Oncobiology), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
32
|
Siegler BH, Brenner T, Uhle F, Weiterer S, Weigand MA, Hofer S. Why a second look might be worth it: immuno-modulatory therapies in the critically ill patient. J Thorac Dis 2016; 8:E424-30. [PMID: 27293871 DOI: 10.21037/jtd.2016.04.37] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Sepsis and septic shock are associated with high mortality rates and remain a serious menace for the critically ill patient. Concurrent activation of pro- and anti-inflammatory pathways and an excessive cytokine release represent initial key features in the deregulation of the humoral and cellular antimicrobial defense. Research of the last decades addressed both the ebullient inflammation as well as the resulting long-term failure of the host immunity. While the reestablishment of an adequate immune-competence is still under investigation, many promising experimental trials to limit the inflammatory response during sepsis were challenged by missing beneficial effects in clinical studies. Nevertheless, due to advanced knowledge about the complex regulation of inflammatory mediators and their overlapping involvement in other potentially life-threatening diseases, further evaluation of these approaches in relevant subgroups could help to identify critically ill patients with potential benefit from anti-inflammatory therapies.
Collapse
Affiliation(s)
- Benedikt H Siegler
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Thorsten Brenner
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Florian Uhle
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Sebastian Weiterer
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Markus A Weigand
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Hofer
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW The review will update readers on research examining the influence of genetic variation and epigenetics on the immune system and whether genetic variation influences the outcome of critically ill children. RECENT FINDINGS Although there have been few recent studies examining the role of genetic variation in the severity of disease or outcome in critically ill children, studies in critically ill adults have been informative. For example, genetic variations in the genes coding for various components of the immune response, such as the Toll-like receptor 1, interleukin-1RA, proprotein convertase subtilisin/kexin type 9, adoponectin, nuclear factor erythroid 2-related factor 2, elafin, sphingosine 1-phosphate receptor 3, and sushi, von Willebrand factor type A, EGF and pentraxin domain containing 1 have been associated with various outcomes in critically ill adult populations. Many of the variants demonstrate functional consequences in the protein levels or activities. In critically ill children, there is an association with increased ICU length of stay in children with septic shock with one of the Toll-like receptor 1 variants. SUMMARY The degree of influence of host genetic variation in the outcome in critically ill children remains a much understudied area of research. However, it remains important because it may not only help identify children at risk for worse outcomes but it may provide insight into mechanisms of critical illnesses and novel therapies.
Collapse
|
34
|
Palakshappa JA, Anderson BJ, Reilly JP, Shashaty MGS, Ueno R, Wu Q, Ittner CAG, Tommasini A, Dunn TG, Charles D, Kazi A, Christie JD, Meyer NJ. Low Plasma Levels of Adiponectin Do Not Explain Acute Respiratory Distress Syndrome Risk: a Prospective Cohort Study of Patients with Severe Sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2016; 20:71. [PMID: 26984771 PMCID: PMC4794929 DOI: 10.1186/s13054-016-1244-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/17/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND Obesity is associated with the development of acute respiratory distress syndrome (ARDS) in at-risk patients. Low plasma levels of adiponectin, a circulating hormone-like molecule, have been implicated as a possible mechanism for this association. The objective of this study was to determine the association of plasma adiponectin level at ICU admission with ARDS and 30-day mortality in patients with severe sepsis and septic shock. METHODS This is a prospective cohort study of patients admitted to the medical ICU at the Hospital of the University of Pennsylvania. Plasma adiponectin was measured at the time of ICU admission. ARDS was defined by Berlin criteria. Multivariable logistic regression was used to determine the association of plasma adiponectin with the development of ARDS and mortality at 30 days. RESULTS The study included 164 patients. The incidence of ARDS within 5 days of admission was 45%. The median initial plasma adiponectin level was 7.62 mcg/ml (IQR: 3.87, 14.90) in those without ARDS compared to 8.93 mcg/ml (IQR: 4.60, 18.85) in those developing ARDS. The adjusted odds ratio for ARDS associated with each 5 mcg increase in adiponectin was 1.12 (95% CI 1.01, 1.25), p-value 0.025). A total of 82 patients (51%) of the cohort died within 30 days of ICU admission. There was a statistically significant association between adiponectin and mortality in the unadjusted model (OR 1.11, 95% CI 1.00, 1.23, p-value 0.04) that was no longer significant after adjusting for potential confounders. CONCLUSIONS In this study, low levels of adiponectin were not associated with an increased risk of ARDS in patients with severe sepsis and septic shock. This argues against low levels of adiponectin as a mechanism explaining the association of obesity with ARDS. At present, it is unclear whether circulating adiponectin is involved in the pathogenesis of ARDS or simply represents an epiphenomenon of other unknown functions of adipose tissue or metabolic alterations in sepsis.
Collapse
Affiliation(s)
- Jessica A Palakshappa
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA.
| | - Brian J Anderson
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - John P Reilly
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Blockley Hall, 423 Guardian Drive, Philadelphia, PA, 19104, USA
| | - Michael G S Shashaty
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Blockley Hall, 423 Guardian Drive, Philadelphia, PA, 19104, USA
| | - Ryo Ueno
- Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 1130033, Japan
| | - Qufei Wu
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Caroline A G Ittner
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Anna Tommasini
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Thomas G Dunn
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Dudley Charles
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Altaf Kazi
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Jason D Christie
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Blockley Hall, 423 Guardian Drive, Philadelphia, PA, 19104, USA
| | - Nuala J Meyer
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| |
Collapse
|
35
|
Wang F, Meyer NJ, Walley KR, Russell JA, Feng R. Causal Genetic Inference Using Haplotypes as Instrumental Variables. Genet Epidemiol 2015; 40:35-44. [PMID: 26625855 DOI: 10.1002/gepi.21940] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 08/15/2015] [Accepted: 09/19/2015] [Indexed: 02/03/2023]
Abstract
In genomic studies with both genotypes and gene or protein expression profile available, causal effects of gene or protein on clinical outcomes can be inferred through using genetic variants as instrumental variables (IVs). The goal of introducing IV is to remove the effects of unobserved factors that may confound the relationship between the biomarkers and the outcome. A valid inference under the IV framework requires pairwise associations and pathway exclusivity. Among these assumptions, the IV expression association needs to be strong for the casual effect estimates to be unbiased. However, a small number of single nucleotide polymorphisms (SNPs) often provide limited explanation of the variability in the gene or protein expression and can only serve as weak IVs. In this study, we propose to replace SNPs with haplotypes as IVs to increase the variant-expression association and thus improve the casual effect inference of the expression. In the classical two-stage procedure, we developed a haplotype regression model combined with a model selection procedure to identify optimal instruments. The performance of the new method was evaluated through simulations and compared with the IV approaches using observed multiple SNPs. Our results showed the gain of power to detect a causal effect of gene or protein on the outcome using haplotypes compared with using only observed SNPs, under either complete or missing genotype scenarios. We applied our proposed method to a study of the effect of interleukin-1 beta (IL-1β) protein expression on the 90-day survival following sepsis and found that overly expressed IL-1β is likely to increase mortality.
Collapse
Affiliation(s)
- Fan Wang
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Nuala J Meyer
- Center for Translational Lung Biology, Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Keith R Walley
- Center for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - James A Russell
- Center for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rui Feng
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
36
|
Hill-Batorski L, Halfmann P, Marzi A, Lopes TJS, Neumann G, Feldmann H, Kawaoka Y. Loss of Interleukin 1 Receptor Antagonist Enhances Susceptibility to Ebola Virus Infection. J Infect Dis 2015. [PMID: 26209680 DOI: 10.1093/infdis/jiv335] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The current outbreak of Ebola virus (EBOV) infection in West Africa is unprecedented, with nearly 26 000 confirmed cases and >10 000 deaths. Comprehensive data on the pathogenesis of EBOV infection are lacking; however, recent studies suggested that fatal EBOV infections are characterized by dysregulation of the innate immune response and a subsequent cytokine storm. Specifically, several studies suggested that hypersecretion of interleukin 1 receptor antagonist (IL-1Ra) correlates with lethal EBOV infections. To examine the significance of IL-1Ra in EBOV infections, we infected mice that lack the gene encoding IL-1Ra, Il1rn (IL-1RN-KO), and mice with wild-type Il1rn (IL-1RN-WT) with a mouse-adapted EBOV (MA-EBOV). Infected IL-1RN-KO mice lost more weight and had a lower survival rate than IL-1RN-WT mice infected with MA-EBOV. In addition, IL-1RN-KO mice infected with wild-type EBOV, which does not cause lethal infection in adult immunocompetent mice, such as C57BL/6 mice, experienced greater weight loss than IL-1RN-WT mice infected with wild-type EBOV. Further studies revealed that the levels of 6 cytokines in spleens-IL-1α, IL-1β, interleukin 12p40, interleukin 17, granulocyte colony-stimulating factor, and regulated on activation, normal T-cell expressed and secreted-were significantly different between IL-1RN-KO mice and IL-1RN-WT mice infected with MA-EBOV. Collectively, our data suggest that IL-1Ra may have a protective effect upon EBOV infection, likely by damping an overactive proinflammatory immune response.
Collapse
Affiliation(s)
- Lindsay Hill-Batorski
- Department of Pathobiological Sciences, School of Veterinary Medicine, Influenza Research Institute, University of Wisconsin-Madison
| | - Peter Halfmann
- Department of Pathobiological Sciences, School of Veterinary Medicine, Influenza Research Institute, University of Wisconsin-Madison
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Tiago J S Lopes
- Department of Pathobiological Sciences, School of Veterinary Medicine, Influenza Research Institute, University of Wisconsin-Madison Division of Virology, Department of Microbiology and Immunology, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo
| | - Gabriele Neumann
- Department of Pathobiological Sciences, School of Veterinary Medicine, Influenza Research Institute, University of Wisconsin-Madison
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, School of Veterinary Medicine, Influenza Research Institute, University of Wisconsin-Madison Division of Virology, Department of Microbiology and Immunology, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo ERATO Infection-Induced Host Responses Project, Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
37
|
Schortgen F, Asfar P. Update in Sepsis and Acute Kidney Injury 2014. Am J Respir Crit Care Med 2015; 191:1226-31. [DOI: 10.1164/rccm.201502-0307up] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
38
|
Wong HR. Personalized medicine, endotypes, and intensive care medicine. Intensive Care Med 2015; 41:1138-40. [PMID: 25904185 DOI: 10.1007/s00134-015-3812-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 04/07/2015] [Indexed: 11/24/2022]
Affiliation(s)
- Hector R Wong
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center and Cincinnati Children's Research Foundation, Cincinnati, OH, USA,
| |
Collapse
|
39
|
Patel PN, Shah RY, Ferguson JF, Reilly MP. Human experimental endotoxemia in modeling the pathophysiology, genomics, and therapeutics of innate immunity in complex cardiometabolic diseases. Arterioscler Thromb Vasc Biol 2015; 35:525-34. [PMID: 25550206 PMCID: PMC4344396 DOI: 10.1161/atvbaha.114.304455] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 12/18/2014] [Indexed: 01/16/2023]
Abstract
Inflammation is a fundamental feature of several complex cardiometabolic diseases. Indeed, obesity, insulin resistance, metabolic dyslipidemia, and atherosclerosis are all closely linked inflammatory states. Increasing evidence suggests that the infectious, biome-related, or endogenous activation of the innate immune system may contribute to the development of metabolic syndrome and cardiovascular disease. Here, we describe the human experimental endotoxemia model for the specific study of innate immunity in understanding further the pathogenesis of cardiometabolic disease. In a controlled, experimental setting, administration of an intravenous bolus of purified Escherichia coli endotoxin activates innate immunity in healthy human volunteers. During endotoxemia, changes emerge in glucose metabolism, lipoprotein composition, and lipoprotein functions that closely resemble those observed chronically in inflammatory cardiovascular disease risk states. In this review, we describe the transient systemic inflammation and specific metabolic consequences that develop during human endotoxemia. Such a model provides a controlled induction of systemic inflammation, eliminates confounding, undermines reverse causation, and possesses unique potential as a starting point for genomic screening and testing of novel therapeutics for treatment of the inflammatory underpinning of cardiometabolic disease.
Collapse
Affiliation(s)
- Parth N Patel
- From the Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (P.N.P., R.Y.S., M.P.R.); and Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN (J.F.F.)
| | - Rhia Y Shah
- From the Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (P.N.P., R.Y.S., M.P.R.); and Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN (J.F.F.)
| | - Jane F Ferguson
- From the Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (P.N.P., R.Y.S., M.P.R.); and Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN (J.F.F.)
| | - Muredach P Reilly
- From the Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (P.N.P., R.Y.S., M.P.R.); and Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN (J.F.F.).
| |
Collapse
|
40
|
Wurfel MM, Mikacenic C. IL1RN single-nucleotide polymorphisms in septic shock: can genetics clear the fog of the cytokine storm? Am J Respir Crit Care Med 2014; 190:599-600. [PMID: 25221876 DOI: 10.1164/rccm.201408-1506ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Mark M Wurfel
- 1 Division of Pulmonary and Critical Care Medicine University of Washington Seattle, Washington
| | | |
Collapse
|
41
|
Yang SA. Exonic polymorphism (rs315952, Ser133Ser) of interleukin 1 receptor antagonist (IL1RN) is related to overweigh/obese with hypertension. J Exerc Rehabil 2014; 10:332-6. [PMID: 25426473 PMCID: PMC4237851 DOI: 10.12965/jer.140155] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 10/15/2014] [Indexed: 02/03/2023] Open
Abstract
Recent studies demonstrated that interleukin 1 receptor antagonist (IL-1RN) plays an important role in metabolic effects. To investigate whether IL1RN polymorphisms are associated with obesity, two single nucleotide polymorphisms (SNPs) of the IL1RN gene [rs4251961 (-828, T> C) and rs315952 (Ser133Ser)] were analyzed in 122 overweigh/obese and 123 control subjects. Overweigh/obese subjects were classified according to body mass index (BMI). SNPStats was used to obtain odds ratios (ORs), 95% confidence intervals (CIs), and P values. Multiple logistic regression models (codominant1, codominant2, dominant, recessive, and log-additive) were conducted to analyze the genetic data. Synonymous SNP (rs315952) of the IL1RN gene was associated with overweigh/obese with hypertension (OR= 4.98, 95% CI= 1.74-14.19, P = 0.003 in codominant 1 model and OR= 3.98, 95% CI= 1.48-10.74, P= 0.0029 in dominant model). However, another SNP (rs4151961) did not show association with overweigh/obese or overweigh/obese with hypertension. These results suggest that exonic SNP of IL1RN (rs 315952, Ser133Ser) may be contributed to overweigh/obese with hyper-tension.
Collapse
Affiliation(s)
- Seung-Ae Yang
- College of Nursing, Sungshin Women's University, Seoul, Korea
| |
Collapse
|