1
|
Gregory DJ, Han F, Li P, Gritsenko MA, Kyle J, Riley FE, Chavez D, Yotova V, Sindeaux RHM, Hawash MBF, Xu F, Hung LY, Hayden DL, Tompkins RG, Lanford RE, Kobzik L, Hellman J, Jacobs JM, Barreiro LB, Xiao W, Warren HS. Molecular profiles of blood from numerous species that differ in sensitivity to acute inflammation. Mol Med 2024; 30:280. [PMID: 39730996 DOI: 10.1186/s10020-024-01052-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024] Open
Abstract
Vertebrates differ over 100,000-fold in responses to pro-inflammatory agonists such as bacterial lipopolysaccharide (LPS), complicating use of animal models to study human sepsis or inflammatory disorders. We compared transcriptomes of resting and LPS-exposed blood from six LPS-sensitive species (rabbit, pig, sheep, cow, chimpanzee, human) and four LPS-resilient species (mice, rats, baboon, rhesus), as well as plasma proteomes and lipidomes. Unexpectedly, at baseline, sensitive species already had enhanced expression of LPS-responsive genes relative to resilient species. After LPS stimulation, maximally different genes in resilient species included genes that detoxify LPS, diminish bacterial growth, discriminate sepsis from SIRS, and play roles in autophagy and apoptosis. The findings reveal the molecular landscape of species differences in inflammation. This may inform better selection of species for pre-clinical models and could lead to new therapeutic strategies that mimic mechanisms in inflammation-resilient species to limit inflammation without causing immunosuppression.
Collapse
Affiliation(s)
- David J Gregory
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Feifei Han
- Harvard Medical School, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Peng Li
- Harvard Medical School, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Marina A Gritsenko
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Jennifer Kyle
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Frank E Riley
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
| | - Deborah Chavez
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Vania Yotova
- Centre Hospitalier Universitaire Sainte-Justine, Montréal, QC, Canada
| | | | - Mohamed B F Hawash
- Centre Hospitalier Universitaire Sainte-Justine, Montréal, QC, Canada
- Department of Biochemistry, University of Montréal, Montréal, QC, Canada
- Hospital for Sick Children, Toronto, Canada
| | - Fengyun Xu
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Li-Yuan Hung
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Douglas L Hayden
- Harvard Medical School, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ronald G Tompkins
- Harvard Medical School, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Robert E Lanford
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Lester Kobzik
- Program in Molecular and Integrative Physiological Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Judith Hellman
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Jon M Jacobs
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Luis B Barreiro
- Centre Hospitalier Universitaire Sainte-Justine, Montréal, QC, Canada
- Department of Biochemistry, University of Montréal, Montréal, QC, Canada
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Committee On Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, IL, USA
- Committee On Immunology, University of Chicago, Chicago, IL, USA
| | - Wenzhong Xiao
- Harvard Medical School, Boston, MA, USA.
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - H Shaw Warren
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
2
|
Jonassen TB, Jørgensen SE, Mitchell NH, Mogensen TH, Berg RMG, Ronit A, Plovsing RR. Alveolar cytokines and interferon autoantibodies in COVID-19 ARDS. Front Immunol 2024; 15:1353012. [PMID: 38571960 PMCID: PMC10987806 DOI: 10.3389/fimmu.2024.1353012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 02/26/2024] [Indexed: 04/05/2024] Open
Abstract
Background Type I interferon (IFN-I) and IFN autoantibodies play a crucial role in controlling SARS-CoV-2 infection. The levels of these mediators have only rarely been studied in the alveolar compartment in patients with COVID-19 acute respiratory distress syndrome (CARDS) but have not been compared across different ARDS etiologies, and the potential effect of dexamethasone (DXM) on these mediators is not known. Methods We assessed the integrity of the alveolo-capillary membrane, interleukins, type I, II, and III IFNs, and IFN autoantibodies by studying the epithelial lining fluid (ELF) volumes, alveolar concentration of protein, and ELF-corrected concentrations of cytokines in two patient subgroups and controls. Results A total of 16 patients with CARDS (four without and 12 with DXM treatment), eight with non-CARDS, and 15 healthy controls were included. The highest ELF volumes and protein levels were observed in CARDS. Systemic and ELF-corrected alveolar concentrations of interleukin (IL)-6 appeared to be particularly low in patients with CARDS receiving DXM, whereas alveolar levels of IL-8 were high regardless of DXM treatment. Alveolar levels of IFNs were similar between CARDS and non-CARDS patients, and IFNα and IFNω autoantibody levels were higher in patients with CARDS and non-CARDS than in healthy controls. Conclusions Patients with CARDS exhibited greater alveolo-capillary barrier disruption with compartmentalization of IL-8, regardless of DXM treatment, whereas systemic and alveolar levels of IL-6 were lower in the DXM-treated subgroup. IFN-I autoantibodies were higher in the BALF of CARDS patients, independent of DXM, whereas IFN autoantibodies in plasma were similar to those in controls.
Collapse
Affiliation(s)
- Trine B. Jonassen
- Department of Anesthesiology and Intensive Care, Copenhagen University Hospital-Amager and Hvidovre Hospitals, Hvidovre, Denmark
| | - Sofie E. Jørgensen
- Department of Infectious Diseases, Aarhus University Hospital (AUH), Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Nikki H. Mitchell
- Department of Clinical Biochemistry, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | - Trine H. Mogensen
- Department of Infectious Diseases, Aarhus University Hospital (AUH), Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Ronan M. G. Berg
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Centre for Physical Activity Research, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Neurovascular Research Laboratory, Faculty of Life Sciences and Education, University of South Wales, Pontypridd, United Kingdom
| | - Andreas Ronit
- Department of Infectious Diseases, Copenhagen University Hospital-Amager and Hvidovre Hospitals, Hvidovre, Denmark
| | - Ronni R. Plovsing
- Department of Anesthesiology and Intensive Care, Copenhagen University Hospital-Amager and Hvidovre Hospitals, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Shukla PK, Rao RG, Meena AS, Giorgianni F, Lee SC, Raju P, Shashikanth N, Shekhar C, Beranova S, Balazs L, Tigyi G, Gosain A, Rao R. Paneth cell dysfunction in radiation injury and radio-mitigation by human α-defensin 5. Front Immunol 2023; 14:1174140. [PMID: 37638013 PMCID: PMC10448521 DOI: 10.3389/fimmu.2023.1174140] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 07/14/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction The mechanism underlying radiation-induced gut microbiota dysbiosis is undefined. This study examined the effect of radiation on the intestinal Paneth cell α-defensin expression and its impact on microbiota composition and mucosal tissue injury and evaluated the radio-mitigative effect of human α-defensin 5 (HD5). Methods Adult mice were subjected to total body irradiation, and Paneth cell α-defensin expression was evaluated by measuring α-defensin mRNA by RT-PCR and α-defensin peptide levels by mass spectrometry. Vascular-to-luminal flux of FITC-inulin was measured to evaluate intestinal mucosal permeability and endotoxemia by measuring plasma lipopolysaccharide. HD5 was administered in a liquid diet 24 hours before or after irradiation. Gut microbiota was analyzed by 16S rRNA sequencing. Intestinal epithelial junctions were analyzed by immunofluorescence confocal microscopy and mucosal inflammatory response by cytokine expression. Systemic inflammation was evaluated by measuring plasma cytokine levels. Results Ionizing radiation reduced the Paneth cell α-defensin expression and depleted α-defensin peptides in the intestinal lumen. α-Defensin down-regulation was associated with the time-dependent alteration of gut microbiota composition, increased gut permeability, and endotoxemia. Administration of human α-defensin 5 (HD5) in the diet 24 hours before irradiation (prophylactic) significantly blocked radiation-induced gut microbiota dysbiosis, disruption of intestinal epithelial tight junction and adherens junction, mucosal barrier dysfunction, and mucosal inflammatory response. HD5, administered 24 hours after irradiation (treatment), reversed radiation-induced microbiota dysbiosis, tight junction and adherens junction disruption, and barrier dysfunction. Furthermore, HD5 treatment also prevents and reverses radiation-induced endotoxemia and systemic inflammation. Conclusion These data demonstrate that radiation induces Paneth cell dysfunction in the intestine, and HD5 feeding prevents and mitigates radiation-induced intestinal mucosal injury, endotoxemia, and systemic inflammation.
Collapse
Affiliation(s)
- Pradeep K. Shukla
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Roshan G. Rao
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Avtar S. Meena
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Francesco Giorgianni
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sue Chin Lee
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Preeti Raju
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Nitesh Shashikanth
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Chandra Shekhar
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sarka Beranova
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Louisa Balazs
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Gabor Tigyi
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ankush Gosain
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - RadhaKrishna Rao
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
4
|
Brownlow MA, Mizzi JX. Pathophysiology of exertional heat illness in the Thoroughbred racehorse: Broadening perspective to include an exercise‐induced gastrointestinal syndrome in which endotoxaemia and systemic inflammation may contribute to the condition. EQUINE VET EDUC 2022. [DOI: 10.1111/eve.13750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | - James Xavier Mizzi
- Department of Regulation, Welfare and Biosecurity Policy The Hong Kong Jockey Club, Sha Tin Racecourse Sha Tin Hong Kong
| |
Collapse
|
5
|
Antwi S, Oduro-Mensah D, Obiri DD, Osafo N, Antwi AO, Ansah HO, Ocloo A, Okine LKNA. Hydro-ethanol extract of Holarrhena floribunda stem bark exhibits anti-anaphylactic and anti-oedematogenic effects in murine models of acute inflammation. BMC Complement Med Ther 2022; 22:80. [PMID: 35305615 PMCID: PMC8934059 DOI: 10.1186/s12906-022-03565-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 03/10/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Holarrhena floribunda (G.Don) T.Durand & Schinz stem bark has anecdotal use in Ghanaian folk medicine for the management of inflammatory conditions. This study was conducted to investigate the in vivo anti-inflammatory activity of the bark extract using models of acute inflammation in male Sprague Dawley rats, C57BL/6 mice and ICR mice. METHODS A 70% hydro-ethanol extract of the stem bark (HFE) was evaluated at doses of 5-500 mg/kg bw. Local anaphylaxis was modelled by the pinnal cutaneous anaphylactic test. Systemic anaphylaxis or sepsis were modeled by compound 48/80 or lipopolysaccharide, respectively. Clonidine-induced catalepsy was used to investigate the effect on histamine signaling. Anti-oedematogenic effect was assessed by induction with carrageenan. Effects on mediators of biphasic acute inflammation were studied using histamine and serotonin (early phase) or prostaglandin E2 (late phase). RESULTS HFE demonstrated anti-inflammatory and/or anti-oedematogenic activity comparable to standard doses of aspirin and diclofenac (inhibitors of cyclooxygenases-1 and -2), chlorpheniramine (histamine H1-receptor antagonist), dexamethasone (glucocorticoid receptor agonist), granisetron (serotonin receptor antagonist) and sodium cromoglycate (inhibitor of mast cell degranulation). All observed HFE bioactivities increased with dose. CONCLUSIONS The data provide evidence that the extract of H. floribunda stem bark has anti-anaphylactic and anti-oedematogenic effects; by interfering with signalling or metabolism of histamine, serotonin and prostaglandin E2 which mediate the progression of inflammation. The anti-inflammatory and antihistaminic activities of HFE may be relevant in the context of the management of COVID-19.
Collapse
Affiliation(s)
- Stephen Antwi
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Pharmacology/Toxicology Department, Centre for Plant Medicine Research (CPMR), Mampong-Akuapem, Ghana
| | - Daniel Oduro-Mensah
- Department of Biochemistry, Cell and Molecular Biology, School of Biological Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Ghana.
| | - David Darko Obiri
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Newman Osafo
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Aaron Opoku Antwi
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Helena Owusu Ansah
- Pharmacology/Toxicology Department, Centre for Plant Medicine Research (CPMR), Mampong-Akuapem, Ghana
| | - Augustine Ocloo
- Department of Biochemistry, Cell and Molecular Biology, School of Biological Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Ghana
| | - Laud K N-A Okine
- Department of Biochemistry, Cell and Molecular Biology, School of Biological Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Ghana
| |
Collapse
|
6
|
Negrini TDC, Carlos IZ, Duque C, Caiaffa KS, Arthur RA. Interplay Among the Oral Microbiome, Oral Cavity Conditions, the Host Immune Response, Diabetes Mellitus, and Its Associated-Risk Factors-An Overview. FRONTIERS IN ORAL HEALTH 2022; 2:697428. [PMID: 35048037 PMCID: PMC8757730 DOI: 10.3389/froh.2021.697428] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
This comprehensive review of the literature aimed to investigate the interplay between the oral microbiome, oral cavity conditions, and host immune response in Diabetes mellitus (DM). Moreover, this review also aimed to investigate how DM related risk factors, such as advanced age, hyperglycemia, hyperlipidemia, obesity, hypertension and polycystic ovary syndrome (PCOS), act in promoting or modifying specific mechanisms that could potentially perpetuate both altered systemic and oral conditions. We found that poorly controlled glycemic index may exert a negative effect on the immune system of affected individuals, leading to a deficient immune response or to an exacerbation of the inflammatory response exacerbating DM-related complications. Hyperglycemia induces alterations in the oral microbiome since poor glycemic control is associated with increased levels and frequencies of periodontal pathogens in the subgingival biofilm of individuals with DM. A bidirectional relationship between periodontal diseases and DM has been suggested: DM patients may have an exaggerated inflammatory response, poor repair and bone resorption that aggravates periodontal disease whereas the increased levels of systemic pro-inflammatory mediators found in individuals affected with periodontal disease exacerbates insulin resistance. SARS-CoV-2 infection may represent an aggravating factor for individuals with DM. Individuals with DM tend to have low salivary flow and a high prevalence of xerostomia, but the association between prevalence/experience of dental caries and DM is still unclear. DM has also been associated to the development of lesions in the oral mucosa, especially potentially malignant ones and those associated with fungal infections. Obesity plays an important role in the induction and progression of DM. Co-affected obese and DM individuals tend to present worse oral health conditions. A decrease in HDL and, an increase in triglycerides bloodstream levels seem to be associated with an increase on the load of periodontopathogens on oral cavity. Moreover, DM may increase the likelihood of halitosis. Prevalence of impaired taste perception and impaired smell recognition tend to be greater in DM patients. An important interplay among oral cavity microbiome, DM, obesity and hypertension has been proposed as the reduction of nitrate into nitrite, in addition to contribute to lowering of blood pressure, reduces oxidative stress and increases insulin secretion, being these effects desirable for the control of obesity and DM. Women with PCOS tend to present a distinct oral microbial composition and an elevated systemic response to selective members of this microbial community, but the association between oral microbiome, PCOS are DM is still unknown. The results of the studies presented in this review suggest the interplay among the oral microbiome, oral cavity conditions, host immune response and DM and some of the DM associated risk factors exist. DM individuals need to be encouraged and motivated for an adequate oral health care. In addition, these results show the importance of adopting multidisciplinary management of DM and of strengthening physicians-dentists relationship focusing on both systemic and on oral cavity conditions of DM patients.
Collapse
Affiliation(s)
- Thais de Cássia Negrini
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| | - Iracilda Zeppone Carlos
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| | - Cristiane Duque
- Department of Restorative and Preventive Dentistry, Araçatuba Dental School, São Paulo State University, Araçatuba, Brazil
| | - Karina Sampaio Caiaffa
- Department of Restorative and Preventive Dentistry, Araçatuba Dental School, São Paulo State University, Araçatuba, Brazil
| | - Rodrigo Alex Arthur
- Department of Preventive and Community Dentistry, Dental School, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
7
|
OUP accepted manuscript. J Antimicrob Chemother 2022; 77:1424-1431. [DOI: 10.1093/jac/dkac039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/14/2022] [Indexed: 11/13/2022] Open
|
8
|
Matsumoto H, Scicluna BP, Jim KK, Falahi F, Qin W, Gürkan B, Malmström E, Meijer MT, Butler JM, Khan HN, Takagi T, Ishii S, Schultz MJ, van de Beek D, de Vos AF, van 't Veer C, van der Poll T. HIVEP1 Is a Negative Regulator of NF-κB That Inhibits Systemic Inflammation in Sepsis. Front Immunol 2021; 12:744358. [PMID: 34804025 PMCID: PMC8602905 DOI: 10.3389/fimmu.2021.744358] [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: 07/20/2021] [Accepted: 10/12/2021] [Indexed: 11/23/2022] Open
Abstract
Our previous work identified human immunodeficiency virus type I enhancer binding protein 1 (HIVEP1) as a putative driver of LPS-induced NF-κB signaling in humans in vivo. While HIVEP1 is known to interact with NF-ĸB binding DNA motifs, its function in mammalian cells is unknown. We report increased HIVEP1 mRNA expression in monocytes from patients with sepsis and monocytes stimulated by Toll-like receptor agonists and bacteria. In complementary overexpression and gene deletion experiments HIVEP1 was shown to inhibit NF-ĸB activity and induction of NF-ĸB responsive genes. RNA sequencing demonstrated profound transcriptomic changes in HIVEP1 deficient monocytic cells and transcription factor binding site analysis showed enrichment for κB site regions. HIVEP1 bound to the promoter regions of NF-ĸB responsive genes. Inhibition of cytokine production by HIVEP1 was confirmed in LPS-stimulated murine Hivep1-/- macrophages and HIVEP1 knockdown zebrafish exposed to the common sepsis pathogen Streptococcus pneumoniae. These results identify HIVEP1 as a negative regulator of NF-κB in monocytes/macrophages that inhibits proinflammatory reactions in response to bacterial agonists in vitro and in vivo.
Collapse
Affiliation(s)
- Hisatake Matsumoto
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Brendon P Scicluna
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Kin Ki Jim
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Neurology, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Fahimeh Falahi
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Wanhai Qin
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Berke Gürkan
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Erik Malmström
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Mariska T Meijer
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Joe M Butler
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Hina N Khan
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Tsuyoshi Takagi
- Department of Disease Model, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Aichi, Japan
| | | | - Marcus J Schultz
- Department of Intensive Care Medicine, Laboratory of Experimental Intensive Care and Anesthesiology (L·E·I·C·A), Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Mahidol-Oxford Tropical Medicine Research Unit (MORU), Mahidol University, Bangkok, Thailand.,Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Diederik van de Beek
- Department of Neurology, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Alex F de Vos
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Cornelis van 't Veer
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Division of Infectious Diseases, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
9
|
Eleveld N, Hoedemaekers CWE, van Kaam CR, Leijte GP, van den Brule JMD, Pickkers P, Aries MJH, Maurits NM, Elting JWJ. Near-Infrared Spectroscopy-Derived Dynamic Cerebral Autoregulation in Experimental Human Endotoxemia-An Exploratory Study. Front Neurol 2021; 12:695705. [PMID: 34566840 PMCID: PMC8461327 DOI: 10.3389/fneur.2021.695705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 08/11/2021] [Indexed: 11/13/2022] Open
Abstract
Cerebral perfusion may be altered in sepsis patients. However, there are conflicting findings on cerebral autoregulation (CA) in healthy participants undergoing the experimental endotoxemia protocol, a proxy for systemic inflammation in sepsis. In the current study, a newly developed near-infrared spectroscopy (NIRS)-based CA index is investigated in an endotoxemia study population, together with an index of focal cerebral oxygenation. Methods: Continuous-wave NIRS data were obtained from 11 healthy participants receiving a continuous infusion of bacterial endotoxin for 3 h (ClinicalTrials.gov NCT02922673) under extensive physiological monitoring. Oxygenated–deoxygenated hemoglobin phase differences in the (very)low frequency (VLF/LF) bands and the Tissue Saturation Index (TSI) were calculated at baseline, during systemic inflammation, and at the end of the experiment 7 h after the initiation of endotoxin administration. Results: The median (inter-quartile range) LF phase difference was 16.2° (3.0–52.6°) at baseline and decreased to 3.9° (2.0–8.8°) at systemic inflammation (p = 0.03). The LF phase difference increased from systemic inflammation to 27.6° (12.7–67.5°) at the end of the experiment (p = 0.005). No significant changes in VLF phase difference were observed. The TSI (mean ± SD) increased from 63.7 ± 3.4% at baseline to 66.5 ± 2.8% during systemic inflammation (p = 0.03) and remained higher at the end of the experiment (67.1 ± 4.2%, p = 0.04). Further analysis did not reveal a major influence of changes in several covariates such as blood pressure, heart rate, PaCO2, and temperature, although some degree of interaction could not be excluded. Discussion: A reversible decrease in NIRS-derived cerebral autoregulation phase difference was seen after endotoxin infusion, with a small, sustained increase in TSI. These findings suggest that endotoxin administration in healthy participants reversibly impairs CA, accompanied by sustained microvascular vasodilation.
Collapse
Affiliation(s)
- Nick Eleveld
- Department of Neurology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Cornelia W E Hoedemaekers
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud University, Nijmegen, Netherlands
| | - C Ruud van Kaam
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud University, Nijmegen, Netherlands
| | - Guus P Leijte
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud University, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Radboud University, Nijmegen, Netherlands
| | - Judith M D van den Brule
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud University, Nijmegen, Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud University, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Radboud University, Nijmegen, Netherlands
| | - Marcel J H Aries
- Department of Intensive Care Medicine, School of Mental Health and NeuroSciences (MHeNS), University Medical Center Maastricht (MUMC+), Maastricht University, Maastricht, Netherlands
| | - Natasha M Maurits
- Department of Neurology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jan Willem J Elting
- Department of Neurology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
10
|
Osafo N, Antwi AO, Mante PK, Osei YA, Yeboah OK, Otu-Boakye S. Cutaneous and systemic anti-allergic potential of xylopic acid in rodents. ADVANCES IN TRADITIONAL MEDICINE 2021. [DOI: 10.1007/s13596-021-00595-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
11
|
Zazula R, Moravec M, Pehal F, Nejtek T, Protuš M, Müller M. Myristic Acid Serum Levels and Their Significance for Diagnosis of Systemic Inflammatory Response, Sepsis, and Bacteraemia. J Pers Med 2021; 11:jpm11040306. [PMID: 33923419 PMCID: PMC8074080 DOI: 10.3390/jpm11040306] [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: 03/15/2021] [Revised: 04/08/2021] [Accepted: 04/14/2021] [Indexed: 01/10/2023] Open
Abstract
Myristic acid is identified as a metabolite with the highest diagnostic sensitivity and specificity in the metabolome of patients with bacteraemia. Its significant decrease has been observed in patients with septic shock not responding to treatment. Another study has reported a close correlation of myristic acid levels with the outcome of severe trauma patients. Myristic acid concentrations were investigated in a cohort of septic patients and patients with Systemic Inflammatory Response Syndrome (SIRS) in 5 consecutive days following diagnosis and compared to healthy controls. The study population groups-Sepsis 34, SIRS 31, and Healthy Control 120 patients were included. Serum samples were analyzed using gas chromatography and mass spectrometry. The myristic acid levels in the Sepsis Group and SIRS Group were found to be significantly higher when compared to healthy controls. The serum concentration of myristic acid in septic patients with bacteraemia was higher than in septic patients without bacteraemia. Most patients with sepsis and SIRS had the highest levels of myristic acid within 24 h after an established diagnosis. Myristic acid should be considered as a new candidate marker of severe inflammation and sepsis. A simplified analysis and sufficient body of validated data are necessary steps towards the introduction of this metabolite into routine clinical practice.
Collapse
Affiliation(s)
- Roman Zazula
- Department of Anesthesiology and Intensive Care, First Faculty of Medicine, Charles University and Thomayer University Hospital, 140 59 Prague, Czech Republic; (M.M.); (F.P.); (T.N.); (M.M.)
- Correspondence: ; Tel.: +420-261-083-811
| | - Michal Moravec
- Department of Anesthesiology and Intensive Care, First Faculty of Medicine, Charles University and Thomayer University Hospital, 140 59 Prague, Czech Republic; (M.M.); (F.P.); (T.N.); (M.M.)
| | - František Pehal
- Department of Anesthesiology and Intensive Care, First Faculty of Medicine, Charles University and Thomayer University Hospital, 140 59 Prague, Czech Republic; (M.M.); (F.P.); (T.N.); (M.M.)
| | - Tomáš Nejtek
- Department of Anesthesiology and Intensive Care, First Faculty of Medicine, Charles University and Thomayer University Hospital, 140 59 Prague, Czech Republic; (M.M.); (F.P.); (T.N.); (M.M.)
| | - Marek Protuš
- Department of Anaesthesiology, Resuscitation and Intensive Care, Institute for Clinical and Experimental Medicine, First Faculty of Medicine, Charles University, 140 21 Prague, Czech Republic;
| | - Martin Müller
- Department of Anesthesiology and Intensive Care, First Faculty of Medicine, Charles University and Thomayer University Hospital, 140 59 Prague, Czech Republic; (M.M.); (F.P.); (T.N.); (M.M.)
| |
Collapse
|
12
|
Pecchiari M, Pontikis K, Alevrakis E, Vasileiadis I, Kompoti M, Koutsoukou A. Cardiovascular Responses During Sepsis. Compr Physiol 2021; 11:1605-1652. [PMID: 33792902 DOI: 10.1002/cphy.c190044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Sepsis is the life-threatening organ dysfunction arising from a dysregulated host response to infection. Although the specific mechanisms leading to organ dysfunction are still debated, impaired tissue oxygenation appears to play a major role, and concomitant hemodynamic alterations are invariably present. The hemodynamic phenotype of affected individuals is highly variable for reasons that have been partially elucidated. Indeed, each patient's circulatory condition is shaped by the complex interplay between the medical history, the volemic status, the interval from disease onset, the pathogen, the site of infection, and the attempted resuscitation. Moreover, the same hemodynamic pattern can be generated by different combinations of various pathophysiological processes, so the presence of a given hemodynamic pattern cannot be directly related to a unique cluster of alterations. Research based on endotoxin administration to healthy volunteers and animal models compensate, to an extent, for the scarcity of clinical studies on the evolution of sepsis hemodynamics. Their results, however, cannot be directly extrapolated to the clinical setting, due to fundamental differences between the septic patient, the healthy volunteer, and the experimental model. Numerous microcirculatory derangements might exist in the septic host, even in the presence of a preserved macrocirculation. This dissociation between the macro- and the microcirculation might account for the limited success of therapeutic interventions targeting typical hemodynamic parameters, such as arterial and cardiac filling pressures, and cardiac output. Finally, physiological studies point to an early contribution of cardiac dysfunction to the septic phenotype, however, our defective diagnostic tools preclude its clinical recognition. © 2021 American Physiological Society. Compr Physiol 11:1605-1652, 2021.
Collapse
Affiliation(s)
- Matteo Pecchiari
- Dipartimento di Fisiopatologia Medico Chirurgica e dei Trapianti, Università degli Studi di Milano, Milan, Italy
| | - Konstantinos Pontikis
- Intensive Care Unit, 1st Department of Pulmonary Medicine, National & Kapodistrian University of Athens, General Hospital for Diseases of the Chest 'I Sotiria', Athens, Greece
| | - Emmanouil Alevrakis
- 4th Department of Pulmonary Medicine, General Hospital for Diseases of the Chest 'I Sotiria', Athens, Greece
| | - Ioannis Vasileiadis
- Intensive Care Unit, 1st Department of Pulmonary Medicine, National & Kapodistrian University of Athens, General Hospital for Diseases of the Chest 'I Sotiria', Athens, Greece
| | - Maria Kompoti
- Intensive Care Unit, Thriassio General Hospital of Eleusis, Magoula, Greece
| | - Antonia Koutsoukou
- Intensive Care Unit, 1st Department of Pulmonary Medicine, National & Kapodistrian University of Athens, General Hospital for Diseases of the Chest 'I Sotiria', Athens, Greece
| |
Collapse
|
13
|
Participation of Monocyte Subpopulations in Progression of Experimental Endotoxemia (EE) and Systemic Inflammation. J Immunol Res 2021; 2021:1762584. [PMID: 33628841 PMCID: PMC7895567 DOI: 10.1155/2021/1762584] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 01/26/2021] [Accepted: 02/04/2021] [Indexed: 01/01/2023] Open
Abstract
Systemic inflammation plays a crucial role in formation of various pathological conditions, including sepsis, burns, and traumas. The main effector cells participating in progression of systemic inflammation response and sepsis are monocytes, which regulate both innate and acquired immunity via phagocytosis, synthesis of cytokines and chemokines, antigen presentation, and lymphocyte activation. Thus, the monocytes are considered as a link between innate and acquired immunity. The monocyte subpopulations taken into consideration in the study essentially determine the progression of systemic inflammation and could serve as targets for therapeutic intervention. The complexity of the analysis of pathophysiology of systemic inflammation lies in its high variability conditioned by individual peculiarities of the patients and inflammation progression specifications. To overcome these limitation, model of experimental endotoxemia (EE) is used. The results of EE, in turn, cannot be directly extrapolated on patients with the systemic inflammatory response. This review is dedicated to discussing the role of monocyte subpopulations in progression of systemic inflammation/sepsis and EE.
Collapse
|
14
|
Stančič B, Qvarfordt B, Berglund MM, Brenden N, Sydow Bäckman M, Fransson M, Nordling S, Magnusson PU. The blood endothelial cell chamber - An innovative system to study immune responses in drug development. Int Immunopharmacol 2020; 90:107237. [PMID: 33310662 DOI: 10.1016/j.intimp.2020.107237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/04/2020] [Accepted: 11/23/2020] [Indexed: 10/22/2022]
Abstract
The risk for adverse immune-mediated reactions, associated with the administration of certain immunotherapeutic agents, should be mitigated early. Infusion reactions to monoclonal antibodies and other biopharmaceuticals, known as cytokine release syndrome, can arise from the release of cytokines via the drug target cell, as well as the recruitment of immune effector cells. While several in vitro cytokine release assays have been proposed up to date, many of them lack important blood components, required for this response to occur. The blood endothelial cell chamber model is an in vitro assay, composed of freshly drawn human whole blood and cultured human primary endothelial cells. Herein, its potential to study the compatibility of immunotherapeutics with the human immune system was studied by evaluating three commercially available monoclonal antibodies and bacterial endotoxin lipopolysaccharide. We demonstrate that the anti-CD28 antibody TGN1412 displayed an adaptive cytokine release profile and a distinct IL-2 response, accompanied with increased CD3+ cell recruitment. Alemtuzumab exhibited a clear cytokine response with a mixed adaptive/innate source (IFNγ, TNFα and IL-6). Its immunosuppressive nature is observed in depleted CD3+ cells. Cetuximab, associated with low infusion reactions, showed a very low or absent stimulatory effect on proinflammatory cytokines. In contrast, bacterial endotoxin demonstrated a clear innate cytokine response, defined by TNFα, IL-6 and IL-1β release, accompanied with a strong recruitment of CD14+CD16+ cells. Therefore, the blood endothelial cell chamber model is presented as a valuable in vitro tool to investigate therapeutic monoclonal antibodies with respect to cytokine release and vascular immune cell recruitment.
Collapse
Affiliation(s)
- Brina Stančič
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Dag Hammarskjöldsväg 20, 751 85, Uppsala University, Uppsala, Sweden; Department of Molecular Biology, Universidad Autónoma de Madrid, and Department of Molecular Neuropathology, Center of Molecular Biology Severo Ochoa (UAM-CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain
| | - Bodil Qvarfordt
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Dag Hammarskjöldsväg 20, 751 85, Uppsala University, Uppsala, Sweden
| | | | - Nina Brenden
- Swedish Orphan Biovitrum AB, Tomtebodavägen 23A, 112 76 Solna, Sweden
| | | | - Moa Fransson
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Dag Hammarskjöldsväg 20, 751 85, Uppsala University, Uppsala, Sweden
| | - Sofia Nordling
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Dag Hammarskjöldsväg 20, 751 85, Uppsala University, Uppsala, Sweden
| | - Peetra U Magnusson
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Dag Hammarskjöldsväg 20, 751 85, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
15
|
Scicluna BP, Uhel F, van Vught LA, Wiewel MA, Hoogendijk AJ, Baessman I, Franitza M, Nürnberg P, Horn J, Cremer OL, Bonten MJ, Schultz MJ, van der Poll T. The leukocyte non-coding RNA landscape in critically ill patients with sepsis. eLife 2020; 9:58597. [PMID: 33305733 PMCID: PMC7775110 DOI: 10.7554/elife.58597] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 12/10/2020] [Indexed: 12/26/2022] Open
Abstract
The extent of non-coding RNA alterations in patients with sepsis and their relationship to clinical characteristics, soluble mediators of the host response to infection, as well as an advocated in vivo model of acute systemic inflammation is unknown. Here we obtained whole blood from 156 patients with sepsis and 82 healthy subjects among whom eight were challenged with lipopolysaccharide in a clinically controlled setting (human endotoxemia). Via next-generation microarray analysis of leukocyte RNA we found that long non-coding RNA and, to a lesser extent, small non-coding RNA were significantly altered in sepsis relative to health. Long non-coding RNA expression, but not small non-coding RNA, was largely recapitulated in human endotoxemia. Integrating RNA profiles and plasma protein levels revealed known as well as previously unobserved pathways, including non-sensory olfactory receptor activity. We provide a benchmark dissection of the blood leukocyte ‘regulome’ that can facilitate prioritization of future functional studies.
Collapse
Affiliation(s)
- Brendon P Scicluna
- Amsterdam UMC, University of Amsterdam, Center for Experimental Molecular Medicine, Amsterdam Infection & Immunity, Amsterdam, Netherlands.,Amsterdam UMC, University of Amsterdam, Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam, Netherlands
| | - Fabrice Uhel
- Amsterdam UMC, University of Amsterdam, Center for Experimental Molecular Medicine, Amsterdam Infection & Immunity, Amsterdam, Netherlands
| | - Lonneke A van Vught
- Amsterdam UMC, University of Amsterdam, Center for Experimental Molecular Medicine, Amsterdam Infection & Immunity, Amsterdam, Netherlands
| | - Maryse A Wiewel
- Amsterdam UMC, University of Amsterdam, Center for Experimental Molecular Medicine, Amsterdam Infection & Immunity, Amsterdam, Netherlands
| | - Arie J Hoogendijk
- Amsterdam UMC, University of Amsterdam, Center for Experimental Molecular Medicine, Amsterdam Infection & Immunity, Amsterdam, Netherlands
| | - Ingelore Baessman
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Marek Franitza
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Janneke Horn
- Amsterdam UMC, University of Amsterdam, Department of Intensive Care Medicine, Amsterdam, Netherlands
| | - Olaf L Cremer
- Department of Intensive Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marc J Bonten
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands.,Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marcus J Schultz
- Amsterdam UMC, University of Amsterdam, Department of Intensive Care Medicine, Amsterdam, Netherlands
| | - Tom van der Poll
- Amsterdam UMC, University of Amsterdam, Center for Experimental Molecular Medicine, Amsterdam Infection & Immunity, Amsterdam, Netherlands.,Amsterdam UMC, University of Amsterdam, Division of Infectious Diseases, Amsterdam, Netherlands
| | | |
Collapse
|
16
|
Zhang R, Trower J, Wu T. Degradation of bacterial permeability family member A1 (BPIFA1) by house dust mite (HDM) cysteine protease Der p 1 abrogates immune modulator function. Int J Biol Macromol 2020; 164:4022-4031. [PMID: 32890564 PMCID: PMC7467078 DOI: 10.1016/j.ijbiomac.2020.08.214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/19/2020] [Accepted: 08/28/2020] [Indexed: 01/01/2023]
Abstract
Bacterial permeability family member A1 (BPIFA1) is one of the most abundant proteins present in normal airway surface liquid (ASL). It is known to be diminished in asthmatic patients' sputum, which causes airway hyperresponsiveness (AHR). What is currently unclear is how environmental factors, such as allergens' impact on BPIFA1's abundance and functions in the context of allergic asthma. House dust mite (HDM) is a predominant domestic source of aeroallergens. The group of proteases found in HDM is thought to cleave multiple cellular protective mechanisms, and therefore foster the development of allergic asthma. Here, we show that BPIFA1 is cleaved by HDM proteases in a time-, dose-, and temperature-dependent manner. We have also shown the main component in HDM that is responsible for BPIFA1's degradation is Der p1. Fragmented BPIFA1 failed to bind E. coli lipopolysaccharide (LPS), and hence elevated TNFα and IL-6 secretion in human whole blood. BPIFA1 degradation is also observed in vivo in bronchoalveolar fluid (BALF) of mice which are intranasally instilled with HDM. These data suggest that proteases associated with environmental allergens such as HDM cleave BPIFA1 and therefore impair its immune modulator function.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, PR China
| | - Jessika Trower
- Department of Pharmaceutical Sciences, 302 East Lawson Street, North Carolina Central University, Durham, NC 27707, USA
| | - Tongde Wu
- Department of Pharmaceutical Sciences, 302 East Lawson Street, North Carolina Central University, Durham, NC 27707, USA; Biomanufacturing Research Institute & Technology Enterprise (BRITE), 302 East Lawson Street, North Carolina Central University, Durham, NC 27707, USA.
| |
Collapse
|
17
|
Driedonks TA, Mol S, de Bruin S, Peters AL, Zhang X, Lindenbergh MF, Beuger BM, van Stalborch AMD, Spaan T, de Jong EC, van der Vries E, Margadant C, van Bruggen R, Vlaar AP, Groot Kormelink T, Nolte-‘T Hoen EN. Y-RNA subtype ratios in plasma extracellular vesicles are cell type- specific and are candidate biomarkers for inflammatory diseases. J Extracell Vesicles 2020; 9:1764213. [PMID: 32944168 PMCID: PMC7448942 DOI: 10.1080/20013078.2020.1764213] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/20/2019] [Accepted: 02/25/2020] [Indexed: 02/07/2023] Open
Abstract
Major efforts are made to characterize the presence of microRNA (miRNA) and messenger RNA in blood plasma to discover novel disease-associated biomarkers. MiRNAs in plasma are associated to several types of macromolecular structures, including extracellular vesicles (EV), lipoprotein particles (LPP) and ribonucleoprotein particles (RNP). RNAs in these complexes are recovered at variable efficiency by commonly used EV- and RNA isolation methods, which causes biases and inconsistencies in miRNA quantitation. Besides miRNAs, various other non-coding RNA species are contained in EV and present within the pool of plasma extracellular RNA. Members of the Y-RNA family have been detected in EV from various cell types and are among the most abundant non-coding RNA types in plasma. We previously showed that shuttling of full-length Y-RNA into EV released by immune cells is modulated by microbial stimulation. This indicated that Y-RNAs could contribute to the functional properties of EV in immune cell communication and that EV-associated Y-RNAs could have biomarker potential in immune-related diseases. Here, we investigated which macromolecular structures in plasma contain full length Y-RNA and whether the levels of three Y-RNA subtypes in plasma (Y1, Y3 and Y4) change during systemic inflammation. Our data indicate that the majority of full length Y-RNA in plasma is stably associated to EV. Moreover, we discovered that EV from different blood-related cell types contain cell-type-specific Y-RNA subtype ratios. Using a human model for systemic inflammation, we show that the neutrophil-specific Y4/Y3 ratios and PBMC-specific Y3/Y1 ratios were significantly altered after induction of inflammation. The plasma Y-RNA ratios strongly correlated with the number and type of immune cells during systemic inflammation. Cell-type-specific "Y-RNA signatures" in plasma EV can be determined without prior enrichment for EV, and may be further explored as simple and fast test for diagnosis of inflammatory responses or other immune-related diseases.
Collapse
Affiliation(s)
- Tom A.P. Driedonks
- Department Of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Sanne Mol
- Department Of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- Department Of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Sanne de Bruin
- Department of Intensive Care, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Anna-Linda Peters
- Department Of Anesthesiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Xiaogang Zhang
- Department Of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Marthe F.S. Lindenbergh
- Department Of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Boukje M. Beuger
- Department Of Blood Cell Research, Sanquin Research, and Landsteiner Laboratory, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Anne-Marieke D. van Stalborch
- Molecular Cell Biology Laboratory, Department Of Molecular and Cellular Hemostasis, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Thom Spaan
- Department Of Infectious Diseases & Immunity, Division of Virology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Esther C. de Jong
- Department Of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Erhard van der Vries
- Department Of Infectious Diseases & Immunity, Division of Virology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Coert Margadant
- Molecular Cell Biology Laboratory, Department Of Molecular and Cellular Hemostasis, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Robin van Bruggen
- Department Of Blood Cell Research, Sanquin Research, and Landsteiner Laboratory, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Alexander P.J. Vlaar
- Department of Intensive Care, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Tom Groot Kormelink
- Department Of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Esther N.M. Nolte-‘T Hoen
- Department Of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
18
|
Brands X, Haak BW, Klarenbeek AM, Otto NA, Faber DR, Lutter R, Scicluna BP, Wiersinga WJ, van der Poll T. Concurrent Immune Suppression and Hyperinflammation in Patients With Community-Acquired Pneumonia. Front Immunol 2020; 11:796. [PMID: 32477337 PMCID: PMC7232566 DOI: 10.3389/fimmu.2020.00796] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/07/2020] [Indexed: 12/16/2022] Open
Abstract
Background The nature and timing of the host immune response during infections remain uncertain and most knowledge is derived from critically ill sepsis patients. We aimed to test the hypothesis that community-acquired pneumonia (CAP) is associated with concurrent immune suppression and systemic inflammation. Methods Blood was collected from 79 CAP patients within 24 h after hospitalization and 1 month after discharge; 42 age- and sex-matched subjects without acute infection served as controls. Blood leukocytes were stimulated with lipopolysaccharide (LPS) or Klebsiella pneumoniae, and cytokines were measured in supernatants. Fifteen plasma biomarkers reflective of key host response pathways were compared between CAP patients with the strongest immune suppression (lowest 25% blood leukocyte tumor necrosis factor (TNF)-α production in response to LPS) and those with the least immune suppression (highest 25% of LPS-induced TNF-α production). Results Blood leukocytes of CAP patients (relative to control subjects) showed a reduced capacity to release TNF-α, interleukin (IL)-1β, IL-6 and IL-10 upon stimulation with LPS or K. pneumoniae, with a concurrently enhanced ability to release the anti-inflammatory mediator IL-1 receptor antagonist, irrespective of the presence of sepsis (18.9% of cases). Low (relative to high) TNF-α producers displayed higher plasma levels of biomarkers reflecting systemic inflammation, neutrophil degranulation, endothelial cell activation, a disturbed vascular barrier function and coagulation activation. Conclusion CAP replicates a common feature of immune suppression in sepsis. The coexistence of immune suppression and hyperinflammation in CAP argues against the theory of two distinct phases during the host response to sepsis.
Collapse
Affiliation(s)
- Xanthe Brands
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Bastiaan W Haak
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Augustijn M Klarenbeek
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Natasja A Otto
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Daniël R Faber
- Department of Internal Medicine, BovenIJ Hospital, Amsterdam, Netherlands
| | - René Lutter
- Respiratory Medicine and Experimental Immunology, Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Brendon P Scicluna
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - W Joost Wiersinga
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, Netherlands.,Division of Infectious Diseases, Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, Netherlands.,Division of Infectious Diseases, Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
19
|
Van Eldik LJ, Sawaki L, Bowen K, Laskowitz DT, Noveck RJ, Hauser B, Jordan L, Spears TG, Wu H, Watt K, Raja S, Roy SM, Watterson DM, Guptill JT. First-in-Human Studies of MW01-6-189WH, a Brain-Penetrant, Antineuroinflammatory Small-Molecule Drug Candidate: Phase 1 Safety, Tolerability, Pharmacokinetic, and Pharmacodynamic Studies in Healthy Adult Volunteers. Clin Pharmacol Drug Dev 2020; 10:131-143. [PMID: 32255549 PMCID: PMC7541708 DOI: 10.1002/cpdd.795] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/18/2020] [Indexed: 11/18/2022]
Abstract
MW01‐6‐189WH (MW189) is a novel central nervous system–penetrant small‐molecule drug candidate that selectively attenuates stressor‐induced proinflammatory cytokine overproduction and is efficacious in intracerebral hemorrhage and traumatic brain injury animal models. We report first‐in‐human, randomized, double‐blind, placebo‐controlled phase 1 studies to evaluate the safety, tolerability, and pharmacokinetics (PK) of single and multiple ascending intravenous doses of MW189 in healthy adult volunteers. MW189 was safe and well tolerated in single and multiple doses up to 0.25 mg/kg, with no clinically significant concerns. The most common drug‐related treatment‐emergent adverse event was infusion‐site reactions, likely related to drug solution acidity. No clinically concerning changes were seen in vital signs, electrocardiograms, physical or neurological examinations, or safety laboratory results. PK analysis showed dose‐proportional increases in plasma concentrations of MW189 after single or multiple doses, with approximately linear kinetics and no significant drug accumulation. Steady state was achieved by dose 3 for all dosing cohorts. A pilot pharmacodynamic study administering low‐dose endotoxin to induce a systemic inflammatory response was done to evaluate the effects of a single intravenous dose of MW189 on plasma cytokine levels. MW189 treatment resulted in lower levels of the proinflammatory cytokine TNF‐α and higher levels of the anti‐inflammatory cytokine IL‐10 compared with placebo treatment. The outcomes are consistent with the pharmacological mechanism of MW189. Overall, the safety profile, PK properties, and pharmacodynamic effect support further development of MW189 for patients with acute brain injury.
Collapse
Affiliation(s)
- Linda J. Van Eldik
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Department of NeuroscienceUniversity of KentuckyLexingtonKentuckyUSA
- Spinal Cord and Brain Injury Research CenterUniversity of KentuckyLexingtonKentuckyUSA
| | - Lumy Sawaki
- Department of Physical Medicine & RehabilitationUniversity of KentuckyLexingtonKentuckyUSA
| | - Karen Bowen
- Bluegrass Research Consultants, Inc.VersaillesKentuckyUSA
| | - Daniel T. Laskowitz
- Department of NeurologyDuke UniversityDurhamNorth CarolinaUSA
- Duke Clinical Research InstituteDurhamNorth CarolinaUSA
| | | | - Byron Hauser
- Duke Early Phase Clinical Research UnitDurhamNorth CarolinaUSA
| | - Lynn Jordan
- Duke Early Phase Clinical Research UnitDurhamNorth CarolinaUSA
| | | | - Huali Wu
- Duke Clinical Research InstituteDurhamNorth CarolinaUSA
| | - Kevin Watt
- Duke Clinical Research InstituteDurhamNorth CarolinaUSA
| | - Shruti Raja
- Department of NeurologyDuke UniversityDurhamNorth CarolinaUSA
- Duke Early Phase Clinical Research UnitDurhamNorth CarolinaUSA
| | | | | | - Jeffrey T. Guptill
- Department of NeurologyDuke UniversityDurhamNorth CarolinaUSA
- Duke Clinical Research InstituteDurhamNorth CarolinaUSA
- Duke Early Phase Clinical Research UnitDurhamNorth CarolinaUSA
| |
Collapse
|
20
|
Chang YT, Huang WC, Cheng CC, Ke MW, Tsai JS, Hung YM, Huang NC, Huang MS, Wann SR. Effects of epinephrine on heart rate variability and cytokines in a rat sepsis model. Bosn J Basic Med Sci 2020; 20:88-98. [PMID: 29984678 PMCID: PMC7029199 DOI: 10.17305/bjbms.2018.3565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 06/06/2018] [Indexed: 01/31/2023] Open
Abstract
Catecholamines have both anti-inflammatory and vasoactive properties. A decreased cardiac response to catecholamines has been associated with a high risk of death in sepsis and septic shock. The aim of this study was to investigate the effects of epinephrine (EPI) on heart rate variability (HRV) and autonomic balance, as well as cytokine levels, in a rat sepsis model. Thirty-six male Sprague-Dawley rats were assigned to 4 experimental groups and 2 control groups of 6 rats each. The rats in the experimental groups were inoculated with a lipopolysaccharide (LPS, endotoxin) to establish a sepsis model. Group A received only LPS; group B received LPS, antecedent EPI and the nonselective β-blocker propranolol; group C received LPS and antecedent EPI; and group D received LPS, antecedent EPI and the selective β1-blocker esmolol. One control group received EPI and the other received saline placebo. Heart rate variability (HRV) was analyzed and tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interleukin-1β (IL-1β) levels were measured. Measurements were carried out at baseline, at 0 hour after EPI infusion, and at 0.5, 2, and 4 hours after LPS inoculation. There were significant differences in HRV and cytokine levels between the groups, indicating that LPS infusion caused autonomic imbalance. Antecedent EPI significantly decreased the level of TNF-α in group C compared with group A in which TNF-α level peaked at 2 hours and then declined. Propranolol (group B) but not esmolol (group D) administration resulted in elevated TNF-α levels, comparable to those observed in group A. In conclusion, antecedent administration of EPI in a rat sepsis model inhibits the production of TNF-α possibly via the β2-adrenoceptor.
Collapse
Affiliation(s)
- Yun-Te Chang
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung City; Department of Nursing, Yuh-Ing Junior College of Health Care and Management, Kaohsiung City; Department of Physical Therapy, Shu-Zen Junior College of Medicine and Management, Kaohsiung City; School of Medicine, National Yang-Ming University, Taipei City, Taiwan, Republic of China.
| | - Wei-Chun Huang
- Cardiovascular Medical Center, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan, Republic of China
| | - Chin-Chang Cheng
- Cardiovascular Medical Center, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan, Republic of China
| | - Meng-Wei Ke
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan, Republic of China
| | - Jung-Shun Tsai
- Department of Nursing, Yuh-Ing Junior College of Health Care and Management, Kaohsiung City, Taiwan, Republic of China
| | - Yao-Min Hung
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan, Republic of China
| | - Neng-Chyan Huang
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan, Republic of China
| | - Mu-Shun Huang
- School of Medicine, National Yang-Ming University, Taipei City, Taiwan, Republic of China.
| | - Shue-Ren Wann
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan, Republic of China.
| |
Collapse
|
21
|
Yeager MP, Guyre CA, Sites BD, Collins JE, Pioli PA, Guyre PM. The Stress Hormone Cortisol Enhances Interferon-υ-Mediated Proinflammatory Responses of Human Immune Cells. Anesth Analg 2019; 127:556-563. [PMID: 30028389 DOI: 10.1213/ane.0000000000003481] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Cortisol is a prototypical human stress hormone essential for life, yet the precise role of cortisol in the human stress response to injury or infection is still uncertain. Glucocorticoids (GCs) such as cortisol are widely understood to suppress inflammation and immunity. However, recent research shows that GCs also induce delayed immune effects manifesting as immune stimulation. In this study, we show that cortisol enhances the immune-stimulating effects of a prototypical proinflammatory cytokine, interferon-υ (IFN-υ). We tested the hypothesis that cortisol enhances IFN-υ-mediated proinflammatory responses of human mononuclear phagocytes (monocyte/macrophages [MOs]) stimulated by bacterial endotoxin (lipopolysaccharide [LPS]). METHODS Human MOs were cultured for 18 hours with or without IFN-υ and/or cortisol before LPS stimulation. MO differentiation factors granulocyte-macrophage colony stimulating factor (GM-CSF) or M-CSF were added to separate cultures. We also compared the inflammatory response with an acute, 4-hour MO incubation with IFN-υ plus cortisol and LPS to a delayed 18-hour incubation with cortisol before LPS exposure. MO activation was assessed by interleukin-6 (IL-6) release and by multiplex analysis of pro- and anti-inflammatory soluble mediators. RESULTS After the 18-hour incubation, we observed that cortisol significantly increased LPS-stimulated IL-6 release from IFN-υ-treated undifferentiated MOs. In GM-CSF-pretreated MOs, cortisol increased IFN-υ-mediated IL-6 release by >4-fold and release of the immune stimulant IFN-α2 (IFN-α2) by >3-fold, while suppressing release of the anti-inflammatory mediator, IL-1 receptor antagonist to 15% of control. These results were reversed by either the GC receptor antagonist RU486 or by an IFN-υ receptor type 1 antibody antagonist. Cortisol alone increased expression of the IFN-υ receptor type 1 on undifferentiated and GM-CSF-treated MOs. In contrast, an acute 4-hour incubation of MOs with IFN-υ and cortisol showed classic suppression of the IL-6 response to LPS. CONCLUSIONS These results reveal a surprisingly robust proinflammatory interaction between the human stress response hormone cortisol and the immune activating cytokine IFN-υ. The results support an emerging physiological model with an adaptive role for cortisol, wherein acute release of cortisol suppresses early proinflammatory responses but also primes immune cells for an augmented response to a subsequent immune challenge. These findings have broad clinical implications and provide an experimental framework to examine individual differences, mechanisms, and translational implications of cortisol-enhanced immune responses in humans.
Collapse
Affiliation(s)
- Mark P Yeager
- From the Department of Anesthesiology and Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | | | - Brian D Sites
- Department of Anesthesiology and Orthopedics, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Jane E Collins
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Patricia A Pioli
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Paul M Guyre
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| |
Collapse
|
22
|
Khan HN, Perlee D, Schoenmaker L, van der Meer A, Franitza M, Toliat MR, Nürnberg P, Zwinderman AH, van der Poll T, Scicluna BP. Leukocyte transcriptional signatures dependent on LPS dosage in human endotoxemia. J Leukoc Biol 2019; 106:1153-1160. [PMID: 31280495 PMCID: PMC6852106 DOI: 10.1002/jlb.4a0219-050r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/29/2019] [Accepted: 06/15/2019] [Indexed: 12/18/2022] Open
Abstract
The host immune response is characterized by a complex interplay of signal‐specific cellular transcriptional responses. The magnitude of the immune response is dependent on the strength of the external stimulus. Knowledge on leukocyte transcriptional responses altered in response to different stimulus dosages in man is lacking. Here, we sought to identify leukocyte transcriptional signatures dependent on LPS dose in humans. Healthy human volunteers were administered 1 ng/kg (n = 7), 2 ng/kg (n = 6), or 4 ng/kg (n = 7) LPS intravenously. Blood was collected before (pre‐LPS) and 4 h after LPS administration. Total RNA was analyzed by microarrays and generalized linear models. Pathway analysis was performed by using Ingenuity pathway analysis. Leukocyte transcriptomes altered per LPS dosage were predominantly shared, with 47% common signatures relative to pre‐LPS. A univariate linear model identified a set of 3736 genes that exhibited a dependency on differing LPS dosages. Neutrophil, monocyte, and lymphocyte counts explained 38.9% of the variance in the LPS dose‐dependent gene set. A multivariate linear model including leukocyte composition delineated a set of 295 genes with a dependency on LPS dose. Evaluation of the 295 gene signature in patients with sepsis due to abdominal infections showed significant correlations. Promoter regions of the LPS dose gene set were enriched for YY1, EGR1, ELK1, GABPA, KLF4, and REL transcription factor binding sites. Intravenous injection of 1, 2, or 4 ng/kg LPS was accompanied by both shared and distinct leukocyte transcriptional alterations. These data may assist in assessing the severity of the insult in patients with abdominal sepsis.
Collapse
Affiliation(s)
- Hina N. Khan
- Center for Experimental Molecular MedicineAmsterdam University Medical CentersAcademic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Clinical EpidemiologyBiostatistics and BioinformaticsAmsterdam University Medical Centers, Academic Medical CenterAmsterdamThe Netherlands
| | - Desiree Perlee
- Center for Experimental Molecular MedicineAmsterdam University Medical CentersAcademic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Lieke Schoenmaker
- Center for Experimental Molecular MedicineAmsterdam University Medical CentersAcademic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Anne‐Jan van der Meer
- Center for Experimental Molecular MedicineAmsterdam University Medical CentersAcademic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Marek Franitza
- Cologne Center for Genomics (CCG)University of CologneCologneGermany
| | | | - Peter Nürnberg
- Cologne Center for Genomics (CCG)University of CologneCologneGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Aeilko H. Zwinderman
- Department of Clinical EpidemiologyBiostatistics and BioinformaticsAmsterdam University Medical Centers, Academic Medical CenterAmsterdamThe Netherlands
| | - Tom van der Poll
- Center for Experimental Molecular MedicineAmsterdam University Medical CentersAcademic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
- Division of Infectious DiseasesAmsterdam University Medical CentersAcademic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Brendon P. Scicluna
- Center for Experimental Molecular MedicineAmsterdam University Medical CentersAcademic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Clinical EpidemiologyBiostatistics and BioinformaticsAmsterdam University Medical Centers, Academic Medical CenterAmsterdamThe Netherlands
| |
Collapse
|
23
|
Remote ischaemic preconditioning does not modulate the systemic inflammatory response or renal tubular stress biomarkers after endotoxaemia in healthy human volunteers: a single-centre, mechanistic, randomised controlled trial. Br J Anaesth 2019; 123:177-185. [PMID: 31084985 DOI: 10.1016/j.bja.2019.03.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/15/2019] [Accepted: 03/02/2019] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Remote ischaemic preconditioning (RIPC) consists of repeated cycles of limb ischaemia and reperfusion, which may reduce perioperative myocardial ischaemic damage and kidney injury. We hypothesised that RIPC may be beneficial by attenuating the systemic inflammatory response. We investigated whether RIPC affects the response in humans to bacterial endotoxin (lipopolysaccharide [LPS]) by measuring plasma cytokines and renal cell-cycle arrest mediators, which reflect renal tubular stress. METHODS Healthy male volunteers were randomised to receive either daily RIPC for 6 consecutive days (RIPCmultiple, n=10) plus RIPC during the 40 min preceding i.v. LPS (2 ng kg-1), RIPC only during the 40 min before LPS (RIPCsingle, n=10), or no RIPC preceding LPS (control, n=10). As a surrogate marker of renal tubular stress, the product of urinary concentrations of two cell-cycle arrest markers was calculated (tissue inhibitor of metalloproteinases-2 [TIMP2]*insulin-like growth factor binding protein-7 [IGFBP7]). Data are presented as median (inter-quartile range). RESULTS In both RIPC groups, RIPC alone increased [TIMP2]*[IGFBP7]. LPS administration resulted in fever, flu-like symptoms, and haemodynamic alterations. Plasma cytokine concentrations increased profoundly during endotoxaemia (control group: tumor necrosis factor alpha [TNF-α] from 14 [9-16] pg ml-1 at baseline to 480 [284-709] pg ml-1 at 1.5 h after LPS; interleukin-6 [IL-6] from 4 [4-4] pg ml-1 at baseline to 659 [505-1018] pg ml-1 at 2 h after LPS). LPS administration also increased urinary [TIMP2[*[IGFBP7]. RIPC had no effect on LPS-induced cytokine release or [TIMP2]*[IGFBP7]. CONCLUSIONS RIPC neither modulated systemic cytokine release nor attenuated inflammation-induced tubular stress after LPS. However, RIPC alone induced renal markers of cell-cycle arrest. CLINICAL TRIAL REGISTRATION NCT02602977.
Collapse
|
24
|
TLR4 counteracts BVRA signaling in human leukocytes via differential regulation of AMPK, mTORC1 and mTORC2. Sci Rep 2019; 9:7020. [PMID: 31065010 PMCID: PMC6504875 DOI: 10.1038/s41598-019-43347-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/16/2019] [Indexed: 12/30/2022] Open
Abstract
TLR4 is implicated in diseases associated with chronic low-grade inflammation, yet homeostatic signaling mechanisms that prevent and/or are affected by chronic TLR4 activation are largely uncharacterized. We recently reported that LPS/TLR4 activates in human leukocytes signaling intermediates (SI), abbreviated TLR4-SI, which include mTORC1-specific effectors and targets, and that leukocytes of patients with T2D or after cardiopulmonary bypass (CPB) expressed similar SI. Extending these previous findings, here we show that TLR4-SI expression post-CPB was associated with low serum bilirubin and reduced preoperative expression of biliverdin reductase A (BVRA), the enzyme that converts biliverdin to bilirubin, in patient’s leukocytes. Biliverdin inhibited TLR4 signaling in leukocytes and triggered phosphorylation of mTORC2-specific targets, including Akt, PKCζ, AMPKα-LKB1-TSC1/2, and their association with BVRA. Torin, PP242, and a PKCζ inhibitory peptide, but not rapamycin, prevented these biliverdin-induced responses and TLR4 inhibition. In contrast, LPS/TLR4 triggered decreases in BVRA, AMPKα and PKCζ expression, and an increase in haptoglobin, a heme binding protein, in leukocytes in vivo and in vitro, indicating that activated TLR4 may suppress biliverdin/BVRA signaling. Significantly, compared to non-diabetics, BVRA and PKCζ expression was low and haptoglobin was high in T2D patients leukocytes. Sustained TLR4 activation may deregulate homeostatic anti-inflammatory BVRA/mTORC2 signaling and thereby contribute to chronic inflammatory diseases.
Collapse
|
25
|
Allergic Airway-Induced Hypersensitivity Is Attenuated by Bergapten in Murine Models of Inflammation. Adv Pharmacol Sci 2019; 2019:6097349. [PMID: 30863445 PMCID: PMC6378071 DOI: 10.1155/2019/6097349] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 12/13/2018] [Indexed: 12/02/2022] Open
Abstract
Bergapten (5-methoxypsoralen, 5-MOP) is a plant-derived furocoumarin with demonstrated anti-inflammatory action. The present study investigated its effects on allergic inflammation in two related pathways of mast cell degranulation. Compound 48/80 and lipopolysaccharide (LPS) were used to activate the IgE-independent pathway while bovine serum albumin (BSA) was used as allergen for the IgE-dependent pathway. The modulatory effect of bergapten on mast cell degranulation, neutrophil extravasation, protein concentration, lung histopathology, and oxidative stress was assessed. Bergapten at 10, 30, and 100 μg/ml for 15 min stabilized mast cells in rat mesenteric tissue from disruption in vitro and when administered in vivo at 3, 10, and 30 mg kg−1 for 1 h protected mice from fatal anaphylaxis induced by compound 48/80. Similarly, treatment of LPS-challenged mice with bergapten (3, 10, and 30 mg kg−1) for 24 h significantly decreased neutrophil infiltration into bronchoalveolar lavage fluid, mean protein concentration, and inflammatory cell infiltration of pulmonary tissues when compared to the saline-treated LPS-challenged control. In addition, lung histology of the bergapten-treated LPS-challenged mice showed significantly less oedema, congestion, and alveolar septa thickening when compared to the saline-treated LPS-challenged disease control. LPS-induced oxidative stress was significantly reduced through increased tissue activities of catalase and superoxide dismutase and reduced malondialdehyde levels on treatment with bergapten. In the triple antigen-induced active anaphylaxis, daily administration of bergapten at 3, 10, and 30 mg kg−1 for 10 days, respectively, protected previously sensitized and challenged mice against anaphylactic shock. Overall, our study demonstrates the ability of bergapten to attenuate allergic airway-induced hypersensitivity in murine models of inflammation, suggesting its possible therapeutic benefit in this condition.
Collapse
|
26
|
Perlee D, van Vught LA, Scicluna BP, Maag A, Lutter R, Kemper EM, van ‘t Veer C, Punchard MA, González J, Richard MP, Dalemans W, Lombardo E, de Vos AF, van der Poll T. Intravenous Infusion of Human Adipose Mesenchymal Stem Cells Modifies the Host Response to Lipopolysaccharide in Humans: A Randomized, Single-Blind, Parallel Group, Placebo Controlled Trial. Stem Cells 2018; 36:1778-1788. [DOI: 10.1002/stem.2891] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/06/2018] [Accepted: 07/02/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Desiree Perlee
- Center of Experimental & Molecular Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Lonneke A. van Vught
- Center of Experimental & Molecular Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Brendon P. Scicluna
- Center of Experimental & Molecular Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
- Department of Clinical Epidemiology and Biostatistics, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Anja Maag
- Center of Experimental & Molecular Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - René Lutter
- Department of Experimental Immunology & Respiratory Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Elles M. Kemper
- Department of Pharmacy, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Cornelis van ‘t Veer
- Center of Experimental & Molecular Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | | | | | | | | | | | - Alex F. de Vos
- Center of Experimental & Molecular Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Tom van der Poll
- Center of Experimental & Molecular Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
- Division of Infectious Diseases, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| |
Collapse
|
27
|
Experimental human endotoxemia as a model of systemic inflammation. Biochimie 2018; 159:99-106. [PMID: 29936295 DOI: 10.1016/j.biochi.2018.06.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/20/2018] [Indexed: 12/31/2022]
Abstract
Systemic inflammation plays a pivotal role in a multitude of conditions, including sepsis, trauma, major surgery and burns. However, comprehensive analysis of the pathophysiology underlying this systemic inflammatory response is greatly complicated by variations in the immune response observed in critically ill patients, which is a result of inter-individual differences in comorbidity, comedication, source of infection, causative pathogen, and onset of the inflammatory response. During experimental human endotoxemia, human subjects are challenged with purified endotoxin (lipopolysaccharide) intravenously which induces a short-lived, well-tolerated and controlled systemic inflammatory response, similar to that observed during sepsis. The human endotoxemia model can be conducted in a highly standardized and reproducible manner, using a carefully selected homogenous study population. As such, the experimental human endotoxemia model does not share the aforementioned clinical limitations and enables us to investigate both the mechanisms of systemic inflammation, as well as to evaluate novel (pharmacological) interventions in humans in vivo. The present review provides a detailed overview of the various designs, organ-specific changes, and strengths and limitations of the experimental human endotoxemia model, with the main focus on its use as a translational model for sepsis research.
Collapse
|
28
|
Vasopressors Do Not Influence Cerebral Critical Closing Pressure During Systemic Inflammation Evoked by Experimental Endotoxemia and Sepsis in Humans. Shock 2018; 49:529-535. [DOI: 10.1097/shk.0000000000001003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
29
|
Rao RT, Scherholz ML, Hartmanshenn C, Bae SA, Androulakis IP. On the analysis of complex biological supply chains: From Process Systems Engineering to Quantitative Systems Pharmacology. Comput Chem Eng 2017; 107:100-110. [PMID: 29353945 DOI: 10.1016/j.compchemeng.2017.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The use of models in biology has become particularly relevant as it enables investigators to develop a mechanistic framework for understanding the operating principles of living systems as well as in quantitatively predicting their response to both pathological perturbations and pharmacological interventions. This application has resulted in a synergistic convergence of systems biology and pharmacokinetic-pharmacodynamic modeling techniques that has led to the emergence of quantitative systems pharmacology (QSP). In this review, we discuss how the foundational principles of chemical process systems engineering inform the progressive development of more physiologically-based systems biology models.
Collapse
Affiliation(s)
- Rohit T Rao
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854
| | - Megerle L Scherholz
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854
| | - Clara Hartmanshenn
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854
| | - Seul-A Bae
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854
| | - Ioannis P Androulakis
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854.,Department of Biomedical Engineering, Rutgers The State University of New Jersey, 599 Taylor Road, Piscataway, NJ 08854
| |
Collapse
|
30
|
Paardekooper LM, van den Bogaart G, Kox M, Dingjan I, Neerincx AH, Bendix MB, Beest MT, Harren FJM, Risby T, Pickkers P, Marczin N, Cristescu SM. Ethylene, an early marker of systemic inflammation in humans. Sci Rep 2017; 7:6889. [PMID: 28761087 PMCID: PMC5537290 DOI: 10.1038/s41598-017-05930-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 06/06/2017] [Indexed: 11/09/2022] Open
Abstract
Ethylene is a major plant hormone mediating developmental processes and stress responses to stimuli such as infection. We show here that ethylene is also produced during systemic inflammation in humans and is released in exhaled breath. Traces of ethylene were detected by laser spectroscopy both in vitro in isolated blood leukocytes exposed to bacterial lipopolysaccharide (LPS) as well as in vivo following LPS administration in healthy volunteers. Exposure to LPS triggers formation of ethylene as a product of lipid peroxidation induced by the respiratory burst. In humans, ethylene was detected prior to the increase of blood levels of inflammatory cytokines and stress-related hormones. Our results highlight that ethylene release is an early and integral component of in vivo lipid peroxidation with important clinical implications as a breath biomarker of bacterial infection.
Collapse
Affiliation(s)
- Laurent M Paardekooper
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Geert van den Bogaart
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Matthijs Kox
- Intensive Care Medicine, Nijmegen Institute for Infection, Inflammation and Immunity, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ilse Dingjan
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anne H Neerincx
- Department of Molecular and Laser Physics, Institute of Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| | - Maura B Bendix
- Department of Molecular and Laser Physics, Institute of Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| | - Martin Ter Beest
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frans J M Harren
- Department of Molecular and Laser Physics, Institute of Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| | - Terence Risby
- Department of Environmental Health Sciences, Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Peter Pickkers
- Intensive Care Medicine, Nijmegen Institute for Infection, Inflammation and Immunity, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nandor Marczin
- Department of Anaesthesia, Royal Brompton and Harefield NHS Foundation Trust, Harefield, UK
- Section of Anaesthesia, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Simona M Cristescu
- Department of Molecular and Laser Physics, Institute of Molecules and Materials, Radboud University, Nijmegen, The Netherlands.
| |
Collapse
|
31
|
Human CD62Ldim neutrophils identified as a separate subset by proteome profiling and in vivo pulse-chase labeling. Blood 2017; 129:3476-3485. [DOI: 10.1182/blood-2016-07-727669] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 05/10/2017] [Indexed: 01/08/2023] Open
Abstract
Abstract
During acute inflammation, 3 neutrophil subsets are found in the blood: neutrophils with a conventional segmented nucleus, neutrophils with a banded nucleus, and T-cell–suppressing CD62Ldim neutrophils with a high number of nuclear lobes. In this study, we compared the in vivo kinetics and proteomes of banded, mature, and hypersegmented neutrophils to determine whether these cell types represent truly different neutrophil subsets or reflect changes induced by lipopolysaccharide (LPS) activation. Using in vivo pulse-chase labeling of neutrophil DNA with 6,6-2H2-glucose, we found that 2H-labeled banded neutrophils appeared much earlier in blood than labeled CD62Ldim and segmented neutrophils, which shared similar label kinetics. Comparison of the proteomes by cluster analysis revealed that CD62Ldim neutrophils were clearly separate from conventional segmented neutrophils despite having similar kinetics in peripheral blood. Interestingly, the conventional segmented cells were more related at a proteome level to banded cells despite a 2-day difference in maturation time. The differences between CD62Ldim and mature neutrophils are unlikely to have been a direct result of LPS-induced activation, because of the extremely low transcriptional capacity of CD62Ldim neutrophils and the fact that neutrophils do not directly respond to the low dose of LPS used in the study (2 ng/kg body weight). Therefore, we propose CD62Ldim neutrophils are a truly separate neutrophil subset that is recruited to the bloodstream in response to acute inflammation. This trial was registered at www.clinicaltrials.gov as #NCT01766414.
Collapse
|
32
|
Kamisoglu K, Acevedo A, Almon RR, Coyle S, Corbett S, Dubois DC, Nguyen TT, Jusko WJ, Androulakis IP. Understanding Physiology in the Continuum: Integration of Information from Multiple - Omics Levels. Front Pharmacol 2017; 8:91. [PMID: 28289389 PMCID: PMC5327699 DOI: 10.3389/fphar.2017.00091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 02/13/2017] [Indexed: 01/18/2023] Open
Abstract
In this paper, we discuss approaches for integrating biological information reflecting diverse physiologic levels. In particular, we explore statistical and model-based methods for integrating transcriptomic, proteomic and metabolomics data. Our case studies reflect responses to a systemic inflammatory stimulus and in response to an anti-inflammatory treatment. Our paper serves partly as a review of existing methods and partly as a means to demonstrate, using case studies related to human endotoxemia and response to methylprednisolone (MPL) treatment, how specific questions may require specific methods, thus emphasizing the non-uniqueness of the approaches. Finally, we explore novel ways for integrating -omics information with PKPD models, toward the development of more integrated pharmacology models.
Collapse
Affiliation(s)
- Kubra Kamisoglu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo NY, USA
| | - Alison Acevedo
- Department of Biomedical Engineering, Rutgers University, Piscataway NJ, USA
| | - Richard R Almon
- Department of Biological Sciences, University at Buffalo, Buffalo NY, USA
| | - Susette Coyle
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick NJ, USA
| | - Siobhan Corbett
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick NJ, USA
| | - Debra C Dubois
- Department of Biological Sciences, University at Buffalo, Buffalo NY, USA
| | - Tung T Nguyen
- BioMaPS Institute for Quantitative Biology, Rutgers University, Piscataway NJ, USA
| | - William J Jusko
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo NY, USA
| | - Ioannis P Androulakis
- Department of Biomedical Engineering, Rutgers University, PiscatawayNJ, USA; Department of Chemical Engineering, Rutgers University, PiscatawayNJ, USA
| |
Collapse
|
33
|
Regulation of Lipolysis and Adipose Tissue Signaling during Acute Endotoxin-Induced Inflammation: A Human Randomized Crossover Trial. PLoS One 2016; 11:e0162167. [PMID: 27627109 PMCID: PMC5023116 DOI: 10.1371/journal.pone.0162167] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 08/17/2016] [Indexed: 01/19/2023] Open
Abstract
Background Lipolysis is accelerated during the acute phase of inflammation, a process being regulated by pro-inflammatory cytokines (e.g. TNF-α), stress-hormones, and insulin. The intracellular mechanisms remain elusive and we therefore measured pro- and anti-lipolytic signaling pathways in adipocytes after in vivo endotoxin exposure. Methods Eight healthy, lean, male subjects were investigated using a randomized cross over trial with two interventions: i) bolus injection of saline (Placebo) and ii) bolus injection of lipopolysaccharide endotoxin (LPS). A 3H-palmitate tracer was used to measure palmitate rate of appearance (Rapalmitate) and indirect calorimetry was performed to measure energy expenditures and lipid oxidation rates. A subcutaneous abdominal fat biopsy was obtained during both interventions and subjected to western blotting and qPCR quantifications. Results LPS caused a mean increase in serum free fatty acids (FFA) concentrations of 90% (CI-95%: 37–142, p = 0.005), a median increase in Rapalmitate of 117% (CI-95%: 77–166, p<0.001), a mean increase in lipid oxidation of 49% (CI-95%: 1–96, p = 0.047), and a median increase in energy expenditure of 28% (CI-95%: 16–42, p = 0.001) compared with Placebo. These effects were associated with increased phosphorylation of hormone sensitive lipase (pHSL) at ser650 in adipose tissue (p = 0.03), a trend towards elevated pHSL at ser552 (p = 0.09) and cAMP-dependent protein kinase A (PKA) phosphorylation of perilipin 1 (PLIN1) (p = 0.09). Phosphatase and tensin homolog (PTEN) also tended to increase (p = 0.08) while phosphorylation of Akt at Thr308 tended to decrease (p = 0.09) during LPS compared with Placebo. There was no difference between protein or mRNA expression of ATGL, G0S2, and CGI-58. Conclusion LPS stimulated lipolysis in adipose tissue and is associated with increased pHSL and signs of increased PLIN1 phosphorylation combined with a trend toward decreased insulin signaling. The combination of these mechanisms appear to be the driving forces behind the increased lipolysis observed in the early stages of acute inflammation and sepsis. Trial Registration ClinicalTrials.gov NCT01705782
Collapse
|
34
|
Alvarez SM, Katsamanis Karavidas M, Coyle SM, Lu SE, Macor M, Oikawa LO, Lehrer PM, Calvano SE, Lowry SF. Low-dose steroid alters in vivo endotoxin-induced systemic inflammation but does not influence autonomic dysfunction. ACTA ACUST UNITED AC 2016; 13:358-68. [DOI: 10.1177/0968051907086465] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Severe injury and infection are associated with autonomic dysfunction. Diminished heart rate variability (HRV) is also observed as a component of autonomic dysfunction and is induced by endotoxin administration to healthy subjects. It is established that low-dose glucocorticoid administration diminishes the systemic inflammatory manifestations of endotoxinemia but the influence of this anti-inflammatory intervention on overall autonomic dysfunction and HRV responses to endotoxin is unknown. This study was designed to assess the influence of a low-dose hydrocortisone infusion upon endotoxin-elicited systemic inflammatory responses including phenotypic features, cytokine production, and parameters of HRV. Of 19 subjects studied, nine received a continuous infusion of hydrocortisone (3 µg/kg/min continuously over 6 h) prior to intravenous administration of Escherichia coli endotoxin (2 ng/kg, CC-RE, Lot #2) while 10 healthy subjects received only the endotoxin after a 6-h period of saline control infusion. Serial determinations of vital signs, heart rate variability assessments, and cytokine levels were obtained over the subsequent 24 h. Prior cortisol infusion diminished the peak TNF-α ( P < 0.01) and IL-6 ( P < 0.0001) responses after endotoxin challenge, as compared to saline infusion controls and diminished the peak core temperature response to endotoxin ( P < 0.01). In contrast to the influence of cortisol on the above parameters of systemic inflammation, the significant endotoxin-induced decreases in HRV time and frequency domains were not influenced by prior hydrocortisone treatment. Hence, alterations in autonomic dysfunction occur despite hydrocortisone attenuation of other traditional systemic manifestations of endotoxinemia. The maintenance or restoration of autonomic balance is not influenced by glucocorticoid administration.
Collapse
Affiliation(s)
- Sonia M. Alvarez
- Division of Surgical Sciences, UMDNJ-Robert Wood Johnson Medical School, Now Brunswick, New Jersey, USA
| | | | - Susette M. Coyle
- Division of Surgical Sciences, UMDNJ-Robert Wood Johnson Medical School, Now Brunswick, New Jersey, USA
| | - Shou-En Lu
- Department of Biostatistics, UMDNJ-School of Public Health, New Brunswick, New Jersey, USA
| | - Marie Macor
- Division of Surgical Sciences, UMDNJ-Robert Wood Johnson Medical School, Now Brunswick, New Jersey, USA
| | - Leo O. Oikawa
- Department of Psychiatry, UMDNJ-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Paul M. Lehrer
- Department of Psychiatry, UMDNJ-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Steve E. Calvano
- Division of Surgical Sciences, UMDNJ-Robert Wood Johnson Medical School, Now Brunswick, New Jersey, USA
| | - Stephen F. Lowry
- Division of Surgical Sciences, UMDNJ-Robert Wood Johnson Medical School, Now Brunswick, New Jersey, USA,
| |
Collapse
|
35
|
Bahador M, Cross AS. Review: From therapy to experimental model: a hundred years of endotoxin administration to human subjects. ACTA ACUST UNITED AC 2016; 13:251-79. [DOI: 10.1177/0968051907085986] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This article is a review of studies in which endotoxin has been administered to human subjects for experimental purposes. Data are presented in tabular form so the reader can better appreciate the objectives of individual studies. Although the original intention was to focus on the adverse events associated with these studies, unexpected serious adverse events rarely have been reported.
Collapse
Affiliation(s)
- Marjan Bahador
- Center for Vaccine Development and Division of Infectious Diseases, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA,
| | - Alan S. Cross
- Center for Vaccine Development and Division of Infectious Diseases, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
36
|
Janum S, Nielsen ST, Werner MU, Mehlsen J, Kehlet H, Møller K. Pain perception in healthy volunteers: effect of repeated exposure to experimental systemic inflammation. Innate Immun 2016; 22:546-56. [PMID: 27554053 DOI: 10.1177/1753425916663638] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 07/08/2016] [Indexed: 11/15/2022] Open
Abstract
We aimed to study the relationship between pain perception and cytokine release during systemic inflammation. We present a randomized crossover trial in healthy volunteers (n = 17) in 37 individual trials. Systemic inflammation was induced by an i.v. bolus of Escherichia coli LPS (2 ng/kg) on two separate trial days, with or without a nicotine patch applied 10 h previously. Pain perception at baseline, and 2 and 6 h after LPS was assessed by pressure algometry and tonic heat stimulation at an increasing temperature (45-48℃) during both trials. Compared with baseline, pain pressure threshold was reduced 2 and 6 h after LPS, while heat pain perception was accentuated at all testing temperatures after 2 but not 6 h. The magnitude of changes in pain perception did not correlate to cytokine release. No effect of transdermal nicotine or training status was observed. In conclusion, LPS administration in healthy human volunteers leads to reduction in pain pressure threshold and an increase in pain perception to heat stimuli, supporting a relationship between acute systemic inflammation and pain perception.
Collapse
Affiliation(s)
- Susanne Janum
- Center of Inflammation and Metabolism, CIM 7641, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark Department of Anesthesiology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Signe T Nielsen
- Center of Inflammation and Metabolism, CIM 7641, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Mads U Werner
- Multidisciplinary Pain Center, Department of Neuroanesthesiology, Rigshospitalet, Copenhagen, Denmark
| | - Jesper Mehlsen
- Coordinating Research Centre, Frederiksberg Hospital, Frederiksberg, Denmark
| | - Henrik Kehlet
- Institute of Surgical Pathophysiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Kirsten Møller
- Department of Neuroanesthesiology, Rigshospitalet, University of Copenhagen, Denmark
| |
Collapse
|
37
|
Zhang Z, Amorosa LF, Coyle SM, Macor MA, Birnbaum MJ, Lee LY, Haimovich B. Insulin-Dependent Regulation of mTORC2-Akt-FoxO Suppresses TLR4 Signaling in Human Leukocytes: Relevance to Type 2 Diabetes. Diabetes 2016; 65:2224-34. [PMID: 27207509 DOI: 10.2337/db16-0027] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 05/02/2016] [Indexed: 11/13/2022]
Abstract
Leukocyte signaling in patients with systemic insulin resistance is largely unexplored. We recently discovered the presence of multiple Toll-like receptor 4 (TLR4) signaling intermediates in leukocytes from patients with type 2 diabetes or acute insulin resistance associated with cardiopulmonary bypass surgery. We extend this work to show that in addition to matrix metalloproteinase 9, hypoxia-inducible factor 1α, and cleaved AMPKα, patient leukocytes also express IRS-1 phosphorylated on Ser(312), Akt phosphorylated on Thr(308), and elevated TLR4 expression. Similar signaling intermediates were detected in leukocytes and neutrophils treated with lipopolysaccharide (LPS), a ligand of TLR4, in vitro. In contrast, insulin, but not LPS, induced mammalian target of rapamycin complex 2 (mTORC2)-dependent phosphorylation of Akt on Ser(473) and FoxO1/O3a on Thr(24/32) in leukocytes and neutrophils. Insulin suppressed LPS-induced responses in a dose- and time-dependent manner. AS1842856, a FoxO1 inhibitor, also suppressed TLR4 signaling. We propose that insulin is a homeostatic regulator of leukocyte responses to LPS/TLR4 and that the signaling intermediates expressed in leukocytes of patients with type 2 diabetes indicate TLR4 signaling dominance and deficient insulin signaling. The data suggest that insulin suppresses LPS/TLR4 signals in leukocytes through the mTORC2-Akt-FoxO signaling axis. Better understanding of leukocyte signaling in patients with type 2 diabetes may shed new light on disease causation and progression.
Collapse
Affiliation(s)
- Zhiyong Zhang
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Louis F Amorosa
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Susette M Coyle
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Marie A Macor
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Morris J Birnbaum
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Leonard Y Lee
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Beatrice Haimovich
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| |
Collapse
|
38
|
Pierre K, Schlesinger N, Androulakis IP. The role of the hypothalamic-pituitary-adrenal axis in modulating seasonal changes in immunity. Physiol Genomics 2016; 48:719-738. [PMID: 27341833 DOI: 10.1152/physiolgenomics.00006.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 06/23/2016] [Indexed: 12/21/2022] Open
Abstract
Seasonal changes in environmental conditions are accompanied by significant adjustment of multiple biological processes. In temperate regions, the day fraction, or photoperiod, is a robust environmental cue that synchronizes seasonal variations in neuroendocrine and metabolic function. In this work, we propose a semimechanistic mathematical model that considers the influence of seasonal photoperiod changes as well as cellular and molecular adaptations to investigate the seasonality of immune function. Our model predicts that the circadian rhythms of cortisol, our proinflammatory mediator, and its receptor exhibit seasonal differences in amplitude and phase, oscillating at higher amplitudes in the winter season with peak times occurring later in the day. Furthermore, the reduced photoperiod of winter coupled with seasonal alterations in physiological activity induces a more exacerbated immune response to acute stress, simulated in our studies as the administration of an acute dose of endotoxin. Our findings are therefore in accordance with experimental data that reflect the predominance of a proinflammatory state during the winter months. These changes in circadian rhythm dynamics may play a significant role in the seasonality of disease incidence and regulate the diurnal and seasonal variation of disease symptom severity.
Collapse
Affiliation(s)
- Kamau Pierre
- Biomedical Engineering Department, Rutgers University, Piscataway, New Jersey
| | - Naomi Schlesinger
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Ioannis P Androulakis
- Biomedical Engineering Department, Rutgers University, Piscataway, New Jersey; Chemical and Biochemical Engineering Department, Rutgers University, Piscataway, New Jersey; and Department of Surgery, Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey
| |
Collapse
|
39
|
Abstract
OBJECTIVE To evaluate the prevalence and time course of systemic endotoxemia following severe multiple trauma, to define its risk factors, and to explore the correlation between post-trauma endotoxemia and organ dysfunction. DESIGN Prospective single-center cohort study. SETTING Emergency department and ICU of adult tertiary care level I trauma center. PATIENTS Forty-eight severely injured (Injury Severity Score ≥ 16) patients, admitted to ICU within 24 hours of injury. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Endotoxemia was not evident on initial presentation, but developed subsequently in 75% of patients, even in the absence of Gram-negative infection. Nonsurviving patients had higher endotoxin levels than survivors on day 1 (endotoxemia, 0.48 vs 0.28; p = 0.048). Shock at admission, or surgery within the first 48 hours after trauma, was associated with higher endotoxin levels and predicted subsequent maximal endotoxemia, after adjusting for other significant covariates. Maximal endotoxemia levels were higher in patients who developed organ dysfunction, reflected in a cumulative Multiple Organ Dysfunction Score greater than 25, and patients with an intermediate endotoxemia level (≥ 0.4) had more cardiovascular dysfunction. CONCLUSIONS It is the first study to detect increasing levels of endotoxemia following multiple trauma. Shock and early surgery predict the development of endotoxemia; endotoxemia is particularly associated with cardiovascular dysfunction. However, Gram-negative infections are uncommon in these patients, suggesting that the gastrointestinal tract is the dominant reservoir of endotoxin. Endotoxin may be an appropriate therapeutic target in patients who have sustained severe multiple trauma.
Collapse
|
40
|
Fullerton JN, Segre E, De Maeyer RPH, Maini AAN, Gilroy DW. Intravenous Endotoxin Challenge in Healthy Humans: An Experimental Platform to Investigate and Modulate Systemic Inflammation. J Vis Exp 2016. [PMID: 27213711 PMCID: PMC4942172 DOI: 10.3791/53913] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Activation of inflammatory pathways represents a central mechanism in multiple disease states both acute and chronic. Triggered via either pathogen or tissue damage-associated molecular motifs, common biochemical pathways lead to conserved yet variable physiological and immunological alterations. Dissection and delineation of the determinants and mechanisms underlying phenotypic variance in response is expected to yield novel therapeutic advances. Intravenous (IV) administration of endotoxin (gram-negative bacterial lipopolysaccharide), a specific Toll-like receptor 4 agonist, represents an in vivo model of systemic inflammation in man. National Institutes for Health Clinical Center Reference Endotoxin (CCRE, Escherichia coli O:113:H10:K negative) is employed to reliably and reproducibly generate vascular, hematological, endocrine, immunological and organ-specific functional effects that parallel, to varying degrees, those seen in the early stages of pathological states. Alteration of dose (0.06 - 4 ng/kg) and time-scale of exposure (bolus vs. infusion) allows replication of either acute or chronic inflammation and a range of severity to be elicited, with higher doses (2 - 4 ng/kg) frequently being used to create a 'sepsis-like' state. Established and novel medicinal compounds may additionally be administered prior to or post endotoxin exposure to appreciate their effect on the inflammatory cascade. Despite limitations in scope and generalizability, human IV endotoxin challenge offers a unique platform to gain mechanistic insights into inducible physiological responses and inflammatory pathways. Rationally employed it may aid translation of this knowledge into therapeutic innovations.
Collapse
Affiliation(s)
- James N Fullerton
- Centre for Clinical Pharmacology, Division of Medicine, University College London;
| | - Elisabetta Segre
- Centre for Clinical Pharmacology, Division of Medicine, University College London
| | - Roel P H De Maeyer
- Centre for Clinical Pharmacology, Division of Medicine, University College London
| | - Alexander A N Maini
- Centre for Clinical Pharmacology, Division of Medicine, University College London
| | - Derek W Gilroy
- Centre for Clinical Pharmacology, Division of Medicine, University College London
| |
Collapse
|
41
|
Plovsing RR, Berg RMG, Munthe-Fog L, Konge L, Iversen M, Møller K, Garred P. Alveolar recruitment of ficolin-3 in response to acute pulmonary inflammation in humans. Immunobiology 2016; 221:690-7. [PMID: 26868430 DOI: 10.1016/j.imbio.2015.11.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 11/21/2015] [Accepted: 11/22/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND Ficolins serve as soluble recognition molecules in the lectin pathway of complement. They are known to participate in the systemic host-response to infection but their role in local pulmonary defence is still incompletely understood. The purpose of this study was to clarify whether acute lung and systemic inflammation induce recruitment of lectins in humans. METHODS Fifteen healthy volunteers received LPS intravenously (IV) or in a lung subsegment on two different occasions. Volunteers were evaluated by consecutive blood samples and by bronchoalveolar lavage 2, 4, 6, 8, or 24h after LPS (n=3 in all groups), and gene expression patterns and protein levels of mannose-binding lectin (MBL) and ficolins were determined. RESULTS Endobronchial LPS was associated with an increase in alveolar ficolin-3 and MBL levels (p<0.04 and p<0.001, respectively). IV LPS elicited a pronounced acute phase response with an increase in CRP (p<0.001) and plasma ficolin-1 protein levels (p<0.001), whereas no changes were observed in ficolin-1 gene expression patterns (p=0.11) or plasma protein levels of MBL, ficolin-2, or ficolin-3. CONCLUSIONS LPS induces a tissue-specific recruitment of ficolin-3 and ficolin-1 in the lung and systemic compartment, respectively, suggesting an important role of distinct lectin complement pathway initiators in the local pulmonary and systemic host defence.
Collapse
Affiliation(s)
- Ronni R Plovsing
- Department of Intensive Care, University Hospital Rigshospitalet, Copenhagen Ø, Denmark.
| | - Ronan M G Berg
- Department of Clinical Physiology, Nuclear Medicine & PET, University Hospital Rigshospitalet, Copenhagen Ø, Denmark; Centre of Inflammation and Metabolism, University Hospital Rigshospitalet, Copenhagen Ø, Denmark
| | - Lea Munthe-Fog
- Department of Clinical Immunology, Laboratory of Molecular Medicine, University Hospital Rigshospitalet, Copenhagen Ø, Denmark
| | - Lars Konge
- Centre for Clinical Education, University of Copenhagen and the Capital Region of Denmark, Copenhagen, Denmark
| | - Martin Iversen
- Department of Lung Transplantation, University Hospital Rigshospitalet, Copenhagen Ø, Denmark
| | - Kirsten Møller
- Department of Neuroanaesthesiology, Neurointensive Care Unit, University Hospital Rigshospitalet, Copenhagen Ø, Denmark; Centre of Inflammation and Metabolism, University Hospital Rigshospitalet, Copenhagen Ø, Denmark
| | - Peter Garred
- Department of Clinical Immunology, Laboratory of Molecular Medicine, University Hospital Rigshospitalet, Copenhagen Ø, Denmark
| |
Collapse
|
42
|
Mooberry MJ, Bradford R, Hobl EL, Lin FC, Jilma B, Key NS. Procoagulant microparticles promote coagulation in a factor XI-dependent manner in human endotoxemia. J Thromb Haemost 2016; 14:1031-42. [PMID: 26857798 PMCID: PMC4870121 DOI: 10.1111/jth.13285] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Indexed: 11/30/2022]
Abstract
UNLABELLED Essentials The procoagulant effects of microparticles (MPs) on coagulation in endotoxemia are not known. MPs from endotoxemia volunteers were evaluated for procoagulant activity in a plasma milieu. MPs from endotoxemia volunteers shortened clotting times and enhanced thrombin generation. MP procoagulant effects were mediated in a factor XI-dependent manner. SUMMARY Background Human endotoxemia is characterized by acute inflammation and activation of coagulation, as well as increased numbers of circulating microparticles (MPs). Whether these MPs directly promote coagulation and through which pathway their actions are mediated, however, has not been fully explored. Objectives In this study, we aimed to further characterize endotoxin-induced MPs and their procoagulant properties using several approaches. Methods Enumeration and characterization of MPs were performed using a new-generation flow cytometer. Relative contributions of the extrinsic and intrinsic pathways in MP-mediated procoagulant activity were assessed using plasmas deficient in factor (F) VII or FXI or with blocking antibodies to tissue factor (TF) or FXIa. Results Total MPs and platelet MPs were significantly elevated in plasma at 6 h after infusion of endotoxin in healthy human subjects. MPs isolated from plasma following endotoxin infusion also demonstrated increased TF activity in a reconstituted buffer system. When added to recalcified platelet-poor plasma, these MPs also promoted coagulation, as judged by a decreased clotting time with shortening of the lag time and time to peak thrombin using calibrated automated thrombography (CAT). However, the use of FVII-deficient plasma or blocking antibody to TF did not inhibit these procoagulant effects. In contrast, plasma clotting time was prolonged in FXI-deficient plasma and a blocking antibody to FXIa inhibited all MP-mediated parameters in the CAT assay. Conclusions The initiation of coagulation by cellular TF in endotoxemia is in contrast to (and presumably complemented by) the intrinsic pathway-mediated procoagulant effects of circulating MPs.
Collapse
Affiliation(s)
- M J Mooberry
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - R Bradford
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - E L Hobl
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - F C Lin
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - B Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - N S Key
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
43
|
Zhang Z, Amorosa LF, Coyle SM, Macor MA, Lubitz SE, Carson JL, Birnbaum MJ, Lee LY, Haimovich B. Proteolytic Cleavage of AMPKα and Intracellular MMP9 Expression Are Both Required for TLR4-Mediated mTORC1 Activation and HIF-1α Expression in Leukocytes. THE JOURNAL OF IMMUNOLOGY 2015; 195:2452-60. [PMID: 26232429 DOI: 10.4049/jimmunol.1500944] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 07/05/2015] [Indexed: 12/26/2022]
Abstract
LPS-induced TLR4 activation alters cellular bioenergetics and triggers proteolytic cleavage of AMPKα and HIF-1α expression in leukocytes. In human leukocytes, and more specifically neutrophils, AMPKα cleavage yields 55- and 35-kDa protein fragments. In this study, we address the mechanism by which AMPKα is cleaved and its relevance to human health. Our data indicate that AMPKα cleavage is linked to MMP9 expression and that both are required for mammalian target of rapamycin complex-1 and S6K1 activation and HIF-1α expression in LPS-stimulated human and mice leukocytes. Three key observations support this conclusion. First, no changes in AMPKα and TLR4 signaling intermediates (mammalian target of rapamycin complex-1/S6 kinase 1/HIF-1α) were detected in LPS-stimulated MMP9-deficient mice leukocytes. Second, rMMP9 cleaved human AMPKα ex vivo, producing degradation products similar in size to those detected following LPS stimulation. Third, MMP9 inhibitors prevented AMPKα degradation and HIF-1α expression in LPS-activated human leukocytes, whereas AMPK activators blocked MMP9 and HIF-1α expression. Significantly, AMPKα degradation, MMP9, and TLR4 signaling intermediates were all detected in leukocytes from patients with type 2 diabetes mellitus and patients following cardiopulmonary bypass surgery. Plasma from these two patient cohorts induced AMPKα cleavage and TLR4 signaling intermediates in healthy donor leukocytes and either a TLR4 inhibitor or polymyxin prevented these outcomes. Detection of AMPKα degradation, MMP9 expression, and TLR4 signaling intermediates described in this study in leukocytes, the most readily available human cells for clinical investigation, may provide a powerful tool for further exploring the role of TLR4 signaling in human diseases and lead to identification of new, context-specific therapeutic modalities for precision medicine.
Collapse
Affiliation(s)
- Zhiyong Zhang
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08903
| | - Louis F Amorosa
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08903; and
| | - Susette M Coyle
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08903
| | - Marie A Macor
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08903
| | - Sara E Lubitz
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08903; and
| | - Jeffrey L Carson
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08903; and
| | - Morris J Birnbaum
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Leonard Y Lee
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08903
| | - Beatrice Haimovich
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08903;
| |
Collapse
|
44
|
Khan ASA, Gibson JM, Carlson GL, Rooyackers O, New JP, Soop M. Protein kinetics in human endotoxaemia and their temporal relation to metabolic, endocrine and proinflammatory cytokine responses. Br J Surg 2015; 102:767-75. [PMID: 25950998 DOI: 10.1002/bjs.9817] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 11/03/2014] [Accepted: 03/03/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND Sepsis is associated with profound alterations in protein metabolism. The unpredictable time course of sepsis and the multiplicity of confounding factors prevent studies of temporal relations between the onset of endocrine and proinflammatory cytokine responses and the onset of protein catabolism. This study aimed to determine the time course of whole-body protein catabolism, and relate it to the endocrine, metabolic and cytokine responses in a human endotoxaemia model of early sepsis. METHODS Six healthy male volunteers were studied twice in random order, before and for 600 min after administration of either an intravenous bolus of Escherichia coli lipopolysaccharide (LPS) or sterile saline. Whole-body protein synthesis, breakdown and net protein breakdown were measured by amino acid tracer infusion, and related to changes in plasma levels of growth hormone, glucagon, cortisol, insulin-like growth factor (IGF) 1, tumour necrosis factor (TNF) α and interleukin (IL) 6. RESULTS Protein synthesis, breakdown and net protein breakdown increased and peaked 120 min after LPS administration (P < 0·001), the alterations persisting for up to 480 min. These peaks coincided with peaks in plasma growth hormone, TNF-α and IL-6 concentrations (P = 0·049, P < 0·001 and P < 0·001 for LPS versus saline), whereas plasma cortisol concentration peaked later. No alterations in plasma insulin or glucagon concentrations, or in the IGF axis were observed during the period of abnormalities of protein metabolism. CONCLUSION LPS administration induced an early protein catabolic response in young men and this coincided with changes in plasma growth hormone, TNF-α and IL-6 concentrations, rather than changes in cortisol, glucagon, insulin or the IGF axis. Surgical relevance Sepsis in surgical patients is common and remains associated with substantial mortality. Although sepsis is a heterogeneous condition and its pathophysiology therefore difficult to study, a universal and profound clinical problem is protein catabolism not responsive to nutritional support. Human experimental endotoxaemia is a promising model of clinical sepsis that can be used to elucidate underlying pathophysiology and explore novel therapeutic approaches. This study demonstrates that human experimental endotoxaemia replicates the changes in whole-body protein turnover seen in clinical sepsis. Frequent measurements allowed identification of tumour necrosis factor (TNF) α, interleukin (IL) 6 and growth hormone as putative mediators. Human experimental endotoxaemia is a valid model for further study of mechanisms and putative therapies of catabolism associated with sepsis. In particular, effects of TNF-α and IL-6 blockade should be evaluated.
Collapse
Affiliation(s)
- A S A Khan
- Vascular Research Group, Salford Royal NHS Foundation Trust, Salford, UK
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
In this meta-study, we aimed to integrate biological insights gained from two levels of -omics analyses on the response to systemic inflammation induced by lipopolysaccharide in humans. We characterized the interplay between plasma metabolite compositions and transcriptional response of leukocytes through integration of transcriptomics with plasma metabonomics. We hypothesized that the drastic changes in the immediate environment of the leukocytes might have an adaptive effect on shaping their transcriptional response in conjunction with the initial inflammatory stimuli. Indeed, we observed that leukocytes, most notably, tune the activity of lipid- and protein-associated processes at the transcriptional level in accordance with the fluctuations in metabolite compositions of surrounding plasma. A closer look into the transcriptional control of only metabolic pathways uncovered alterations in bioenergetics and defenses against oxidative stress closely associated with mitochondrial dysfunction and shifts in energy production observed during inflammatory processes.
Collapse
|
46
|
Hormone and Cytokine Responses to Repeated Endotoxin Exposures—No Evidence of Endotoxin Tolerance After 5 Weeks in Humans. Shock 2015; 44:32-5. [DOI: 10.1097/shk.0000000000000384] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
47
|
Kox M, van Eijk LT, Verhaak T, Frenzel T, Kiers HD, Gerretsen J, van der Hoeven JG, Kornet L, Scheiner A, Pickkers P. Transvenous vagus nerve stimulation does not modulate the innate immune response during experimental human endotoxemia: a randomized controlled study. Arthritis Res Ther 2015; 17:150. [PMID: 26049730 PMCID: PMC4480894 DOI: 10.1186/s13075-015-0667-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/29/2015] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION Vagus nerve stimulation (VNS) exerts beneficial anti-inflammatory effects in various animal models of inflammation, including collagen-induced arthritis, and is implicated in representing a novel therapy for rheumatoid arthritis. However, evidence of anti-inflammatory effects of VNS in humans is very scarce. Transvenous VNS (tVNS) is a newly developed and less invasive method to stimulate the vagus nerve. In the present study, we determined whether tVNS is a feasible and safe procedure and investigated its putative anti-inflammatory effects during experimental human endotoxemia. METHODS We performed a randomized double-blind sham-controlled study in healthy male volunteers. A stimulation catheter was inserted in the left internal jugular vein at spinal level C5-C7, adjacent to the vagus nerve. In the tVNS group (n = 10), stimulation was continuously performed for 30 minutes (0-10 V, 1 ms, 20 Hz), starting 10 minutes before intravenous administration of 2 ng kg(-1) Escherichia coli lipopolysaccharide (LPS). Sham-instrumented subjects (n = 10) received no electrical stimulation. RESULTS No serious adverse events occurred throughout the study. In the tVNS group, stimulation of the vagus nerve was achieved as indicated by laryngeal vibration. Endotoxemia resulted in fever, flu-like symptoms, and hemodynamic changes that were unaffected by tVNS. Furthermore, plasma levels of inflammatory cytokines increased sharply during endotoxemia, but responses were similar between groups. Finally, cytokine production by leukocytes stimulated with LPS ex vivo, as well as neutrophil phagocytosis capacity, were not influenced by tVNS. CONCLUSIONS tVNS is feasible and safe, but does not modulate the innate immune response in humans in vivo during experimental human endotoxemia. TRIAL REGISTRATION Clinicaltrials.gov NCT01944228. Registered 12 September 2013.
Collapse
Affiliation(s)
- Matthijs Kox
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Lucas T van Eijk
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Tim Verhaak
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Tim Frenzel
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Harmke D Kiers
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Jelle Gerretsen
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Johannes G van der Hoeven
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Lilian Kornet
- Medtronic Inc., Endepolsdomein 5, 6229, GW, Maastricht, The Netherlands.
| | - Avram Scheiner
- Medtronic Inc., 8200 Coral Sea St NE, Mounds View, MN, 55112, USA.
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| |
Collapse
|
48
|
Rittig N, Bach E, Thomsen HH, Johannsen M, Jørgensen JO, Richelsen B, Jessen N, Møller N. Amino acid supplementation is anabolic during the acute phase of endotoxin-induced inflammation: A human randomized crossover trial. Clin Nutr 2015; 35:322-330. [PMID: 25896101 DOI: 10.1016/j.clnu.2015.03.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/10/2015] [Accepted: 03/30/2015] [Indexed: 01/16/2023]
Abstract
BACKGROUND & AIMS Inflammation is catabolic and causes muscle loss. It is unknown if amino acid supplementation reverses these effects during the acute phase of inflammation. The aim was to test whether amino acid supplementation counteracts endotoxin-induced catabolism. METHODS Eight young, healthy, lean males were investigated three times in randomized order: (i) normal conditions (Placebo), (ii) endotoxemia (LPS), and (iii) endotoxemia with amino acid supplementation (LPS + A). Protein kinetics were determined using phenylalanine, tyrosine, and urea tracers. Each study day consisted of a four-hour non-insulin stimulated period and a two-hour hyperinsulinemic euglycemic clamp period. Muscle biopsies were collected once each period. RESULTS Endotoxin administration created a significant inflammatory response (cytokines, hormones, and vital parameters) without significant differences between LPS and LPS + A. Whole body protein breakdown was elevated during LPS compared with Placebo and LPS + A (p < 0.05). Whole body protein synthesis was higher during LPS + A than both Placebo and LPS (p < 0.003). Furthermore, protein synthesis was higher during LPS than during Placebo (p < 0.02). Net muscle phenylalanine release was markedly decreased during LPS + A (p < 0.004), even though muscle protein synthesis and breakdown rates did not differ significantly between interventions. LPS + A increased mammalian target of rapamycin (mTOR) phosphorylation (p < 0.05) and eukaryotic translation factor 4E-binding protein 1 (4EBP1) phosphorylation (p = 0.007) without activating AMPK or affecting insulin signaling through Akt. During insulin stimulation net muscle phenylalanine release and protein degradation were further reduced. CONCLUSIONS Amino acid supplementation in the acute phase of inflammation reduces whole body and muscle protein loss, and this effect is associated with activation of mTOR and downstream signaling to protein synthesis through mTORC1, suggesting a therapeutic role for intravenous amino acids in inflammatory states. CLINICAL TRIAL REGISTRY The Central Denmark Region Ethics Commitee (1-10-71-410-12) www.clinicaltrials.gov (identification number NCT01705782).
Collapse
Affiliation(s)
- N Rittig
- Department of Internal Medicine and Endocrinology (MEA) and Medical Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, DK-8000 Aarhus C, Denmark.
| | - E Bach
- Department of Internal Medicine and Endocrinology (MEA) and Medical Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, DK-8000 Aarhus C, Denmark
| | - H H Thomsen
- Department of Internal Medicine and Endocrinology (MEA) and Medical Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, DK-8000 Aarhus C, Denmark
| | - M Johannsen
- Section for Forensic Chemistry, Department of Forensic Medicine, Aarhus University Hospital, Brendstrupgaardsvej 100, 8200 Aarhus N, Denmark
| | - J O Jørgensen
- Department of Internal Medicine and Endocrinology (MEA) and Medical Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, DK-8000 Aarhus C, Denmark
| | - B Richelsen
- Department of Internal Medicine and Endocrinology (MEA) and Medical Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, DK-8000 Aarhus C, Denmark
| | - N Jessen
- Department of Internal Medicine and Endocrinology (MEA) and Medical Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, DK-8000 Aarhus C, Denmark; Department of Biochemical Pathology, Institute for Clinical Medicine, Aarhus University Hospital, Denmark
| | - N Møller
- Department of Internal Medicine and Endocrinology (MEA) and Medical Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, DK-8000 Aarhus C, Denmark
| |
Collapse
|
49
|
Kamisoglu K, Haimovich B, Calvano SE, Coyle SM, Corbett SA, Langley RJ, Kingsmore SF, Androulakis IP. Human metabolic response to systemic inflammation: assessment of the concordance between experimental endotoxemia and clinical cases of sepsis/SIRS. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2015; 19:71. [PMID: 25887472 PMCID: PMC4383069 DOI: 10.1186/s13054-015-0783-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 02/03/2015] [Indexed: 12/28/2022]
Abstract
Introduction Two recent, independent, studies conducted novel metabolomics analyses relevant to human sepsis progression; one was a human model of endotoxin (lipopolysaccharide (LPS)) challenge (experimental endotoxemia) and the other was community acquired pneumonia and sepsis outcome diagnostic study (CAPSOD). The purpose of the present study was to assess the concordance of metabolic responses to LPS and community-acquired sepsis. Methods We tested the hypothesis that the patterns of metabolic response elicited by endotoxin would agree with those in clinical sepsis. Alterations in the plasma metabolome of the subjects challenged with LPS were compared with those of sepsis patients who had been stratified into two groups: sepsis patients with confirmed infection and non-infected patients who exhibited systemic inflammatory response syndrome (SIRS) criteria. Common metabolites between endotoxemia and both these groups were individually identified, together with their direction of change and functional classifications. Results Response to endotoxemia at the metabolome level elicited characteristics that agree well with those observed in sepsis patients despite the high degree of variability in the response of these patients. Moreover, some distinct features of SIRS have been identified. Upon stratification of sepsis patients based on 28-day survival, the direction of change in 21 of 23 metabolites was the same in endotoxemia and sepsis survival groups. Conclusions The observed concordance in plasma metabolomes of LPS-treated subjects and sepsis survivors strengthens the relevance of endotoxemia to clinical research as a physiological model of community-acquired sepsis, and gives valuable insights into the metabolic changes that constitute a homeostatic response. Furthermore, recapitulation of metabolic differences between sepsis non-survivors and survivors in LPS-treated subjects can enable further research on the development and assessment of rational clinical therapies to prevent sepsis mortality. Compared with earlier studies which focused exclusively on comparing transcriptional dynamics, the distinct metabolomic responses to systemic inflammation with or without confirmed infection, suggest that the metabolome is much better at differentiating these pathophysiologies. Finally, the metabolic changes in the recovering patients shift towards the LPS-induced response pattern strengthening the notion that the metabolic, as well as transcriptional responses, characteristic to the endotoxemia model represent necessary and “healthy” responses to infectious stimuli. Electronic supplementary material The online version of this article (doi:10.1186/s13054-015-0783-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kubra Kamisoglu
- Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, NJ, 08854, USA.
| | - Beatrice Haimovich
- Department of Surgery, Rutgers - Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
| | - Steve E Calvano
- Department of Surgery, Rutgers - Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
| | - Susette M Coyle
- Department of Surgery, Rutgers - Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
| | - Siobhan A Corbett
- Department of Surgery, Rutgers - Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
| | - Raymond J Langley
- Department of Respiratory Immunology, Lovelace Respiratory Research Institute, Albuquerque, NM, 87108, USA.
| | - Stephen F Kingsmore
- Center for Pediatric Genomic Medicine, Children's Mercy, Kansas City, MO, 64108, USA. .,Departments of Pediatrics and Obstetrics/Gynecology, University of Missouri, Kansas City, MO, 64108, USA.
| | - Ioannis P Androulakis
- Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, NJ, 08854, USA. .,Department of Surgery, Rutgers - Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA. .,Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ, 08854, USA.
| |
Collapse
|
50
|
Skulas-Ray AC. Omega-3 fatty acids and inflammation: a perspective on the challenges of evaluating efficacy in clinical research. Prostaglandins Other Lipid Mediat 2015; 116-117:104-11. [PMID: 25698680 DOI: 10.1016/j.prostaglandins.2015.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 02/03/2015] [Accepted: 02/09/2015] [Indexed: 12/31/2022]
Abstract
Chronic inflammation is a common underpinning of many diseases. There is a strong pre-clinical evidence base demonstrating the efficacy of omega-3 fatty acids for ameliorating inflammation and thereby reducing disease burden. Clinically, C-reactive protein (CRP) serves as both a reliable marker for monitoring inflammation and a modifiable endpoint for studies of anti-inflammatory pharmaceuticals. However, clinical omega-3 fatty acid supplementation trials have not replicated pre-clinical findings in terms of consistent CRP reductions. Methodological differences present numerous challenges in translating pre-clinical evidence to clinical results. It is crucial that future clinical nutrition research clearly distinguish between the reversal of established inflammation and preventing the development of inflammation. Future clinical studies evaluating the ability of omega-3 fatty acids to attenuate an excessive inflammatory response, may be advanced by employing new statistical approaches and utilizing models of induced inflammation, such as low-dose human endotoxemia.
Collapse
Affiliation(s)
- Ann C Skulas-Ray
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, United States.
| |
Collapse
|