1
|
Pellon A, Palacios A, Abecia L, Rodríguez H, Anguita J. Friends to remember: innate immune memory regulation by the microbiota. Trends Microbiol 2025; 33:510-520. [PMID: 39794207 DOI: 10.1016/j.tim.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/28/2024] [Accepted: 12/11/2024] [Indexed: 01/13/2025]
Abstract
Innate immune memory (IIM) is the process by which, upon a primary challenge, innate immune cells alter their epigenetic, transcriptional, and immunometabolic profiles, resulting in modified secondary responses. Unlike infections or other immune-system-related diseases, the role of IIM in nonpathogenic contexts is less understood. An increasing body of research has shown that normal microbiota members or their metabolic byproducts induce alternative memory phenotypes, suggesting that memory cells contribute to homeostasis in mucosal areas. In this review, we discuss the newest insights in the emerging field of IIM to the microbiota and the potential of manipulating these long-term responses to promote better mucosal health.
Collapse
Affiliation(s)
- Aize Pellon
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA, Derio, Spain.
| | - Ainhoa Palacios
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA, Derio, Spain; Present address: Research Unit, Basque Center for Blood Transfusion and Human Tissues, Osakidetza; Galdakao, Spain and Cell Therapy, Stem Cells and Tissues Group, BioBizkaia Health Research Institute; Barakaldo, Spain
| | - Leticia Abecia
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA, Derio, Spain; Department of Immunology, Microbiology, and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Héctor Rodríguez
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA, Derio, Spain
| | - Juan Anguita
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA, Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
2
|
Perrine WG, Sauer EL, Love AC, Morris A, Novotny J, DuRant SE. A high-lipid diet leads to greater pathology and lower tolerance during infection. J Exp Biol 2025; 228:JEB249541. [PMID: 39886835 PMCID: PMC11925396 DOI: 10.1242/jeb.249541] [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: 09/11/2024] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Altered food landscapes contribute importantly to wildlife disease dynamics and may play a critical role in host heterogeneity in disease outcomes through changes in host diet composition. We explored the effects of dietary macronutrient composition on disease pathology and feeding behavior of canaries (Serinus canaria domestica) infected with Mycoplasma gallisepticum (MG). In the first experiment, we provided canaries with isocaloric diets composed of identical ingredients that varied in macronutrient content (high protein or high lipid) then MG- or sham-inoculated birds. In the second experiment, we offered both diets to canaries before and after MG or sham inoculation. In experiment one, high-protein diet birds consumed more food than high-lipid diet birds and experienced a more pronounced decrease in food intake after infection. High-protein diet birds were more tolerant to MG infection, exhibiting reduced pathology when compared with high-lipid diet birds, despite the two treatments having similar levels of MG-specific antibodies and MG loads. When birds had access to both diets, they consumed more of the high-protein diet and experienced pathology for less time than lipid- or protein-restricted birds. These results highlight that macronutrient makeup of the diet can shape vertebrate host tolerance and pathology, which has direct implications for host-pathogen transmission dynamics.
Collapse
Affiliation(s)
- Weston G. Perrine
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Erin L. Sauer
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Ashley C. Love
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
- Department of Biology, Miami University, Miami, OH 45056, USA
| | - Ashley Morris
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Johnathan Novotny
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Sarah E. DuRant
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
3
|
Dunbar SB, McCauley LA, Yeager KA, Holstad MM, Corwin EJ, Hertzberg V. Multiple Chronic Conditions, Metabolites, and Symptoms. Nurs Res 2025; 74:4-8. [PMID: 39666466 DOI: 10.1097/nnr.0000000000000789] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
|
4
|
Davis E, Dunbar SB, Higgins MK, Wood K, Ferranti E, Morris AA, Butts B. Western Diet and Inflammatory Mechanisms in African American Adults With Heart Failure. Nurs Res 2025; 74:20-26. [PMID: 39666467 PMCID: PMC11643355 DOI: 10.1097/nnr.0000000000000782] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
BACKGROUND Black adults have a higher risk for heart failure (HF) than others, which may be related to higher cardiovascular risk factors and also inflammatory dietary patterns. The Western diet is associated with inflammation and contributes to HF. Trimethylamine N-oxide is a diet-linked metabolite that contributes to inflammation and is associated with higher tumor necrosis factor-alpha (TNF-α) levels, especially in HF populations. The dietary inflammatory index score measures a diet's inflammatory potential and food's inflammatory effects. OBJECTIVE The purpose of this pilot study was to explore associations between the Western diet, dietary inflammatory index, trimethylamine N-oxide, relevant covariates and variables, and TNF-α in Black persons with HF. METHODS Thirty-one Black participants (mean age = 55 years, 68% women) with HF were enrolled. Trimethylamine N-oxide and TNF-α levels were analyzed using immunoassays. A food frequency questionnaire was completed, and dietary inflammatory index scores and food groups were calculated. Analyses included correlations and I-test statistics. RESULTS Mean dietary inflammatory index score was -0.38, noting an anti-inflammatory diet with slightly higher inflammatory diet scores in men compared to women. The dietary inflammatory index score showed a negative association with dietary choline but not with trimethylamine N-oxide or TNF-α. Trimethylamine N-oxide and age were positively correlated, along with the correlation for TNF-α with a moderate effect size. No relationship was found among dietary inflammatory index, TNF-α, and trimethylamine N-oxide variables. DISCUSSION A greater understanding of intake of inflammatory foods and relationships with immune factors is warranted to inform intervention development. In Black adults with HF, it is important to consider the intake of inflammatory foods as increased age may affect the retention of dietary metabolites. Metabolites may also increase the levels of inflammation. Knowledge about these relationships could lead to tailored dietary interventions based on diet, age, and culture patterns.
Collapse
|
5
|
Spitler KM, Shetty SK, Davies BSJ. Effects of age and diet on triglyceride metabolism in mice. J Lipid Res 2025; 66:100706. [PMID: 39566846 PMCID: PMC11730548 DOI: 10.1016/j.jlr.2024.100706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 11/22/2024] Open
Abstract
Both age and diet can contribute to alterations in triglyceride metabolism and subsequent metabolic disease. In humans, plasma triglyceride levels increase with age. Diets high in saturated fats can increase triglyceride levels while diets high in omega-3 fatty acids decrease triglyceride levels. Here we asked how age and long-term diet altered triglyceride metabolism in mice. We fed male and female C57Bl/6 mice a low-fat diet, a western diet (WD), or a diet high in polyunsaturated and omega-3 fatty acids (n3D) for up to 2 years. We measured survival, body composition, plasma triglyceride levels, chylomicron clearance, and oral fat, glucose, and insulin tolerance. Triglyceride levels in mice did not increase with age, regardless of diet. Oral fat tolerance increased with age, while chylomicron clearance remained unchanged. Decreased survival was observed in WD-fed mice. Interestingly, n3D-fed mice gained more lean mass and had lower insulin levels than WD-fed or LFD-fed mice. Moreover, triglyceride uptake into the hearts of n3D-fed mice was strikingly higher than in other groups. Our data indicate that in C57Bl/6 mice, age-induced changes in triglyceride metabolism differ from those observed in humans. Mice, like humans, appeared to have decreased fat absorption with age, but in mice plasma triglyceride clearance did not decrease with age, resulting in lower plasma triglyceride levels and improved fat tolerance with age. Although a chronic diet high in omega-3 fatty acids increased insulin sensitivity and triglyceride uptake specifically into the heart, how these observations are connected is unclear.
Collapse
Affiliation(s)
- Kathryn M Spitler
- Department of Biochemistry and Molecular Biology, Fraternal Order of Eagles Diabetes Research Center, and Obesity Research and Education Initiative, University of Iowa, Iowa City, IA
| | - Shwetha K Shetty
- Department of Biochemistry and Molecular Biology, Fraternal Order of Eagles Diabetes Research Center, and Obesity Research and Education Initiative, University of Iowa, Iowa City, IA
| | - Brandon S J Davies
- Department of Biochemistry and Molecular Biology, Fraternal Order of Eagles Diabetes Research Center, and Obesity Research and Education Initiative, University of Iowa, Iowa City, IA.
| |
Collapse
|
6
|
Gabrielli M, Zaccaria R, Impagnatiello M, Zileri Dal Verme L, Gasbarrini A. Nutritional Strategies for the Treatment and Prevention of Sepsis Outside the Intensive Care Unit. Nutrients 2024; 16:3985. [PMID: 39683380 DOI: 10.3390/nu16233985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/14/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Sepsis is a life-threatening condition characterized by an imbalanced immune response to infection, posing a significant challenge in hospital settings due to its high morbidity and mortality rates. While much attention has been given to patients in the ICU, uncertainties remain regarding the nutritional management of septic patients in non-intensive wards. This narrative review aims to address these gaps by exploring key aspects of nutritional care in sepsis patients admitted to non-intensive wards. METHODS We examine the pathophysiological mechanisms driving metabolic alterations in sepsis, methods for effective nutritional assessment, and supplementation strategies, including the potential role of specific nutrients. Additionally, we discuss the preventive role of nutrition, with a focus on gut microbiota modulation. CONCLUSIONS By synthesizing the available literature, this review provides evidence-based insights to guide nutritional strategies for managing sepsis in patients hospitalized in non-intensive wards and highlights critical areas for future research.
Collapse
Affiliation(s)
- Maurizio Gabrielli
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Raffaella Zaccaria
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Michele Impagnatiello
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Lorenzo Zileri Dal Verme
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
7
|
Ma Y, Zhao Y, Zhang X. Factors affecting neutrophil functions during sepsis: human microbiome and epigenetics. J Leukoc Biol 2024; 116:672-688. [PMID: 38734968 DOI: 10.1093/jleuko/qiae107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 05/13/2024] Open
Abstract
Sepsis is a severe disease that occurs when the body's immune system reacts excessively to infection. The body's response, which includes an intense antibacterial reaction, can damage its tissues and organs. Neutrophils are the major components of white blood cells in circulation, play a vital role in innate immunity while fighting against infections, and are considered a feature determining sepsis classification. There is a plethora of basic research detailing neutrophil functioning, among which, the study of neutrophil extracellular traps is providing novel insights into mechanisms and treatments of sepsis. This review explores their functions, dysfunctions, and influences in the context of sepsis. The interplay between neutrophils and the human microbiome and the impact of DNA methylation on neutrophil function in sepsis are crucial areas of study. The interaction between neutrophils and the human microbiome is complex, particularly in the context of sepsis, where dysbiosis may occur. We highlight the importance of deciphering neutrophils' functional alterations and their epigenetic features in sepsis because it is critical for defining sepsis endotypes and opening up the possibility for novel diagnostic methods and therapy. Specifically, epigenetic signatures are pivotal since they will provide a novel implication for a sepsis diagnostic method when used in combination with the cell-free DNA. Research is exploring how specific patterns of DNA methylation in neutrophils, detectable in cell-free DNA, could serve as biomarkers for the early detection of sepsis.
Collapse
Affiliation(s)
- Yina Ma
- Department of Urology Surgery, Beijing Chaoyang Hospital, Capital Medical University, Shijingshan District, Beijing 100043, China
| | - Yu Zhao
- Department of Urology Surgery, Beijing Chaoyang Hospital, Capital Medical University, Shijingshan District, Beijing 100043, China
| | - Xin Zhang
- Department of Urology Surgery, Beijing Chaoyang Hospital, Capital Medical University, Shijingshan District, Beijing 100043, China
| |
Collapse
|
8
|
Koga HK, Grodstein F, Williams DR, Demeo DL, Kubzansky LD. Relations of optimism and purpose in life to immune markers in aging. J Psychosom Res 2024; 184:111851. [PMID: 38964200 DOI: 10.1016/j.jpsychores.2024.111851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/13/2024] [Accepted: 06/21/2024] [Indexed: 07/06/2024]
Abstract
OBJECTIVE Optimism and purpose in life are associated with improved health outcomes. More information is needed on biological mechanisms, including immunosenescence. We investigated if psychological well-being is associated with healthier immunosenescence-related measures including naïve and terminally differentiated CD4+ and CD8+ T cell percentages, CD4+:CD8+, and cytomegalovirus (CMV) IgG response. METHODS Participants were adults over age 50 from the Health and Retirement Study. Optimism was measured using the Life Orientation Test Revised. Purpose in life was assessed using the subscale from the Ryff psychological well-being measure. We examined the cross-sectional associations of optimism and purpose in life with measures of T cell subsets using linear regression and with CMV IgG using ordered logit regression, controlling for potential confounding factors. RESULTS The final analytic sample ranged from 7250 to 7870. After adjusting for sociodemographic factors, a 1-SD increment in optimism was associated with the percentage of naïve CD4+ T cells increasing by 0.6 (95%CI 0.2%, 1.0%). A 1-SD increment in purpose in life was associated with the percentage of naïve CD4+ T cells increasing by 0.9 (95%CI 0.5%, 1.3%) after adjusting for sociodemographic factors and the association was maintained after further adjustments for health conditions, depression, and health behaviors. For naïve CD8+ T cell percentages, CD4:CD8 ratios, and CMV IgG antibodies, associations were seen only in models that adjusted for age. No significant associations were seen in any models for the terminally differentiated CD4+ and CD8+ T cells. CONCLUSIONS We found associations of optimism and purpose in life with naïve CD4+ T cell percentages.
Collapse
Affiliation(s)
- Hayami K Koga
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America; Harvard Center for Population and Development Studies, Cambridge, MA, United States of America.
| | - Francine Grodstein
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, United States of America
| | - David R Williams
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America; Department of African and African American Studies, Department of Sociology, Harvard University, Cambridge, MA, United States of America
| | - Dawn L Demeo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America; Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, United States of America
| | - Laura D Kubzansky
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America
| |
Collapse
|
9
|
Shokoples BG, Paradis P, Schiffrin EL. Immunological insights into hypertension: unraveling triggers and potential therapeutic avenues. Hypertens Res 2024; 47:2115-2125. [PMID: 38778172 DOI: 10.1038/s41440-024-01731-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/02/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
Hypertension remains the leading cause of morbidity and mortality worldwide. Despite its prevalence, the development of novel antihypertensive therapies has only recently accelerated, with novel agents not yet commercialized, leaving a substantial proportion of individuals resistant to existing treatments. The intricate pathophysiology of hypertension is now understood to involve chronic low-grade inflammation, which places the immune system in the spotlight as a potential target for new therapeutics. This review explores the factors that initiate and sustain an immune response in hypertension, offering insights into potential targets for new treatments. Several factors contribute to immune activation in hypertension, including diet and damage-associated molecular pattern (DAMP) generation. Diets rich in fat or sodium can promote inflammation by inducing intestinal barrier dysfunction and triggering salt-sensitive receptors in T cells and dendritic cells. DAMPs, such as extracellular adenosine triphosphate and heat-shock protein 70, are released during episodes of increased blood pressure, contributing to immune cell activation and inflammation. Unconventional innate-like γδ T cells contribute to initiating and maintaining an immune response through their potential involvement in antigen presentation and regulating cytokine-mediated responses. Immunologic memory, sustained through the formation of effector memory T cells after exposure to hypertensive insults, likely contributes to maintaining an immune response in hypertension. When exposed to hypertensive insults, these memory cells are rapidly activated and contribute to elevated blood pressure and end-organ damage. Evidence from human hypertension, although limited, supports the relevance of distinct immune pathways in hypertension, and highlights the potential of targeted immune interventions in human hypertension. Diet and acute bouts of high blood pressure result in the release of dietary triggers, neoantigens, and damage-associated molecular patterns (DAMPs), which promote immune system activation. Elements such as lipopolysaccharides (LPS), sodium, heat-shock protein (HSP)70, extracellular adenosine triphosphate (eATP), and growth arrest-specific 6 (GAS6) promote activation of innate immune cells such as dendritic cells (DCs) and monocytes (Mo) through their respective receptors (toll-like receptor [TLR]4, amiloride-sensitive epithelial sodium channel [ENaC], TLR2/4, P2X7 receptor [P2RX7], and Axl) leading to costimulatory molecule expression and interleukin (IL)-1β and IL-23 production. The neoantigens HSP70 and isolevuglandins (IsoLGs) are presented to T cells by DCs and possibly γδ T cells, triggering T cell activation, IL-17 and interferon (IFN)-γ production, and the formation of T effector memory (TEM) cells in the kidney, perivascular adipose tissue, bone marrow, and spleen. Exposure of TEM cells to their cognate antigen or previous activating stimuli causes these cells rapid expansion and activation. Cumulatively, this inflammatory state contributes to hypertension and end-organ damage. The figure was created using images from smart.servier.com and is licensed under a Creative Commons Attribution 4.0 license (CC BY 4.0).
Collapse
Affiliation(s)
- Brandon G Shokoples
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research and McGill University, Montréal, QC, Canada
| | - Pierre Paradis
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research and McGill University, Montréal, QC, Canada
| | - Ernesto L Schiffrin
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research and McGill University, Montréal, QC, Canada.
- Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montréal, QC, Canada.
| |
Collapse
|
10
|
Adolph TE, Tilg H. Western diets and chronic diseases. Nat Med 2024; 30:2133-2147. [PMID: 39085420 DOI: 10.1038/s41591-024-03165-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024]
Abstract
'Westernization', which incorporates industrial, cultural and dietary trends, has paralleled the rise of noncommunicable diseases across the globe. Today, the Western-style diet emerges as a key stimulus for gut microbial vulnerability, chronic inflammation and chronic diseases, affecting mainly the cardiovascular system, systemic metabolism and the gut. Here we review the diet of modern times and evaluate the threat it poses for human health by summarizing recent epidemiological, translational and clinical studies. We discuss the links between diet and disease in the context of obesity and type 2 diabetes, cardiovascular diseases, gut and liver diseases and solid malignancies. We collectively interpret the evidence and its limitations and discuss future challenges and strategies to overcome these. We argue that healthcare professionals and societies must react today to the detrimental effects of the Western diet to bring about sustainable change and improved outcomes in the future.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
11
|
Spitler KM, Shetty SK, Davies BS. Effects of Age and Diet on Triglyceride Metabolism in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.19.602944. [PMID: 39091783 PMCID: PMC11291025 DOI: 10.1101/2024.07.19.602944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Background Both age and diet can contribute to alterations in triglyceride metabolism and subsequent metabolic disease. In humans, plasma triglyceride levels increase with age. Diets high in saturated fats can increase triglyceride levels while diets high in omega-3 fatty acids decrease triglyceride levels. Here we asked how age and long-term diet effected triglyceride metabolism in mice. Methods We fed male and female mice a low-fat diet, a western diet, or a diet high in polyunsaturated and omega-3 (n-3) fatty acids for up to 2 years. We measured survival, body composition, plasma triglyceride levels, chylomicron clearance, and oral fat, glucose, and insulin tolerance. Results Triglyceride levels in mice did not increase with age, regardless of diet. Oral fat tolerance increased with age, while chylomicron clearance remained unchanged. Mice fed western diet had decreased survival. Interestingly, mice fed the n-3 diet gained more lean mass, and had lower insulin levels than mice fed either low-fat or western diet. Moreover, triglyceride uptake into the hearts of mice fed the n-3 diet was strikingly higher than in other groups. Conclusions In mice, age-induced changes in triglyceride metabolism did not match those in humans. Our data suggested that mice, like humans, had decreased fat absorption with age, but plasma triglyceride clearance did not decrease with age in mice, resulting in lower plasma triglyceride levels and improved oral fat tolerance with age. A chronic diet high in n-3 fatty acids increased insulin sensitivity and uptake of triglycerides specifically into the heart but how these observations are connected is unclear.
Collapse
Affiliation(s)
- Kathryn M. Spitler
- Department of Biochemistry and Molecular Biology, Fraternal Order of Eagles Diabetes Research Center, and Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242
| | - Shwetha K. Shetty
- Department of Biochemistry and Molecular Biology, Fraternal Order of Eagles Diabetes Research Center, and Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242
| | - Brandon S.J. Davies
- Department of Biochemistry and Molecular Biology, Fraternal Order of Eagles Diabetes Research Center, and Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
12
|
Zhang N, Wang X, Feng M, Li M, Wang J, Yang H, He S, Xia Z, Shang L, Jiang X, Sun M, Wu Y, Ren C, Zhang X, Li J, Gao F. Early-life exercise induces immunometabolic epigenetic modification enhancing anti-inflammatory immunity in middle-aged male mice. Nat Commun 2024; 15:3103. [PMID: 38600123 PMCID: PMC11006929 DOI: 10.1038/s41467-024-47458-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 04/03/2024] [Indexed: 04/12/2024] Open
Abstract
Exercise is usually regarded to have short-term beneficial effects on immune health. Here we show that early-life regular exercise exerts long-term beneficial effects on inflammatory immunity. Swimming training for 3 months in male mice starting from 1-month-old curbs cytokine response and mitigates sepsis when exposed to lipopolysaccharide challenge, even after an 11-month interval of detraining. Metabolomics analysis of serum and liver identifies pipecolic acid, a non-encoded amino acid, as a pivotal metabolite responding to early-life regular exercise. Importantly, pipecolic acid reduces inflammatory cytokines in bone marrow-derived macrophages and alleviates sepsis via inhibiting mTOR complex 1 signaling. Moreover, early-life exercise increases histone 3 lysine 4 trimethylation at the promoter of Crym in the liver, an enzyme responsible for catalyzing pipecolic acid production. Liver-specific knockdown of Crym in adult mice abolishes this early exercise-induced protective effects. Our findings demonstrate that early-life regular exercise enhances anti-inflammatory immunity during middle-aged phase in male mice via epigenetic immunometabolic modulation, in which hepatic pipecolic acid production has a pivotal function.
Collapse
Affiliation(s)
- Nini Zhang
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
- Department of Pediatrics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinpei Wang
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Mengya Feng
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
- Center for Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Min Li
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Jing Wang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hongyan Yang
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Siyu He
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Ziqi Xia
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Lei Shang
- Key Laboratory of Hazard Assessment and Control in Special Operational Environment, Ministry of Education; Department of Health Statistics, School of Public Health, Fourth Military Medical University, Xi'an, China
| | - Xun Jiang
- Department of Pediatrics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Mao Sun
- Department of Biochemistry and Molecular Biology, Center for DNA Typing, Fourth Military Medical University, Xi'an, China
| | - Yuanming Wu
- Department of Biochemistry and Molecular Biology, Center for DNA Typing, Fourth Military Medical University, Xi'an, China
| | - Chaoxue Ren
- School of Sport and Health Science, Xi'an Physical Education University, Xi'an, China
| | - Xing Zhang
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Jia Li
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China.
- Key Laboratory of Hazard Assessment and Control in Special Operational Environment, Ministry of Education; Department of Health Statistics, School of Public Health, Fourth Military Medical University, Xi'an, China.
| | - Feng Gao
- Key Laboratory of Aerospace Medicine, Ministry of Education; School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
13
|
Zhou Z, Wang H, Tan S, Zhang H, Zhu Y. The alterations of innate immunity and enhanced severity of infections in diabetes mellitus. Immunology 2024; 171:313-323. [PMID: 37849389 DOI: 10.1111/imm.13706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/04/2023] [Indexed: 10/19/2023] Open
Abstract
Diabetes mellitus (DM) is a metabolic inflammatory disease with a high incidence worldwide. Patients with DM are at a high risk for all types of infections. Type 1 DM is characterised with immune destruction of pancreatic β cells, while type 2 diabetes is characterised with insulin resistance and β cell dysfunction, both of which result in disorders of glucose and lipid metabolism. This metabolic disorder causes functional defects of immune cells, aberrant production of inflammatory cytokines, dysregulated immune responses, advanced pathophysiological injury of the body, and increased mortality in populations with DM upon infections. Starting with the change of natural immune system in patients with DM, this paper focused on the enhanced severity of infections in DM and the underlying innate immune alterations in preclinical and clinical studies, aiming to better understand the influence of DM on the susceptibility, pathophysiology, and clinical outcomes in infections.
Collapse
Affiliation(s)
- Zi Zhou
- Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China
| | - Hao Wang
- Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China
| | - Sipin Tan
- Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China
| | - Huali Zhang
- Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China
| | - Yaxi Zhu
- Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China
| |
Collapse
|
14
|
Wang X, Miao S, Yang Y, Yang Q, Meng D, Liang H. Association of early dietary fiber intake and mortality in septic patients with mechanical ventilation based on MIMIC IV 2.1 database: a cohort study. Nutr J 2024; 23:1. [PMID: 38167155 PMCID: PMC10762999 DOI: 10.1186/s12937-023-00894-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Whether early dietary fiber intake in septic patients is associated with a better clinical prognosis remains unclear, especially the time and the amount. Therefore, we assessed the association between early dietary fiber intake and clinical outcomes in septic patients by examining an extensive database. METHODS We conducted a retrospective cohort study using data from the MIMIC IV 2.1 database, focusing on consecutive septic patients requiring mechanical ventilation in medical or mixed medical-surgical ICUs. We collected patient demographics and nutritional data. Dietary fiber amounts were calculated according to enteral nutrition instructions from manufacturers within the first 72 h after admission. After adjusting for covariates, we employed restricted cubic spline (RCS) regression to investigate the relationship between fiber intake (FI) and 28-day mortality. Patients were categorized into three groups based on their fiber index (FI) within 72 h of admission: low fiber index (LFI) group when FI was < 3 g/(%), medium fiber index (MFI) group when FI ranged from 3 to 35 g(%), and high fiber index (HFI) group when FI ≥ 35 g(%). Univariate and multivariate Cox proportional hazards regression models were utilized to assess the association between early FI and 28-day mortality. We ultimately employed Kaplan-Meier (KM) curves and log-rank test visually represent the association between FI and 90-day mortality. The second outcomes include ICU-acquired infections and the hospital and ICU death, length of hospital and ICU stay, and length of mechanical ventilation. RESULTS Among 1057 subjects, 562 (53.2%) were male, with a median age of 64.8 years (IQR 53.4-75.2). We observed a J-shaped relationship between FI and 28-day mortality. The MFI group exhibited the lowest 28-day mortality [adjusted HR 0.64 (0.45-0.91), p = 0.013] and the lowest rate of hospital mortality [adjusted OR 0.60 (0.39-0.93), p = 0.022], with no statistically significant differences noted in the HFI group when compared to the LFI group. Similar patterns were observed for 60-day and 90-day mortality. However, no statistically significant differences were observed in other secondary outcomes after adjusting for covariates. CONCLUSION Early medium fiber index intake improved 28-day mortality and lower hospital mortality in septic M/SICU patients on mechanical ventilation.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Clinical Nutrition, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Shuchuan Miao
- Department of Neurosurgery, Chengdu Seventh People's Hospital, Chengdu, Sichuan Province, China.
| | - Yuanwei Yang
- Department of Intensive Care Unit, Affiliated Minshan Hospital of Chengdu Medical College, Ya'an, Sichuan Province, China
| | - Qilin Yang
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dejiao Meng
- Department of Clinical Nutrition, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Hong Liang
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
15
|
Choi SH, Kim SY, Kim KM, Mony TJ, Bae HJ, Kim MS, Lee CH, Choi SE, Lee SH, Park SJ. Fermented Sprouts of Codonopsis lanceolata Suppress LPS-Induced Inflammatory Responses by Inhibiting NF-κB Signaling Pathway in RAW 264.7 Macrophages and CD1 Mice. Pharmaceutics 2023; 15:1793. [PMID: 37513980 PMCID: PMC10384864 DOI: 10.3390/pharmaceutics15071793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
The interest in bioconversion through fermentation of sprouts produced in smart farms is increasing due to their potential health benefits. Codonopsis lanceolata (CL) is reported to alleviate inflammatory conditions, but much research is still needed to determine which types and parts of CL are most effective. This study investigated the anti-inflammatory effects of a fermented extract of CL sprouts' aerial part (F-CSA) against LPS-stimulated RAW 264.7 macrophages and mice. In the screening test, F-CSA showed the most substantial anti-inflammatory effect among several samples, containing the highest total flavonoids, tannins, and polyphenols. UPLC-ESI-Q/TOF-MS and HPLC analysis revealed that F-CSA had the highest amount of luteolin among all the CL samples analyzed. F-CSA reduced the release of inflammatory cytokines and mediators such as NO and PGE2 by inhibiting the expression levels of iNOS and COX-2 in LPS-stimulated macrophages. Further, we found that the anti-inflammatory effects of F-CSA were mediated by inhibiting the JNK/NF-κB signaling pathway. Moreover, F-CSA improved survival rates and reduced plasma levels of NO and IL-6 in CD1 mice stimulated with LPS. These findings suggest that F-CSA, which contains luteolin, can alleviate inflammation in LPS-induced RAW 264.7 cells and a CD1 mouse model by inhibiting the JNK/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Seung-Hyuk Choi
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - So-Yeon Kim
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Kyeong-Min Kim
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Tamanna Jahan Mony
- Agriculture and Life Science Research Institute, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ho Jung Bae
- Agriculture and Life Science Research Institute, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Min Seok Kim
- Department of Forest Biomaterials Engineering, College of Forest and Environmental Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Chan Ho Lee
- Department of Forest Biomaterials Engineering, College of Forest and Environmental Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Sun-Eun Choi
- Department of Forest Biomaterials Engineering, College of Forest and Environmental Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Sang Ho Lee
- College of Pharmacy, Jeju National University, Jeju 63243, Republic of Korea
| | - Se Jin Park
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Republic of Korea
- Agriculture and Life Science Research Institute, Kangwon National University, Chuncheon 24341, Republic of Korea
- School of Natural Resources and Environmental Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
16
|
Brandão LEM, Popa A, Cedernaes E, Cedernaes C, Lampola L, Cedernaes J. Exposure to a more unhealthy diet impacts sleep microstructure during normal sleep and recovery sleep: A randomized trial. Obesity (Silver Spring) 2023. [PMID: 37245331 DOI: 10.1002/oby.23787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/01/2023] [Accepted: 03/23/2023] [Indexed: 05/30/2023]
Abstract
OBJECTIVE Although intake of specific macronutrients has been associated with sleep parameters, interventional evidence is lacking. Therefore, this randomized trial was conducted to examine how a more unhealthy high-fat/high-sugar (HFHS) diet impacts sleep in humans. METHODS In a crossover study, 15 healthy young men consumed two isocaloric diets in random order for a week: an HFHS and a low-fat/low-sugar diet. Following each diet, in-lab sleep was recorded using polysomnography during a full night of sleep and during recovery sleep after extended wakefulness. Sleep duration, macrostructure, and microstructure (oscillatory pattern and slow waves) were investigated using machine learning-based algorithms. RESULTS Sleep duration did not differ across the diets based on actigraphy and the in-lab polysomnography. Sleep macrostructure was similar after 1 week on each diet. Compared with the low-fat/low-sugar diet, consumption of the HFHS diet resulted in reduced delta power, delta to beta ratio, and slow wave amplitude but increased alpha and theta power during deep sleep. During recovery sleep, similar sleep oscillatory changes were observed. CONCLUSIONS Short-term consumption of a more unhealthy diet alters sleep oscillatory features that regulate the restorative properties of sleep. Whether such changes can mediate adverse health outcomes associated with consumption of an unhealthier diet warrants investigation.
Collapse
Affiliation(s)
| | - Alexandru Popa
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | | | - Lauri Lampola
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Jonathan Cedernaes
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
17
|
Tsai HH, Yu JC, Hsu HM, Chu CH, Chang TM, Hong ZJ, Feng AC, Fu CY, Hsu KF, Dai MS, Liao GS. The Risk of Breast Cancer between Western and Mediterranean Dietary Patterns. Nutrients 2023; 15:2057. [PMID: 37432206 DOI: 10.3390/nu15092057] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 07/12/2023] Open
Abstract
Breast cancer is a significant public health problem globally and prevention strategies have become of great interest as its incidence rises. Exploring the connection between dietary patterns and the reduction of breast cancer risk is considered a promising approach. High levels of fiber, phytochemicals, a good antioxidant profile, and a composition of advantageous fatty acids are characteristics of healthy dietary programs such as the Mediterranean diet. This review summarized and discussed the active compounds that are considered important in preventing breast cancer, including dietary components from recent related reports. These include polyunsaturated fatty acids, fiber, phytochemicals, and alcohol. Although the exact mechanism for preventing breast cancer using these dietary factors is not well understood, the combination of all the elements in a healthy diet plays a role in reducing breast cancer risk. Considering the elevated probability of breast cancer relapse and mortality, it is crucial to investigate the correlation between a nutritious dietary pattern and breast cancer, while identifying bioactive components that have the potential to mitigate the risk of breast cancer incidence.
Collapse
Affiliation(s)
- Hsueh-Han Tsai
- Division of General Surgery, Department of Surgery, Tri-Services General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Jyh-Cherng Yu
- Division of General Surgery, Department of Surgery, Tri-Services General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Huan-Ming Hsu
- Division of General Surgery, Department of Surgery, Tri-Services General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chi-Hong Chu
- Division of General Surgery, Department of Surgery, Tri-Services General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Tzu-Ming Chang
- Division of General Surgery, Department of Surgery, Tri-Services General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Zhi-Jie Hong
- Division of General Surgery, Department of Surgery, Tri-Services General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - An-Chieh Feng
- Division of General Surgery, Department of Surgery, Tri-Services General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chun-Yu Fu
- Division of General Surgery, Department of Surgery, Tri-Services General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Kuo-Feng Hsu
- Division of General Surgery, Department of Surgery, Tri-Services General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Ming-Shen Dai
- Division of Hematology/Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Guo-Shiou Liao
- Division of General Surgery, Department of Surgery, Tri-Services General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| |
Collapse
|
18
|
Hirschberger S, Schmid A, Kreth S. [Immunomodulation by nutritional intervention in critically ill patients]. DIE ANAESTHESIOLOGIE 2023; 72:229-244. [PMID: 36797533 PMCID: PMC9934515 DOI: 10.1007/s00101-023-01258-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 01/11/2023] [Indexed: 04/12/2023]
Abstract
Critically ill patients often suffer from a complex and severe immunological dysfunction. The differentiation and function of human immune cells are fundamentally controlled through metabolic processes. New concepts of immunonutrition therefore try to use enteral and parenteral nutrition to positively impact on the immune function of intensive care unit patients. This review article concisely presents the currently available evidence on the commonly used isolated supplements (anti-oxidative substances, amino acids, essential fatty acids) and difficulties related to their clinical use. The second part presents new and more comprehensive concepts of immunonutrition to influence the intestinal microbiome and to modulate the macronutrient composition. Immunonutrition of critically ill patients bears enormous potential and could become a valuable clinical tool for modulation of the immunometabolism of intensive care unit patients.
Collapse
Affiliation(s)
- Simon Hirschberger
- Klinik für Anaesthesiologie, LMU Klinikum München, München, Deutschland
- Walter-Brendel-Zentrum für experimentelle Medizin, Ludwig-Maximilians-Universität München (LMU), Marchioninistr. 68, 81377, München, Deutschland
| | - Annika Schmid
- Klinik für Anaesthesiologie, LMU Klinikum München, München, Deutschland
- Walter-Brendel-Zentrum für experimentelle Medizin, Ludwig-Maximilians-Universität München (LMU), Marchioninistr. 68, 81377, München, Deutschland
| | - Simone Kreth
- Klinik für Anaesthesiologie, LMU Klinikum München, München, Deutschland.
- Walter-Brendel-Zentrum für experimentelle Medizin, Ludwig-Maximilians-Universität München (LMU), Marchioninistr. 68, 81377, München, Deutschland.
| |
Collapse
|
19
|
Seufert AL, Napier BA. A new frontier for fat: dietary palmitic acid induces innate immune memory. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00021. [PMID: 37197687 PMCID: PMC10184819 DOI: 10.1097/in9.0000000000000021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/20/2023] [Indexed: 05/19/2023]
Abstract
Dietary saturated fats have recently been appreciated for their ability to modify innate immune cell function, including monocytes, macrophages, and neutrophils. Many dietary saturated fatty acids (SFAs) embark on a unique pathway through the lymphatics following digestion, and this makes them intriguing candidates for inflammatory regulation during homeostasis and disease. Specifically, palmitic acid (PA) and diets enriched in PA have recently been implicated in driving innate immune memory in mice. PA has been shown to induce long-lasting hyper-inflammatory capacity against secondary microbial stimuli in vitro and in vivo, and PA-enriched diets alter the developmental trajectory of stem cell progenitors in the bone marrow. Perhaps the most relevant finding is the ability of exogenous PA to enhance clearance of fungal and bacterial burdens in mice; however, the same PA treatment enhances endotoxemia severity and mortality. Westernized countries are becoming increasingly dependent on SFA-enriched diets, and a deeper understanding of SFA regulation of innate immune memory is imperative in this pandemic era.
Collapse
Affiliation(s)
- Amy L. Seufert
- Department of Biology and Center for Life in Extreme Environments, Portland State University, Portland, OR, USA
| | - Brooke A. Napier
- Department of Biology and Center for Life in Extreme Environments, Portland State University, Portland, OR, USA
- *Correspondence: Brooke A. Napier, E-mail:
| |
Collapse
|
20
|
Yu Y, Li X, Han S, Zhang J, Wang J, Chai J. miR-181c, a potential mediator for acute kidney injury in a burn rat model with following sepsis. Eur J Trauma Emerg Surg 2023; 49:1035-1045. [PMID: 36227355 DOI: 10.1007/s00068-022-02124-5] [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: 05/05/2022] [Accepted: 09/27/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND The miRNA profile is changed after burn or sepsis and is involved in regulating inflammatory reactions. However, the function and molecular mechanism of miRNAs in regulating burn sepsis-induced acute kidney injury (AKI) are still unclear. METHODS In this study, animal and cell sepsis models were established after burned rats were injected with lipopolysaccharide (LPS) or NRK-52E cells treated with LPS, respectively. Cytokine expression, inflammatory cell infiltration, serum creatinine (Scr) and kidney injury molecule-1 (KIM-1) levels were analysed after the indicated treatments. RESULTS Burn sepsis increased the expression of inflammatory factors (TNF-α and IL-1β) and chemokines (MIP-1α, MIP-2 and MCP-1). Moreover, burn sepsis promoted macrophage and neutrophil infiltration into the kidney and upregulated the levels of Scr and KIM-1 in the kidney and urine. Ectopic expression of miR-181c significantly reduced LPS-induced TLR4 protein expression, suppressed KIM-1 mRNA levels and subsequently inhibited the activation of inflammatory genes (TNF-α and IL-1β) and chemokine genes (MIP-1α, MIP-2 and MCP-1). CONCLUSIONS Our results demonstrated that miR-181c could suppress TLR4 expression, reduce inflammatory factor and chemokine secretion, mitigate inflammatory cell infiltration into the kidney and downregulate KIM-1 expression, which might ultimately attenuate burn sepsis-induced AKI.
Collapse
Affiliation(s)
- Yonghui Yu
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, 11 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Xiao Li
- The Fourth Medical Center of PLA General Hospital, 51 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Shaofang Han
- The Fourth Medical Center of PLA General Hospital, 51 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Jingjie Zhang
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, 11 Fucheng Road, Haidian District, Beijing, 100048, China.
| | - Jing Wang
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, 11 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Jiake Chai
- The Fourth Medical Center of PLA General Hospital, 51 Fucheng Road, Haidian District, Beijing, 100048, China.
| |
Collapse
|
21
|
Gomes SV, Dias BV, Júnior PAM, Pereira RR, de Souza DMS, Breguez GS, de Lima WG, Magalhães CLDB, Cangussú SD, Talvani A, Queiroz KB, Calsavara AJC, Costa DC. High-fat diet increases mortality and intensifies immunometabolic changes in septic mice. J Nutr Biochem 2023; 116:109315. [PMID: 36921735 DOI: 10.1016/j.jnutbio.2023.109315] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/21/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023]
Abstract
Immunometabolic changes in the liver and white adipose tissue (WAT) caused by high-fat (HF) diet intake may worse metabolic adaptation and protection against pathogens in sepsis. We investigate the effect of chronic HF diet (15 weeks) on mortality and immunometabolic responses in female mice after sepsis induced by cecum ligation and perforation (CLP). At week 14, animals were divided into four groups: sham C diet (C-Sh), sepsis C diet (C-Sp), sham HF diet (HF-Sh) and sepsis HF diet (HF-Sp). The surviving animals were euthanised on the 7th day. The HF diet decreased survival rate (58.3% vs 76.2% C-Sp group), increased serum cytokine storm (IL-6 (1.41 ×; vs HF-Sh), IL-1β (1.37 ×; vs C-Sp), TNF (1.34 ×; vs C-Sp and 1.72 ×; vs HF-Sh), IL-17 (1.44 ×; vs HF-Sh), IL-10 (1.55 ×; vs C-Sp and 1.41 ×; HF-Sh), WAT inflammation (IL-6 (8.7 ×; vs C-Sp and 2.4 ×; vs HF-Sh), TNF (5 ×; vs C-Sp and 1.7 ×;vs HF-Sh), IL-17 (1.7 ×; vs C-Sp), IL-10 (7.4 ×; vs C-Sp and 1.3 ×; vs HF-Sh), and modulated lipid metabolism in septic mice. In the HF-Sp group liver's, we observed hepatomegaly, hydropic degeneration, necrosis, an increase in oxidative stress (reduction of CAT activity (-81.7%; vs HF-Sh); increase MDA levels (82.8%; vs HF-Sh), and hepatic IL-6 (1.9 ×; vs HF-Sh), and TNF (1.3 × %;vs HF-Sh) production. Furthermore, we found a decrease in the total number of inflammatory, mononuclear cells, and in the regenerative processes, and binucleated hepatocytes in a HF-Sp group liver's. Our results suggested that the organism under metabolic stress of a HF diet during sepsis may worsen the inflammatory landscape and hepatocellular injury and may harm the liver regenerative process.
Collapse
Affiliation(s)
- Sttefany Viana Gomes
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Bruna Vidal Dias
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Pedro Alves Machado Júnior
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Renata Rebeca Pereira
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Débora Maria Soares de Souza
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Gustavo Silveira Breguez
- Multiuser Research Laboratory, School of Nutrition, School of Nutrition, Postgraduate Program in Health and Nutrition, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Wanderson Geraldo de Lima
- Morphopathology Laboratory, Department of Biological Sciences (DECBI), Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Cintia Lopes de Brito Magalhães
- Laboratory of Biology and Technology of Microorganisms (LBTM), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Silvia Dantas Cangussú
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Karina Barbosa Queiroz
- Laboratory of Experimental Nutrition (LABNEx), Department of Food, Postgraduate Program in Health and Nutrition, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Allan Jefferson Cruz Calsavara
- Laboratory of Cognition and Health (LACOS), School of Medicine, Department of Pediatric and Adult Clinics (DECPA), Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Daniela Caldeira Costa
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil.
| |
Collapse
|
22
|
Muske J, Knoop K. Contributions of the microbiota to the systemic inflammatory response. MICROBIOTA AND HOST 2023; 1:e230018. [PMID: 38872988 PMCID: PMC11170979 DOI: 10.1530/mah-23-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
The health of the intestinal microbiota impacts tolerance at homeostasis and the strength of the inflammation response during acute bloodstream infections. A complete understanding of the feedback loop between systemic inflammation and dysregulation of the gut microbiota is necessary for inflammation management. Here we will review the many ways in which the microbiota can influence the systemic pro-inflammatory response. Short-chain fatty acids, produced through the microbial metabolism of dietary fibers, can suppress inflammation systemically; in the absence of a balanced diet or disruption of the microbiota through antibiotics, there is disrupted metabolite production, leading to systemic inflammation. Dysbiosis or inflammation in the intestines can lead to a breakdown of the sturdy intestinal-epithelial barrier. When this barrier is perturbed, immunogenic lipopolysaccharides or extracellular vesicles enter the bloodstream and induce excessive inflammation. Necessary clinical treatments, such as antifungals or antibacterials, induce microbiota dysregulation and thus increased risk of endotoxemia; though probiotics may aid in improving the microbiota health and have been shown to deflate inflammation during sepsis. Within this complicated relationship: What is in control, the dysbiotic microbiota or the systemic inflammation?
Collapse
Affiliation(s)
- Josey Muske
- Mayo Graduate School of Biomedical Sciences
- Department of Immunology, Mayo Clinic Rochester, MN USA
| | - Kathryn Knoop
- Department of Immunology, Mayo Clinic Rochester, MN USA
- Department of Pediatrics, Mayo Clinic Rochester, MN USA
| |
Collapse
|
23
|
Li C, Dai J, Liu C, Dong G, Zhang X, Zhang J, Yan F, Zhang H, Wang C, Zhao M, Ning Z, Ma Q, Shi H, Li Z, Xiong H. Pyruvate Dehydrogenase Kinase 2 Accelerates Endotoxin Shock by Promoting Mitogen-Activated Protein Kinase Activation. Inflammation 2023; 46:418-431. [PMID: 36171490 DOI: 10.1007/s10753-022-01744-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/26/2022]
Abstract
Endotoxin shock remains one of the major causes of mortality worldwide. Pyruvate dehydrogenase kinase (PDK) 2 is an important regulatory enzyme involved in glucose metabolism. The purpose of this study was to determine the regulatory effect of PDK2 on LPS-induced endotoxin shock and explore the mechanisms in vivo and in vitro. Here, we showed that PDK2 contributed to Toll-like receptor (TLR)-mediated inflammation. Lipopolysaccharide (LPS) activation of TLR4 pathways resulted in PDK2 upregulation in macrophages and dendritic cells (DCs). PDK2 overexpression enhanced TLR4 signaling pathway activation, whereas downregulating PDK2 expression inhibited TLR4 signaling pathway activation. Pharmacological inhibition of PDK2 significantly decreased the mortality rate and alleviated pathological injury in the lungs and livers of LPS-challenged mice, while significantly suppressing proinflammatory cytokine production. Thus, we confirmed that PDK2 is involved in LPS-induced endotoxin shock by modulating TLR4-mitogen-activated protein kinase signaling and inducing the production of proinflammatory cytokines in macrophages and DCs. Our findings highlight the importance of PDK2 as a novel target to treat septic shock.
Collapse
Affiliation(s)
- Chunxia Li
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, 272067, Shandong, China
| | - Jun Dai
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, 272067, Shandong, China
| | - Chuanbin Liu
- Department of Pediatric Dentistry, Jining Stomatological Hospital, Jining, 272067, Shandong, China
| | - Guanjun Dong
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, 272067, Shandong, China
| | - Xin Zhang
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, 272067, Shandong, China
| | - Junfeng Zhang
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, 272067, Shandong, China
| | - Fenglian Yan
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, 272067, Shandong, China
| | - Hui Zhang
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, 272067, Shandong, China
| | - Changying Wang
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, 272067, Shandong, China
| | - Mingsheng Zhao
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, 272067, Shandong, China
| | - Zhaochen Ning
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, 272067, Shandong, China
| | - Qun Ma
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, 272067, Shandong, China
| | - Hui Shi
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, 272067, Shandong, China
| | - Zhihua Li
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, 272067, Shandong, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, 272067, Shandong, China.
| |
Collapse
|
24
|
Zhao C, Bao L, Zhao Y, Wu K, Qiu M, Feng L, Zhang N, Hu X, Fu Y. A fiber-enriched diet alleviates Staphylococcus aureus-induced mastitis by activating the HDAC3-mediated antimicrobial program in macrophages via butyrate production in mice. PLoS Pathog 2023; 19:e1011108. [PMID: 36656870 PMCID: PMC9888710 DOI: 10.1371/journal.ppat.1011108] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/31/2023] [Accepted: 01/09/2023] [Indexed: 01/20/2023] Open
Abstract
Mounting evidence suggests that the gut microbiota plays an important role in the pathogenesis of mastitis, an important disease affecting the health of lactating women and the development of the dairy industry. However, the effect of the regulation of the gut microbiota by dietary components on mastitis development remains unknown. In this study, we found that a fiber-enriched diet alleviated Staphylococcus aureus (S. au)-induced mastitis in mice, which was dependent on the gut microbiota as depletion of the gut microbiota by antibiotics abolished this protective effect. Likewise, fecal microbiota transplantation (FMT) from high-inulin (HI)-treated mice (HIF) to recipient mice improved S. au-induced mastitis in mice. Consumption of an HI diet and HIF increased fecal short-chain fatty acid (SCFA) levels compared with the control group. Moreover, treatment with SCFAs, especially butyrate, alleviated S. au-induced mastitis in mice. Mechanistically, consumption of an HI diet enhanced the host antimicrobial program in macrophages through inhibiting histone deacetylase 3 by the production of butyrate. Collectively, our results suggest that modulation of the gut microbiota and its metabolism by dietary components is a potential strategy for mastitis intervention and serve as a basis for other infectious diseases.
Collapse
Affiliation(s)
- Caijun Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Lijuan Bao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Yihong Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Keyi Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Min Qiu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Lianjun Feng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Xiaoyu Hu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China,* E-mail: (XH); (YF)
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China,* E-mail: (XH); (YF)
| |
Collapse
|
25
|
Douglas GL, DeKerlegand D, Dlouhy H, Dumont-Leblond N, Fields E, Heer M, Krieger S, Mehta S, Rooney BV, Torralba MG, Whiting SE, Crucian B, Lorenzi H, Smith SM, Young M, Zwart SR. Impact of diet on human nutrition, immune response, gut microbiome, and cognition in an isolated and confined mission environment. Sci Rep 2022; 12:20847. [PMID: 36522361 PMCID: PMC9755260 DOI: 10.1038/s41598-022-21927-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 10/06/2022] [Indexed: 12/23/2022] Open
Abstract
Long-duration spaceflight impacts human physiology, including well documented immune system dysregulation. The space food system has the potential to serve as a countermeasure to maladaptive physiological changes during spaceflight. However, the relationship between dietary requirements, the food system, and spaceflight adaptation requires further investigation to adequately define countermeasures and prioritize resources on future spaceflight missions. We evaluated the impact of an enhanced spaceflight diet, with increased quantity and variety of fruits, vegetables, fish, and other foods rich in flavonoids and omega-3 fatty acids, compared to a standard spaceflight diet on multiple health and performance outcomes in 16 subjects over four 45-day closed chamber missions in the NASA Human Exploration Research Analog (HERA). Subjects consuming the enhanced spaceflight diet had lower cholesterol levels, lower stress (i.e. cortisol levels), better cognitive speed, accuracy, and attention, and a more stable microbiome and metatranscriptome than subjects consuming the standard diet. Although no substantial changes were observed in the immune response, there were also no immune challenges, such as illness or infection, so the full benefits of the diet may not have been apparent in these analog missions. These results indicate that a spaceflight diet rich in fruits, vegetables, and omega-3 fatty acids produces significant health and performance benefits even over short durations. Further investigation is required to fully develop dietary countermeasures to physiological decrements observed during spaceflight. These results will have implications for food resource prioritization on spaceflight missions.
Collapse
Affiliation(s)
- Grace L. Douglas
- grid.419085.10000 0004 0613 2864Human Health and Performance Directorate (SF4), NASA Johnson Space Center, 2101 NASA Parkway, Houston, TX 77058 USA
| | | | - Holly Dlouhy
- grid.481680.30000 0004 0634 8729KBR, Houston, TX USA
| | - Nathan Dumont-Leblond
- grid.421142.00000 0000 8521 1798Centre de Recherche de L’Institut Universitaire de Cardiologie Et de Pneumologie de Québec, Quebec City, QC Canada
| | | | - Martina Heer
- grid.10388.320000 0001 2240 3300IU International University of Applied Sciences and University of Bonn, Bonn, Germany
| | | | | | | | | | | | - Brian Crucian
- grid.419085.10000 0004 0613 2864Human Health and Performance Directorate (SF4), NASA Johnson Space Center, 2101 NASA Parkway, Houston, TX 77058 USA
| | - Hernan Lorenzi
- grid.469946.0J Craig Venter Institute, Rockville, MD USA
| | - Scott M. Smith
- grid.419085.10000 0004 0613 2864Human Health and Performance Directorate (SF4), NASA Johnson Space Center, 2101 NASA Parkway, Houston, TX 77058 USA
| | - Millennia Young
- grid.419085.10000 0004 0613 2864Human Health and Performance Directorate (SF4), NASA Johnson Space Center, 2101 NASA Parkway, Houston, TX 77058 USA
| | - Sara R. Zwart
- grid.176731.50000 0001 1547 9964University of Texas Medical Branch, Galveston, TX USA
| |
Collapse
|
26
|
Ali Q, Ma S, Farooq U, Niu J, Li F, Li D, Wang Z, Sun H, Cui Y, Shi Y. Pasture intake protects against commercial diet-induced lipopolysaccharide production facilitated by gut microbiota through activating intestinal alkaline phosphatase enzyme in meat geese. Front Immunol 2022; 13:1041070. [PMID: 36569878 PMCID: PMC9774522 DOI: 10.3389/fimmu.2022.1041070] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction Diet strongly affects gut microbiota composition, and gut bacteria can influence the intestinal barrier functions and systemic inflammation through metabolic endotoxemia. In-house feeding system (IHF, a low dietary fiber source) may cause altered cecal microbiota composition and inflammatory responses in meat geese via increased endotoxemia (lipopolysaccharides) with reduced intestinal alkaline phosphatase (ALP) production. The effects of artificial pasture grazing system (AGF, a high dietary fiber source) on modulating gut microbiota architecture and gut barrier functions have not been investigated in meat geese. Therefore, this study aimed to investigate whether intestinal ALP could play a critical role in attenuating reactive oxygen species (ROS) generation and ROS facilitating NF-κB pathway-induced systemic inflammation in meat geese. Methods The impacts of IHF and AGF systems on gut microbial composition via 16 sRNA sequencing were assessed in meat geese. The host markers analysis through protein expression of serum and cecal tissues, hematoxylin and eosin (H&E) staining, localization of NF-қB and Nrf2 by immunofluorescence analysis, western blotting analysis of ALP, and quantitative PCR of cecal tissues was evaluated. Results and Discussion In the gut microbiota analysis, meat geese supplemented with pasture showed a significant increase in commensal microbial richness and diversity compared to IHF meat geese demonstrating the antimicrobial, antioxidant, and anti-inflammatory ability of the AGF system. A significant increase in intestinal ALP-induced Nrf2 signaling pathway was confirmed representing LPS dephosphorylation mediated TLR4/MyD88 induced ROS reduction mechanisms in AGF meat geese. Further, the correlation analysis of top 44 host markers with gut microbiota showed that artificial pasture intake protected gut barrier functions via reducing ROS-mediated NF-κB pathway-induced gut permeability, systemic inflammation, and aging phenotypes. In conclusion, the intestinal ALP functions to regulate gut microbial homeostasis and barrier function appear to inhibit pro-inflammatory cytokines by reducing LPS-induced ROS production in AGF meat geese. The AGF system may represent a novel therapy to counteract the chronic inflammatory state leading to low dietary fiber-related diseases in animals.
Collapse
Affiliation(s)
- Qasim Ali
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China
| | - Sen Ma
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Henan Agricultural University, Zhengzhou, Henan, China,Henan Herbage Engineering Technology Research Center, Henan Agricultural University, Zhengzhou, Henan, China
| | - Umar Farooq
- Department of Poultry Science, University of Agriculture Faisalabad, Toba Tek Singh, Pakistan
| | - Jiakuan Niu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China
| | - Fen Li
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China
| | - Defeng Li
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Henan Agricultural University, Zhengzhou, Henan, China,Henan Herbage Engineering Technology Research Center, Henan Agricultural University, Zhengzhou, Henan, China
| | - Zhichang Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Henan Agricultural University, Zhengzhou, Henan, China,Henan Herbage Engineering Technology Research Center, Henan Agricultural University, Zhengzhou, Henan, China
| | - Hao Sun
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Henan Agricultural University, Zhengzhou, Henan, China,Henan Herbage Engineering Technology Research Center, Henan Agricultural University, Zhengzhou, Henan, China
| | - Yalei Cui
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Henan Agricultural University, Zhengzhou, Henan, China,Henan Herbage Engineering Technology Research Center, Henan Agricultural University, Zhengzhou, Henan, China
| | - Yinghua Shi
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Henan Agricultural University, Zhengzhou, Henan, China,Henan Herbage Engineering Technology Research Center, Henan Agricultural University, Zhengzhou, Henan, China,*Correspondence: Yinghua Shi,
| |
Collapse
|
27
|
Miyashita D, Inoue R, Tsuno T, Okuyama T, Kyohara M, Nakahashi-Oda C, Nishiyama K, Fukushima S, Inada Y, Togashi Y, Shibuya A, Terauchi Y, Shirakawa J. Protective effects of S100A8 on sepsis mortality: Links to sepsis risk in obesity and diabetes. iScience 2022; 25:105662. [PMID: 36505926 PMCID: PMC9732389 DOI: 10.1016/j.isci.2022.105662] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/23/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Obesity and diabetes are independent risk factors for death during sepsis. S100A8, an alarmin, is related to inflammation, obesity, and diabetes. Here, we examine the role of S100A8 in sepsis of obesity and diabetes models. Injection of S100A8 prolongs the survival of septic mice induced by lethal endotoxemia, Escherichia coli injection, or cecal ligation and puncture. S100A8 decrease the LPS-induced expression of proinflammatory cytokines in peritoneal macrophages by inhibiting TLR4-mediated signals in an autocrine manner. db/db, ob/ob, and western diet-fed mice demonstrate reduced upregulation of S100A8 induced by LPS treatment in both serum and peritoneal cells. These mice also show shorter survival after LPS injection, and S100A8 supplementation prolonged the survival. While myelomonocytic cells-specific S100A8-deficient mice (Lyz2 cre :S100A8 floxed/floxed ) exhibit shorter survival after LPS treatment, S100A8 supplementation prolonged the survival. Thus, myelomonocytic cell-derived S100A8 is crucial for protection from sepsis, and S100A8 supplementation improves sepsis, particularly in mice with obesity and diabetes.
Collapse
Affiliation(s)
- Daisuke Miyashita
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Ryota Inoue
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Takahiro Tsuno
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Tomoko Okuyama
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Mayu Kyohara
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Chigusa Nakahashi-Oda
- Department of Immunology, Faculty of Medicine, and R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Japan
| | - Kuniyuki Nishiyama
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Setsuko Fukushima
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
| | - Yutaro Inada
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Yu Togashi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, and R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Jun Shirakawa
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
- Corresponding author
| |
Collapse
|
28
|
Seufert AL, Hickman JW, Traxler SK, Peterson RM, Waugh TA, Lashley SJ, Shulzhenko N, Napier RJ, Napier BA. Enriched dietary saturated fatty acids induce trained immunity via ceramide production that enhances severity of endotoxemia and clearance of infection. eLife 2022; 11:e76744. [PMID: 36264059 PMCID: PMC9642993 DOI: 10.7554/elife.76744] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 10/19/2022] [Indexed: 11/25/2022] Open
Abstract
Trained immunity is an innate immune memory response that is induced by a primary inflammatory stimulus that sensitizes monocytes and macrophages to a secondary pathogenic challenge, reprogramming the host response to infection and inflammatory disease. Dietary fatty acids can act as inflammatory stimuli, but it is unknown if they can act as the primary stimuli to induce trained immunity. Here we find mice fed a diet enriched exclusively in saturated fatty acids (ketogenic diet; KD) confer a hyper-inflammatory response to systemic lipopolysaccharide (LPS) and increased mortality, independent of diet-induced microbiome and hyperglycemia. We find KD alters the composition of the hematopoietic stem cell compartment and enhances the response of bone marrow macrophages, monocytes, and splenocytes to secondary LPS challenge. Lipidomics identified enhanced free palmitic acid (PA) and PA-associated lipids in KD-fed mice serum. We found pre-treatment with physiologically relevant concentrations of PA induces a hyper-inflammatory response to LPS in macrophages, and this was dependent on the synthesis of ceramide. In vivo, we found systemic PA confers enhanced inflammation and mortality in response to systemic LPS, and this phenotype was not reversible for up to 7 days post-PA-exposure. Conversely, we find PA exposure enhanced clearance of Candida albicans in Rag1-/- mice. Lastly, we show that oleic acid, which depletes intracellular ceramide, reverses PA-induced hyper-inflammation in macrophages and enhanced mortality in response to LPS. These implicate enriched dietary SFAs, and specifically PA, in the induction of long-lived innate immune memory and highlight the plasticity of this innate immune reprogramming by dietary constituents.
Collapse
Affiliation(s)
- Amy L Seufert
- Department of Biology and Center for Life in Extreme Environments, Portland State UniversityPortlandUnited States
| | - James W Hickman
- Department of Biology and Center for Life in Extreme Environments, Portland State UniversityPortlandUnited States
| | - Ste K Traxler
- Department of Biology and Center for Life in Extreme Environments, Portland State UniversityPortlandUnited States
| | - Rachael M Peterson
- Department of Biology and Center for Life in Extreme Environments, Portland State UniversityPortlandUnited States
| | - Trent A Waugh
- Department of Biology and Center for Life in Extreme Environments, Portland State UniversityPortlandUnited States
| | | | - Natalia Shulzhenko
- Department of Biomedical Sciences, Oregon State UniversityCorvallisUnited States
| | - Ruth J Napier
- VA Portland Health Care SystemPortlandUnited States
- Department of Molecular Microbiology and Immunology, Oregon Health & Science UniversityPortlandUnited States
| | - Brooke A Napier
- Department of Biology and Center for Life in Extreme Environments, Portland State UniversityPortlandUnited States
| |
Collapse
|
29
|
Sperry MM, Novak R, Keshari V, Dinis ALM, Cartwright MJ, Camacho DM, Paré J, Super M, Levin M, Ingber DE. Enhancers of Host Immune Tolerance to Bacterial Infection Discovered Using Linked Computational and Experimental Approaches. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200222. [PMID: 35706367 PMCID: PMC9475558 DOI: 10.1002/advs.202200222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/10/2022] [Indexed: 05/31/2023]
Abstract
Current therapeutic strategies against bacterial infections focus on reduction of pathogen load using antibiotics; however, stimulation of host tolerance to infection in the presence of pathogens might offer an alternative approach. Computational transcriptomics and Xenopus laevis embryos are used to discover infection response pathways, identify potential tolerance inducer drugs, and validate their ability to induce broad tolerance. Xenopus exhibits natural tolerance to Acinetobacter baumanii, Klebsiella pneumoniae, Staphylococcus aureus, and Streptococcus pneumoniae bacteria, whereas Aeromonas hydrophila and Pseudomonas aeruginosa produce lethal infections. Transcriptional profiling leads to definition of a 20-gene signature that discriminates between tolerant and susceptible states, as well as identification of a more active tolerance response to gram negative compared to gram positive bacteria. Gene pathways associated with active tolerance in Xenopus, including some involved in metal ion binding and hypoxia, are found to be conserved across species, including mammals, and administration of a metal chelator (deferoxamine) or a HIF-1α agonist (1,4-DPCA) in embryos infected with lethal A. hydrophila increased survival despite high pathogen load. These data demonstrate the value of combining the Xenopus embryo infection model with computational multiomics analyses for mechanistic discovery and drug repurposing to induce host tolerance to bacterial infections.
Collapse
Affiliation(s)
- Megan M. Sperry
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
- Department of BiologyTufts UniversityMedfordMA02155USA
| | - Richard Novak
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
- Present address:
Unravel Biosciences, Inc.BostonMA02125USA
| | - Vishal Keshari
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Alexandre L. M. Dinis
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
- Present address:
University of Massachusetts Medical SchoolWorcesterMA01655USA
| | - Mark J. Cartwright
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Diogo M. Camacho
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
- Present address:
Rheos MedicinesCambridgeMA02142USA
| | - Jean‐François Paré
- Department of BiologyTufts UniversityMedfordMA02155USA
- Present address:
Queen's UniversityKingstonON K7L 3N6Canada
| | - Michael Super
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
| | - Michael Levin
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
- Department of BiologyTufts UniversityMedfordMA02155USA
- Allen Discovery Center at Tufts UniversityMedfordMA02155USA
| | - Donald E. Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
- Vascular Biology Program and Department of SurgeryBoston Children's Hospital and Harvard Medical SchoolBostonMA02115USA
- Harvard John A. Paulson School of Engineering and Applied SciencesCambridgeMA02138USA
| |
Collapse
|
30
|
Wu C, Liu J, Li Y, Wang N, Yan Q, Jiang Z. Manno-oligosaccharides from cassia seed gum ameliorate inflammation and improve glucose metabolism in diabetic rats. Food Funct 2022; 13:6674-6687. [PMID: 35647651 DOI: 10.1039/d1fo03057d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Functional oligosaccharides show anti-diabetic effects through inflammation regulation with improved glucose metabolism. In this study, novel prebiotics of manno-oligosaccharides from cassia seed gum (CMOS) were incorporated into the diet of streptozotocin (STZ) plus high-fat and high-sugar diet (HFSD)-induced rats. After feeding for 8 weeks, CMOS (300-1200 mg per kg b.w. per d) significantly ameliorated the fasting blood glucose level (7.1-8.2 mmol L-1) as compared with that of the model group (14.2 mmol L-1), where the area under the oral glucose tolerance test curve was decreased by 20.0%-24.5%. Meanwhile, CMOS prevented STZ plus HFSD-induced damage to islet tissue with a clear and integrated morphology and reduced the glucagon/insulin area ratio (by 97.9% for 300 mg per kg b.w. per d CMOS). CMOS also reduced metabolic endotoxemia and maintained intestinal integrity with recovered mRNA expression of Zo-1 and occludin to the normal comparable level. Upon 16S rDNA sequencing, it was found that CMOS regulated the microbiota composition in the cecum with an increased relative abundance of Bifidobacteria, while that of Shigella was decreased. The molecular mechanisms involved in the anti-diabetic effects of CMOS were further studied. CMOS reduced the mRNA expression of Tlr2 and Tlr4 in the intestines of STZ plus HFSD-induced rats. Meanwhile, Nlrp3 associated inflammasome activation in the intestine and liver with glucose metabolism disorder was inhibited by CMOS, resulting in reduced interleukin-1β secretion (by 38.8-46.4% for CMOS of 300-1200 mg per kg b.w. per d) and inflammation. Furthermore, CMOS regulated the AKT/IRS/AMPK signaling pathway and improved glucose metabolism in the liver. Findings obtained here implicated that CMOS could modulate metabolic-inflammation as a functional dietary supplement.
Collapse
Affiliation(s)
- Chenxuan Wu
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Jun Liu
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Yanxiao Li
- Department of Nutrition and Health, College of Engineering, China Agricultural University, Beijing 100083, China
| | - Nannan Wang
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Qiaojuan Yan
- Department of Nutrition and Health, College of Engineering, China Agricultural University, Beijing 100083, China
| | - Zhengqiang Jiang
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
31
|
Hirschberger S, Gellert L, Effinger D, Muenchhoff M, Herrmann M, Briegel JM, Zwißler B, Kreth S. Ketone Bodies Improve Human CD8+ Cytotoxic T-Cell Immune Response During COVID-19 Infection. Front Med (Lausanne) 2022; 9:923502. [PMID: 35783654 PMCID: PMC9243504 DOI: 10.3389/fmed.2022.923502] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/30/2022] [Indexed: 12/15/2022] Open
Abstract
Severe COVID-19 is characterized by profound CD8+ T-cell dysfunction, which cannot be specifically treated to date. We here investigate whether metabolic CD8+ T-cell reprogramming by ketone bodies could be a promising strategy to overcome the immunoparalysis in COVID-19 patients. This approach was triggered by our recent pioneering study, which has provided evidence that CD8+ T-cell capacity in healthy subjects could be significantly empowered by a Ketogenic Diet. These improvements were achieved by immunometabolic rewiring toward oxidative phosphorylation. We here report similar strengthening of CD8+ T cells obtained from severely diseased COVID-19 patients: Flow cytometry and ELISA revealed elevated cytokine expression and secretion (up to + 24%) upon ketone treatment and enhanced cell lysis capacity (+ 21%). Metabolic analyses using Seahorse technology revealed upregulated mitochondrial respiratory chain activity (+ 25%), enabling both superior energy supply (+ 44%) and higher mitochondrial reactive oxygen species signaling. These beneficial effects of ketones might represent evolutionary conserved mechanisms to strengthen human immunity. Our findings pave the road for metabolic treatment studies in COVID-19.
Collapse
Affiliation(s)
- Simon Hirschberger
- Research Unit Molecular Medicine, Department of Anaesthesiology, LMU University Hospital, Ludwig-Maximilian-University Munich (LMU), Munich, Germany
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilian-University Munich (LMU), Munich, Germany
| | - Luca Gellert
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilian-University Munich (LMU), Munich, Germany
| | - David Effinger
- Research Unit Molecular Medicine, Department of Anaesthesiology, LMU University Hospital, Ludwig-Maximilian-University Munich (LMU), Munich, Germany
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilian-University Munich (LMU), Munich, Germany
| | - Maximilian Muenchhoff
- Faculty of Medicine, National Reference Center for Retroviruses, Max von Pettenkofer Institute and Gene Center, Virology, Ludwig-Maximilian-University Munich (LMU), Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), LMU University Hospital, LMU Munich, Munich, Germany
| | - Markus Herrmann
- COVID-19 Registry of the LMU Munich (CORKUM), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Medicine III, LMU University Hospital, Ludwig-Maximilian-University Munich (LMU), Munich, Germany
| | - Josef-Maria Briegel
- Research Unit Molecular Medicine, Department of Anaesthesiology, LMU University Hospital, Ludwig-Maximilian-University Munich (LMU), Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), LMU University Hospital, LMU Munich, Munich, Germany
| | - Bernhard Zwißler
- Research Unit Molecular Medicine, Department of Anaesthesiology, LMU University Hospital, Ludwig-Maximilian-University Munich (LMU), Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), LMU University Hospital, LMU Munich, Munich, Germany
| | - Simone Kreth
- Research Unit Molecular Medicine, Department of Anaesthesiology, LMU University Hospital, Ludwig-Maximilian-University Munich (LMU), Munich, Germany
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilian-University Munich (LMU), Munich, Germany
- *Correspondence: Simone Kreth,
| |
Collapse
|
32
|
Merana GR, Dwyer LR, Dhariwala MO, Weckel A, Gonzalez JR, Okoro JN, Cohen JN, Tamaki CM, Han J, Tasoff P, Palacios-Calderon Y, Ha CWY, Lynch SV, Segre JA, Kong HH, Kattah MG, Ma A, Scharschmidt TC. Intestinal inflammation alters the antigen-specific immune response to a skin commensal. Cell Rep 2022; 39:110891. [PMID: 35649365 PMCID: PMC9248974 DOI: 10.1016/j.celrep.2022.110891] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 04/08/2022] [Accepted: 05/05/2022] [Indexed: 12/13/2022] Open
Abstract
Resident microbes in skin and gut predominantly impact local immune cell function during homeostasis. However, colitis-associated neutrophilic skin disorders suggest possible breakdown of this compartmentalization with disease. Using a model wherein neonatal skin colonization by Staphylococcus epidermidis facilitates generation of commensal-specific tolerance and CD4+ regulatory T cells (Tregs), we ask whether this response is perturbed by gut inflammation. Chemically induced colitis is accompanied by intestinal expansion of S. epidermidis and reduces gut-draining lymph node (dLN) commensal-specific Tregs. It also results in reduced commensal-specific Tregs in skin and skin-dLNs and increased skin neutrophils. Increased CD4+ circulation between gut and skin dLN suggests that the altered cutaneous response is initiated in the colon, and resistance to colitis-induced effects in Cd4creIl1r1fl/fl mice implicate interleukin (IL)-1 in mediating the altered commensal-specific response. These findings provide mechanistic insight into observed connections between inflammatory skin and intestinal diseases.
Collapse
Affiliation(s)
- Geil R Merana
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Sciences Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Laura R Dwyer
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Sciences Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Miqdad O Dhariwala
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Antonin Weckel
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeanmarie R Gonzalez
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Sciences Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Joy N Okoro
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jarish N Cohen
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Courtney M Tamaki
- Parnassus Flow Cytometry CoLab, University of California, San Francisco, San Francisco, 94143, USA
| | - Jungmin Han
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Preston Tasoff
- Benioff Center for Microbiome Medicine, Department of Medicine, University of California, San Francisco, CA 94143, USA
| | | | - Connie W Y Ha
- Benioff Center for Microbiome Medicine, Department of Medicine, University of California, San Francisco, CA 94143, USA
| | - Susan V Lynch
- Benioff Center for Microbiome Medicine, Department of Medicine, University of California, San Francisco, CA 94143, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Julia A Segre
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Heidi H Kong
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael G Kattah
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Averil Ma
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tiffany C Scharschmidt
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
33
|
Zou S, Jiao X, Liu J, Qi D, Pei X, Lu D, Huang S, Li Z. High-Fat Nutritional Challenge Reshapes Circadian Signatures in Murine Extraorbital Lacrimal Glands. Invest Ophthalmol Vis Sci 2022; 63:23. [PMID: 35588356 PMCID: PMC9123521 DOI: 10.1167/iovs.63.5.23] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose A high-fat diet (HFD) increases the risk of developing many systemic diseases; however, the effects of high fat intake on lacrimal gland functions and the molecular mechanisms underlying these effects are unknown. We explored the effects of an HFD on the circadian rhythms of the extraorbital lacrimal glands (ELGs). Methods Male C57BL/6J mice maintained on a 12/12-hour light/dark cycle were fed an ad libitum HFD or normal chow (NC) for 2 weeks. The ELGs were collected from euthanized animals every 3 hours throughout the circadian cycle (24 hours). Using high-throughput RNA-sequencing (RNA-Seq), we studied the circadian transcriptomic profile of the ELGs. Circadian oscillations in cell size, secretion response, lipid deposition, and immune cell trafficking of the ELGs were also analyzed. Results An HFD modulated the circadian transcriptomic profile of the ELGs, including the composition, phase, and amplitude of cyclical transcript oscillations, and affected the associated signaling pathways at spatiotemporal levels. HFD feeding significantly altered the normal rhythmic oscillations of ELG cell size, immune cell trafficking, secretion response, and lipid deposition. After dietary reversal in HFD-fed animals, the activity, core temperature, and lipid accumulation in lacrimal glands recovered partially to the level of NC-fed animals. However, the average cell size of the ELGs, the recruitment of immune cells, and the rhythm of lacrimal secretion did not return to the levels of the NC-fed group. Conclusions HFD perturbation interferes with the cyclical transcriptomic profile, cell size, immune cell trafficking, and secretion function of the ELGs with a strikingly high sensitivity.
Collapse
Affiliation(s)
- Sen Zou
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| | - Xinwei Jiao
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| | - Jiangman Liu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| | - Di Qi
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| | - Xiaoting Pei
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| | - Dingli Lu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| | - Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| | - Zhijie Li
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| |
Collapse
|
34
|
Abu Y, Vitari N, Yan Y, Roy S. Opioids and Sepsis: Elucidating the Role of the Microbiome and microRNA-146. Int J Mol Sci 2022; 23:1097. [PMID: 35163021 PMCID: PMC8835205 DOI: 10.3390/ijms23031097] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
Sepsis has recently been defined as life-threatening organ dysfunction caused by the dysregulated host response to an ongoing or suspected infection. To date, sepsis continues to be a leading cause of morbidity and mortality amongst hospitalized patients. Many risk factors contribute to development of sepsis, including pain-relieving drugs like opioids, which are frequently prescribed post-operatively. In light of the opioid crisis, understanding the interactions between opioid use and the development of sepsis has become extremely relevant, as opioid use is associated with increased risk of infection. Given that the intestinal tract is a major site of origin of sepsis-causing microbes, there has been an increasing focus on how alterations in the gut microbiome may predispose towards sepsis and mediate immune dysregulation. MicroRNAs, in particular, have emerged as key modulators of the inflammatory response during sepsis by tempering the immune response, thereby mediating the interaction between host and microbiome. In this review, we elucidate contributing roles of microRNA 146 in modulating sepsis pathogenesis and end with a discussion of therapeutic targeting of the gut microbiome in controlling immune dysregulation in sepsis.
Collapse
Affiliation(s)
- Yaa Abu
- Medical Scientist Training Program, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Nicolas Vitari
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Yan Yan
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Sabita Roy
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| |
Collapse
|
35
|
Zou ZY, Sun KJ, Fu G, Huang JJ, Yang ZJ, Zhou ZP, Ma SL, Zhu F, Wu M. Impact of early empirical antifungal therapy on prognosis of sepsis patients with positive yeast culture: A retrospective study from the MIMIC-IV database. Front Microbiol 2022; 13:1047889. [PMID: 36466647 PMCID: PMC9712452 DOI: 10.3389/fmicb.2022.1047889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Mortality and other clinical outcomes of culture-negative and culture-positive among patients with fungal sepsis have not been documented, and whether antifungal therapy prior to fungal culture reports is related to decreased mortality among patients remains largely controversial. This study aimed to determine the mortality and other clinical outcomes of patients with positive yeast cultures and further investigate the effects of initial empiric antifungal therapy. METHODS A retrospective study was conducted among septic patients using the Medical Information Mart for Intensive Care (MIMIC)-IV database. Patients with sepsis were divided into two groups based on first fungal culture status during intensive care unit (ICU) stay, and initial empirical antifungal therapy was prescribed based on physician's experience prior to fungal culture reports within 48 h. The primary outcome was in-hospital all-cause mortality. The secondary outcomes were 30-day all-cause mortality, 60-day all-cause mortality, length of ICU stay and length of hospital stay. Multivariate logistic regression, propensity score matching (PSM), subgroup analyses and survival curve analyses were performed. RESULTS This study included 18,496 sepsis patients, of whom 3,477 (18.8%) had positive yeast cultures. Patients with positive yeast cultures had higher in-hospital all-cause mortality, 60-day all-cause mortality, and longer lengths of ICU stay and hospital stay than those with negative yeast cultures after PSM (all p < 0.01). Multivariate logistic regression analysis revealed that positive yeast culture was a risk factor for in-hospital mortality in the extended model. Subgroup analyses showed that the results were robust among the respiratory infection, urinary tract infection, gram-positive bacterial infection and bacteria-free culture subgroups. Interestingly, empiric antifungal therapy was not associated with lower in-hospital mortality among patients with positive yeast cultures, mainly manifested in stratification analysis, which showed that antifungal treatment did not improve outcomes in the bloodstream infection (odds ratio, OR 2.12, 95% CI: 1.16-3.91, p = 0.015) or urinary tract infection groups (OR 3.24, 95% CI: 1.48-7.11, p = 0.003). CONCLUSION Culture positivity for yeast among sepsis patients was associated with worse clinical outcomes, and empiric antifungal therapy did not lower in-hospital all-cause mortality in the bloodstream infection or urinary tract infection groups in the ICU.
Collapse
Affiliation(s)
- Zhi-ye Zou
- Department of Critical Care Medicine and Hospital Infection Prevention and Control, Shenzhen Second People’s Hospital & First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen, China
| | - Kai-jun Sun
- Department of Cardiovascular Medicine, Weifang People's Hospital, Weifang, China
| | - Guang Fu
- Department of Gastroenterology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Jia-jia Huang
- Department of Critical Care Medicine and Hospital Infection Prevention and Control, Shenzhen Second People’s Hospital & First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen, China
- Shantou University Medical College, Shantou, China
| | - Zhen-jia Yang
- Department of Critical Care Medicine and Hospital Infection Prevention and Control, Shenzhen Second People’s Hospital & First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen, China
- Shantou University Medical College, Shantou, China
| | - Zhi-peng Zhou
- Department of Critical Care Medicine and Hospital Infection Prevention and Control, Shenzhen Second People’s Hospital & First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen, China
| | - Shao-lin Ma
- Department of Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Shao-lin Ma,
| | - Feng Zhu
- Department of Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Burn and Trauma ICU, The First Affiliated Hospital, Naval Medical University, Shanghai, China
- Feng Zhu,
| | - Ming Wu
- Department of Critical Care Medicine and Hospital Infection Prevention and Control, Shenzhen Second People’s Hospital & First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen, China
- Shantou University Medical College, Shantou, China
- Ming Wu,
| |
Collapse
|
36
|
He S, Xue J, Cao P, Hou J, Cui Y, Chang J, Huang L, Han Y, Duan X, Tan K, Fan Y. JNK/Itch Axis Mediates the Lipopolysaccharide-Induced Ubiquitin-Proteasome-Dependent Degradation of Ferritin Light Chain in Murine Macrophage Cells. Inflammation 2021; 45:1089-1100. [PMID: 34837126 DOI: 10.1007/s10753-021-01603-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/16/2021] [Indexed: 11/25/2022]
Abstract
Ferritin, which is composed of a heavy chain and a light chain, plays a critical role in maintaining iron homeostasis by sequestering iron. The ferritin light chain (FTL) is responsible for the stability of the ferritin complex. We have previously shown that overexpression of FTL decreases the levels of the labile iron pool (LIP) and reactive oxygen species (ROS) in lipopolysaccharide (LPS)-treated murine macrophage cells. The protein level of FTL was downregulated by LPS within a short treatment period. However, the mechanism underlying the LPS-induced changes in the FTL levels is not known. In the present study, we report that LPS induces the ubiquitin-proteasome-dependent degradation of FTL and that the mechanism of LPS-induced FTL degradation involves the JNK/Itch axis. We found that LPS downregulates the protein and mRNA levels of FTL in a time-dependent manner. The proteasome inhibitor MG-132 significantly reverses the LPS-induced decrease in FTL. Furthermore, we observed that LPS treatment cannot cause ubiquitination of the lysine site (K105 and K144) mutant of FTL. Interestingly, LPS-mediated ubiquitin-dependent degradation of FTL is significantly inhibited by the JNK-specific inhibitor SP600125. Moreover, LPS could upregulate the protein level of E3 ubiquitin ligase Itch, a substrate of JNK kinases. Immunoprecipitation analyses revealed an increase in the association of FTL with Itch, a substrate of JNK kinases, in response to LPS stimulation. SP600125 decreased LPS-induced Itch upregulation. Taken together, these results suggest that LPS stimulation leads to the degradation of FTL through the ubiquitin-proteasome proteolytic pathway, and this FTL degradation is mediated by the JNK/Itch axis in murine macrophage cells.
Collapse
Affiliation(s)
- Shufen He
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Hebei Province, Shijiazhuang, 050024, PR China
| | - Jianqi Xue
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Hebei Province, Shijiazhuang, 050024, PR China
| | - Pengxiu Cao
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Hebei Province, Shijiazhuang, 050024, PR China
| | - Jianyuan Hou
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Hebei Province, Shijiazhuang, 050024, PR China
| | - Yan Cui
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Hebei Province, Shijiazhuang, 050024, PR China
| | - Jing Chang
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Hebei Province, Shijiazhuang, 050024, PR China
| | - Liying Huang
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Hebei Province, Shijiazhuang, 050024, PR China
| | - Yu Han
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Hebei Province, Shijiazhuang, 050024, PR China
| | - Xianglin Duan
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Hebei Province, Shijiazhuang, 050024, PR China
| | - Ke Tan
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Hebei Province, Shijiazhuang, 050024, PR China.
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, PR China.
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, PR China.
| | - Yumei Fan
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Hebei Province, Shijiazhuang, 050024, PR China.
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, PR China.
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, PR China.
| |
Collapse
|
37
|
Saito S, Cao DY, Victor AR, Peng Z, Wu HY, Okwan-Duodu D. RASAL3 Is a Putative RasGAP Modulating Inflammatory Response by Neutrophils. Front Immunol 2021; 12:744300. [PMID: 34777356 PMCID: PMC8579101 DOI: 10.3389/fimmu.2021.744300] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
As first responder cells in host defense, neutrophils must be carefully regulated to prevent collateral tissue injury. However, the intracellular events that titrate the neutrophil’s response to inflammatory stimuli remain poorly understood. As a molecular switch, Ras activity is tightly regulated by Ras GTPase activating proteins (RasGAP) to maintain cellular active-inactive states. Here, we show that RASAL3, a RasGAP, is highly expressed in neutrophils and that its expression is upregulated by exogenous stimuli in neutrophils. RASAL3 deficiency triggers augmented neutrophil responses and enhanced immune activation in acute inflammatory conditions. Consequently, mice lacking RASAL3 (RASAL3-KO) demonstrate accelerated mortality in a septic shock model via induction of severe organ damage and hyperinflammatory response. The excessive neutrophilic hyperinflammation and increased mortality were recapitulated in a mouse model of sickle cell disease, which we found to have low neutrophil RASAL3 expression upon LPS activation. Thus, RASAL3 functions as a RasGAP that negatively regulates the cellular activity of neutrophils to modulate the inflammatory response. These results demonstrate that RASAL3 could serve as a therapeutic target to regulate excessive inflammation in sepsis and many inflammatory disease states.
Collapse
Affiliation(s)
- Suguru Saito
- Bio-fluid Biomarker Center, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Division of Virology, Department of Immunology and Infection, School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Duo-Yao Cao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Aaron R Victor
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Zhenzi Peng
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,College of Animal Science and Technology, Northwest A&F University, Shaanxi, China
| | - Hui-Ya Wu
- College of Health Science, Trans World University, Douliu, Taiwan
| | - Derick Okwan-Duodu
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
38
|
Malesza IJ, Malesza M, Walkowiak J, Mussin N, Walkowiak D, Aringazina R, Bartkowiak-Wieczorek J, Mądry E. High-Fat, Western-Style Diet, Systemic Inflammation, and Gut Microbiota: A Narrative Review. Cells 2021; 10:cells10113164. [PMID: 34831387 PMCID: PMC8619527 DOI: 10.3390/cells10113164] [Citation(s) in RCA: 369] [Impact Index Per Article: 92.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota is responsible for recovering energy from food, providing hosts with vitamins, and providing a barrier function against exogenous pathogens. In addition, it is involved in maintaining the integrity of the intestinal epithelial barrier, crucial for the functional maturation of the gut immune system. The Western diet (WD)—an unhealthy diet with high consumption of fats—can be broadly characterized by overeating, frequent snacking, and a prolonged postprandial state. The term WD is commonly known and intuitively understood. However, the strict digital expression of nutrient ratios is not precisely defined. Based on the US data for 1908–1989, the calory intake available from fats increased from 32% to 45%. Besides the metabolic aspects (hyperinsulinemia, insulin resistance, dyslipidemia, sympathetic nervous system and renin-angiotensin system overstimulation, and oxidative stress), the consequences of excessive fat consumption (high-fat diet—HFD) comprise dysbiosis, gut barrier dysfunction, increased intestinal permeability, and leakage of toxic bacterial metabolites into the circulation. These can strongly contribute to the development of low-grade systemic inflammation. This narrative review highlights the most important recent advances linking HFD-driven dysbiosis and HFD-related inflammation, presents the pathomechanisms for these phenomena, and examines the possible causative relationship between pro-inflammatory status and gut microbiota changes.
Collapse
Affiliation(s)
- Ida Judyta Malesza
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 61-701 Poznań, Poland; (I.J.M.); (J.W.)
| | - Michał Malesza
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznań, Poland; (M.M.); (J.B.-W.)
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 61-701 Poznań, Poland; (I.J.M.); (J.W.)
| | - Nadiar Mussin
- Department of General Surgery, West Kazakhstan Marat Ospanov Medical University, Aktobe 030012, Kazakhstan;
| | - Dariusz Walkowiak
- Department of Organization and Management in Health Care, Poznan University of Medical Sciences, 61-701 Poznań, Poland;
| | - Raisa Aringazina
- Department of Internal Diseases No. 1, West Kazakhstan Marat Ospanov Medical University, Aktobe 030012, Kazakhstan;
| | | | - Edyta Mądry
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznań, Poland; (M.M.); (J.B.-W.)
- Correspondence:
| |
Collapse
|
39
|
Omega-3 Supplementation Prevents Short-Term High-Fat Diet Effects on the α7 Nicotinic Cholinergic Receptor Expression and Inflammatory Response. Mediators Inflamm 2021; 2021:5526940. [PMID: 34421366 PMCID: PMC8371655 DOI: 10.1155/2021/5526940] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/16/2021] [Accepted: 07/13/2021] [Indexed: 12/29/2022] Open
Abstract
The study is aimed at investigating if PUFA supplementation could prevent the effects of a short-term HFD on α7nAChR expression and on the severity of sepsis. Swiss mice were used for the in vivo experiments. For the in vitro experiments, we used a microglia cell line (BV-2) and a hepatoma cell line (Hepa-1c1c7) derived from mice. The animals were either fed standard chow, fed a short-term HFD (60%), or given supplementation with omega-3 fatty acid (2 g/kg or 4 g/kg bw) for 17 days, followed by a short-term HFD. Endotoxemia was induced with an intraperitoneal (i.p.) lipopolysaccharide injection (LPS, 5 or 12 mg/kg), and sepsis was induced by subjecting the animals to cecal ligation and puncture (CLP). BV-2 and Hepa-1c1c7 cells were treated with LPS (100 and 500 ng/mL, respectively) for 3 hours. RT-PCR or Western blotting was used to evaluate α7nAChR expression, inflammatory markers, DNMT1, and overall ubiquitination. LPS and HFD reduced the expression of α7nAChR and increased the expression of inflammatory markers. Omega-3 partially prevented the damage caused by the HFD to the expression of α7nAChR in the bone marrow and hypothalamus, decreased the inflammatory markers, and reduced susceptibility to sepsis-induced death. Exposing the BV-2 cells to LPS increased the protein content of DNMT1 and the overall ubiquitination and reduced the expression of α7nAChR. The inflammation induced by LPS in the BV-2 cell decreased α7nAChR expression and concomitantly increased DNMT1 expression and the ubiquitinated protein levels, indicating the participation of pre- and posttranscriptional mechanisms.
Collapse
|
40
|
Hirschberger S, Strauß G, Effinger D, Marstaller X, Ferstl A, Müller MB, Wu T, Hübner M, Rahmel T, Mascolo H, Exner N, Heß J, Kreth FW, Unger K, Kreth S. Very-low-carbohydrate diet enhances human T-cell immunity through immunometabolic reprogramming. EMBO Mol Med 2021; 13:e14323. [PMID: 34151532 PMCID: PMC8350890 DOI: 10.15252/emmm.202114323] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/16/2022] Open
Abstract
Very-low-carbohydrate diet triggers the endogenous production of ketone bodies as alternative energy substrates. There are as yet unproven assumptions that ketone bodies positively affect human immunity. We have investigated this topic in an in vitro model using primary human T cells and in an immuno-nutritional intervention study enrolling healthy volunteers. We show that ketone bodies profoundly impact human T-cell responses. CD4+ , CD8+ , and regulatory T-cell capacity were markedly enhanced, and T memory cell formation was augmented. RNAseq and functional metabolic analyses revealed a fundamental immunometabolic reprogramming in response to ketones favoring mitochondrial oxidative metabolism. This confers superior respiratory reserve, cellular energy supply, and reactive oxygen species signaling. Our data suggest a very-low-carbohydrate diet as a clinical tool to improve human T-cell immunity. Rethinking the value of nutrition and dietary interventions in modern medicine is required.
Collapse
Affiliation(s)
- Simon Hirschberger
- Walter Brendel Center of Experimental MedicineLudwig‐Maximilian‐University München (LMU)MunichGermany
- Department of Anaesthesiology and Intensive Care MedicineResearch Unit Molecular MedicineLMU University HospitalLudwig‐Maximilian‐University München (LMU)MunichGermany
| | - Gabriele Strauß
- Walter Brendel Center of Experimental MedicineLudwig‐Maximilian‐University München (LMU)MunichGermany
- Department of Anaesthesiology and Intensive Care MedicineResearch Unit Molecular MedicineLMU University HospitalLudwig‐Maximilian‐University München (LMU)MunichGermany
| | - David Effinger
- Walter Brendel Center of Experimental MedicineLudwig‐Maximilian‐University München (LMU)MunichGermany
- Department of Anaesthesiology and Intensive Care MedicineResearch Unit Molecular MedicineLMU University HospitalLudwig‐Maximilian‐University München (LMU)MunichGermany
| | - Xaver Marstaller
- Walter Brendel Center of Experimental MedicineLudwig‐Maximilian‐University München (LMU)MunichGermany
| | - Alicia Ferstl
- Walter Brendel Center of Experimental MedicineLudwig‐Maximilian‐University München (LMU)MunichGermany
| | - Martin B Müller
- Walter Brendel Center of Experimental MedicineLudwig‐Maximilian‐University München (LMU)MunichGermany
- Department of Anaesthesiology and Intensive Care MedicineResearch Unit Molecular MedicineLMU University HospitalLudwig‐Maximilian‐University München (LMU)MunichGermany
| | - Tingting Wu
- Walter Brendel Center of Experimental MedicineLudwig‐Maximilian‐University München (LMU)MunichGermany
| | - Max Hübner
- Walter Brendel Center of Experimental MedicineLudwig‐Maximilian‐University München (LMU)MunichGermany
- Department of Anaesthesiology and Intensive Care MedicineResearch Unit Molecular MedicineLMU University HospitalLudwig‐Maximilian‐University München (LMU)MunichGermany
| | - Tim Rahmel
- Department of AnesthesiaIntensive Care Medicine and Pain TherapyUniversity Hospital Knappschaftskrankenhaus BochumBochumGermany
| | - Hannah Mascolo
- Walter Brendel Center of Experimental MedicineLudwig‐Maximilian‐University München (LMU)MunichGermany
| | - Nicole Exner
- Metabolic BiochemistryBiomedical Center (BMC)Faculty of MedicineLudwig‐Maximilian‐University München (LMU)MunichGermany
| | - Julia Heß
- Helmholtz Center MunichResearch Unit Radiation CytogeneticsNeuherbergGermany
- Department of Radiation OncologyLMU University HospitalLudwig‐Maximilian‐University München (LMU)MunichGermany
| | - Friedrich W Kreth
- Walter Brendel Center of Experimental MedicineLudwig‐Maximilian‐University München (LMU)MunichGermany
| | - Kristian Unger
- Helmholtz Center MunichResearch Unit Radiation CytogeneticsNeuherbergGermany
- Department of Radiation OncologyLMU University HospitalLudwig‐Maximilian‐University München (LMU)MunichGermany
| | - Simone Kreth
- Walter Brendel Center of Experimental MedicineLudwig‐Maximilian‐University München (LMU)MunichGermany
- Department of Anaesthesiology and Intensive Care MedicineResearch Unit Molecular MedicineLMU University HospitalLudwig‐Maximilian‐University München (LMU)MunichGermany
| |
Collapse
|
41
|
Yu C, Zhu X, Zheng C, Luo Y, Wang F, Gao Y, Wu H, Sun X, Kong X. Methyl Diet Enhanced Sepsis-Induced Mortality Through Altering Gut Microbiota. J Inflamm Res 2021; 14:3107-3121. [PMID: 34276224 PMCID: PMC8277458 DOI: 10.2147/jir.s305202] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022] Open
Abstract
Introduction Mortality of sepsis is caused by an inappropriately amplified systemic inflammatory response and bacteremia. Methyl diet has been shown to associate with greater inflammation response in different diseases. This study aimed to determine whether dietary supplementation with methyl donors affects the inflammation response and mortality in sepsis and to investigate the underlying mechanisms. Methods Four-week-old male C57BL/6 mice were fed with a high-methyl diet (HMD) or a regulator diet (RD) till the experiment time. Mice septic model was induced by Cecal ligation and puncture (CLP), lipopolysaccharide (LPS), or E.coli. Inflammatory cytokine was analyzed by ELISA and qRT-PCR. Immune cell infiltration was evaluated by H&E and IHC. The composition of gut microbiota was determined by 16S rRNA sequencing. The effect of gut microbiota on sepsis was further verified by fecal microbiome transplantation. Results Our results showed that the diet riches in methyl donors exacerbated mortality, organ injury, and circulating levels of inflammatory mediators in CLP-induced septic mice model, compared to the control diet group. However, no significant differences have been observed in the inflammatory responses in the LPS-induced septic model and macrophages activation between the two groups of mice. There was a higher bacterial burden in CLP-induced HMD mice suggested that methyl diet might modulate gut microbiota. Bacterial 16S rRNA sequencing results showed that the composition of gut microbiota was altered. The high methyl donor diet reduced the abundance of Akkermansia and Lachnospiraceae, which were associated with protective effects in sepsis, in the gut. Moreover, fecal microbiome transplantation experiment showed that the transfer of feces, which obtained from high methyl diet mice, aggravated the mortality and inflammation responses in recipient mice. Discussion Methyl diet enhanced CLP-induced septic mortality and inflammatory responses through altering the composition of gut microbiota. This result indicated that diet-based gut microbiota may be a new therapeutic strategy for sepsis patients.
Collapse
Affiliation(s)
- Chang Yu
- Central Laboratory, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, People's Republic of China
| | - Xiaojun Zhu
- Central Laboratory, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, People's Republic of China
| | - Chao Zheng
- Central Laboratory, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, People's Republic of China
| | - Yichun Luo
- Central Laboratory, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, People's Republic of China
| | - Fang Wang
- Central Laboratory, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, People's Republic of China
| | - Yueqiu Gao
- Central Laboratory, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, People's Republic of China
| | - Hailong Wu
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, People's Republic of China
| | - Xuehua Sun
- Central Laboratory, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, People's Republic of China
| | - Xiaoni Kong
- Central Laboratory, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, People's Republic of China
| |
Collapse
|
42
|
Pérez-Hernández EG, Delgado-Coello B, Luna-Reyes I, Mas-Oliva J. New insights into lipopolysaccharide inactivation mechanisms in sepsis. Biomed Pharmacother 2021; 141:111890. [PMID: 34229252 DOI: 10.1016/j.biopha.2021.111890] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/03/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
The complex pathophysiology of sepsis makes it a syndrome with limited therapeutic options and a high mortality rate. Gram-negative bacteria containing lipopolysaccharides (LPS) in their outer membrane correspond to the most common cause of sepsis. Since the gut is considered an important source of LPS, intestinal damage has been considered a cause and a consequence of sepsis. Although important in the maintenance of the intestinal epithelial cell homeostasis, the microbiota has been considered a source of LPS. Recent studies have started to shed light on how sepsis is triggered by dysbiosis, and an increased inflammatory state of the intestinal epithelial cells, expanding the understanding of the gut-liver axis in sepsis. Here, we review the gut-liver interaction in Gram-negative sepsis, exploring the mechanisms of LPS inactivation, including the recently described contribution of an isoform of the cholesteryl-ester transfer protein (CETPI). Although several key questions remain to be answered when the pathophysiology of sepsis is reviewed, new contributions coming to light exploring the way LPS might be inactivated in vivo, suggest that new applications might soon reach the clinical setting.
Collapse
Affiliation(s)
| | - Blanca Delgado-Coello
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico
| | - Ismael Luna-Reyes
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico
| | - Jaime Mas-Oliva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico.
| |
Collapse
|
43
|
Candelli M, Franza L, Pignataro G, Ojetti V, Covino M, Piccioni A, Gasbarrini A, Franceschi F. Interaction between Lipopolysaccharide and Gut Microbiota in Inflammatory Bowel Diseases. Int J Mol Sci 2021; 22:6242. [PMID: 34200555 PMCID: PMC8226948 DOI: 10.3390/ijms22126242] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/22/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023] Open
Abstract
Lipopolysaccharides (LPSs) are bacterial surface glycolipids, produced by Gram-negative bacteria. LPS is known to determine acute inflammatory reactions, particularly in the context of sepsis. However, LPS can also trigger chronic inflammation. In this case, the source of LPS is not an external infection, but rather an increase in endogenous production, which is usually sustained by gut microbiota (GM), and LPS contained in food. The first site in which LPS can exert its inflammatory action is the gut: both GM and gut-associated lymphoid tissue (GALT) are influenced by LPS and shift towards an inflammatory pattern. The changes in GM and GALT induced by LPS are quite similar to the ones seen in IBD: GM loses diversity, while GALT T regulatory (Tregs) lymphocytes are reduced in number, with an increase in Th17 and Th1 lymphocytes. Additionally, the innate immune system is triggered, through the activation of toll-like receptor (TLR)-4, while the epithelium is directly damaged, further triggering inflammation. In this review, we will discuss the importance of the crosstalk between LPS, GM, and GALT, and discuss the possible implications.
Collapse
Affiliation(s)
- Marcello Candelli
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (L.F.); (G.P.); (V.O.); (M.C.); (A.P.); (F.F.)
| | - Laura Franza
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (L.F.); (G.P.); (V.O.); (M.C.); (A.P.); (F.F.)
| | - Giulia Pignataro
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (L.F.); (G.P.); (V.O.); (M.C.); (A.P.); (F.F.)
| | - Veronica Ojetti
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (L.F.); (G.P.); (V.O.); (M.C.); (A.P.); (F.F.)
| | - Marcello Covino
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (L.F.); (G.P.); (V.O.); (M.C.); (A.P.); (F.F.)
| | - Andrea Piccioni
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (L.F.); (G.P.); (V.O.); (M.C.); (A.P.); (F.F.)
| | - Antonio Gasbarrini
- Medical and Surgical Science Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, Largo A. Gemelli 8, 00168 Rome, Italy;
| | - Francesco Franceschi
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore di Roma, Largo A. Gemelli 8, 00168 Rome, Italy; (L.F.); (G.P.); (V.O.); (M.C.); (A.P.); (F.F.)
| |
Collapse
|
44
|
|
45
|
Yang L, Li Y, Wang S, Bian X, Jiang X, Wu J, Wang K, Wang Q, Xia J, Jiang S, Zhuge A, Yuan Y, Li S, Li L. Western Diet Aggravated Carbon Tetrachloride-Induced Chronic Liver Injury by Disturbing Gut Microbiota and Bile Acid Metabolism. Mol Nutr Food Res 2021; 65:e2000811. [PMID: 33458949 DOI: 10.1002/mnfr.202000811] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 11/17/2020] [Indexed: 12/13/2022]
Abstract
SCOPE The high-fat, high-sucrose, and low-fiber Western diet (WD) is popular in many countries and affects the onset and progression of many diseases. This study is aimed to explore the influence of the WD on chronic liver disease (CLD) and its possible mechanism. METHODS AND RESULTS C57BL/6 mice are given a control diet (CD) or WD and CLD is induced by intraperitoneally injecting carbon tetrachloride (CCL4 ) twice a week for 8 weeks. The WD aggravated CCL4 -induced chronic liver injury, as evidenced by increased serum transaminase levels, worsened hepatic inflammatory response, and fibrosis. Gut microbiota is disturbed in mice treated with CCL4 +WD (WC group), manifested as the accumulation of Fusobacteria, Streptococcaceae, Streptococcus, Fusobacterium, and Prevotella and the depletion of Firmicutes, Lachnospiraceae, and Roseburia. Additionally, increased hepatic taurocholic acid in the WC group activated sphingosine-1-phosphate receptor 2, which is positively correlated with hepatic fibrosis and inflammation parameters. Mice in the WC group have higher fecal primary bile acid (BA) levels and lower fecal secondary/primary BA ratios. Serum FGF15 levels are also elevated in the WC group, which is positively correlated with hepatic inflammation. CONCLUSION WD accelerates the progression of CLD which is associated with changes in the gut microbiota and BA metabolism.
Collapse
Affiliation(s)
- Liya Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China
| | - Yating Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China
| | - Shuting Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China
| | - Xiaoyuan Bian
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China
| | - Xianwan Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China
| | - Jingjing Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China
| | - Kaicen Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China
| | - Qiangqiang Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China
| | - Jiafeng Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China
| | - Shiman Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China
| | - Aoxiang Zhuge
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China
| | - Yin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China
| | - Shenjie Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China
| |
Collapse
|
46
|
Wiȩckowska-Gacek A, Mietelska-Porowska A, Chutorański D, Wydrych M, Długosz J, Wojda U. Western Diet Induces Impairment of Liver-Brain Axis Accelerating Neuroinflammation and Amyloid Pathology in Alzheimer's Disease. Front Aging Neurosci 2021; 13:654509. [PMID: 33867971 PMCID: PMC8046915 DOI: 10.3389/fnagi.2021.654509] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/05/2021] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) is an aging-dependent, irreversible neurodegenerative disorder and the most common cause of dementia. The prevailing AD hypothesis points to the central role of altered cleavage of amyloid precursor protein (APP) and formation of toxic amyloid-β (Aβ) deposits in the brain. The lack of efficient AD treatments stems from incomplete knowledge on AD causes and environmental risk factors. The role of lifestyle factors, including diet, in neurological diseases is now beginning to attract considerable attention. One of them is western diet (WD), which can lead to many serious diseases that develop with age. The aim of the study was to investigate whether WD-derived systemic disturbances may accelerate the brain neuroinflammation and amyloidogenesis at the early stages of AD development. To verify this hypothesis, transgenic mice expressing human APP with AD-causing mutations (APPswe) were fed with WD from the 3rd month of age. These mice were compared to APPswe mice, in which short-term high-grade inflammation was induced by injection of lipopolysaccharide (LPS) and to untreated APPswe mice. All experimental subgroups of animals were subsequently analyzed at 4-, 8-, and 12-months of age. APPswe mice at 4- and 8-months-old represent earlier pre-plaque stages of AD, while 12-month-old animals represent later stages of AD, with visible amyloid pathology. Already short time of WD feeding induced in 4-month-old animals such brain neuroinflammation events as enhanced astrogliosis, to a level comparable to that induced by the administration of pro-inflammatory LPS, and microglia activation in 8-month-old mice. Also, WD feeding accelerated increased Aβ production, observed already in 8-month-old animals. These brain changes corresponded to diet-induced metabolic disorders, including increased cholesterol level in 4-months of age, and advanced hypercholesterolemia and fatty liver disease in 8-month-old mice. These results indicate that the westernized pattern of nourishment is an important modifiable risk factor of AD development, and that a healthy, balanced, diet may be one of the most efficient AD prevention methods.
Collapse
Affiliation(s)
| | | | | | | | | | - Urszula Wojda
- Laboratory of Preclinical Testing of Higher Standard, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
47
|
Rebai R, Jasmin L, Boudah A. Agomelatine effects on fat-enriched diet induced neuroinflammation and depression-like behavior in rats. Biomed Pharmacother 2021; 135:111246. [PMID: 33453676 DOI: 10.1016/j.biopha.2021.111246] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 11/24/2020] [Accepted: 01/03/2021] [Indexed: 12/12/2022] Open
Abstract
Growing evidence suggests that a high fat diet (HFD) induces oxidative stress on the central nervous system (CNS), which predisposes to mood disorders and neuroinflammation. In this study we postulated that in addition to improving mood, antidepressant therapy would reverse inflammatory changes in the brain of rats exposed to a HFD. To test our hypothesis, we measured the effect of the antidepressant agomelatine (AGO) on anxiety- and depressive-like behaviors, as well as on CNS markers of inflammation in rats rendered obese. Agomelatine is an agonist of the melatonin receptors MT1 and MT2 and an antagonist of the serotonin receptors 5HT2B and 5HT2C. A subset of rats was also treated with lipopolysaccharides (LPS) to determine how additional neuroinflammation alters behavior and affects the response to the antidepressant. Specifically, rats were subjected to a 14-week HFD, during which time behavior was evaluated twice, first at the 10th week prior to LPS and/or agomelatine, and then at the 14th week after a bi-weekly exposure to LPS (250 μg/kg) and daily treatment with agomelatine (40 mg/kg). Immediately after the second behavioral testing we measured the proinflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6) and interleukin 1 beta (IL-1β), markers of oxidative stress thiobarbituric acid reactive substances (TABRS), catalase (CAT) and glutathione peroxidase (GPx), the growth factor BDNF, as well as the apoptosis marker caspase-3. Our results show that a HFD induced an anxiety-like behavior in the open field test (OFT) at the 10th week, followed by a depressive-like behavior in the forced swim test (FST) at the 14th week. In the prefrontal and hippocampal cortices of rats exposed to a HFD we noted an overproduction of TNF-α, IL-6, IL-1β, and TABRS, together with an increase in caspase-3 activity. We also observed a decrease in BDNF, as well as reduced CAT and GPx activity in the same brain areas. Treatment with agomelatine reversed the signs of anxiety and depression, and decreased the cytokines (TNF-α, IL-6 and IL-1β), TABRS, as well as caspase-3 activity. Agomelatine also restored BDNF levels and the activity of antioxidant enzymes CAT and GPx. Our findings suggest that the anxiolytic/antidepressant effect of agomelatine in obese rats could result from a reversal of the inflammatory and oxidative stress brought about by their diet.
Collapse
Affiliation(s)
- Redouane Rebai
- Department of Natural and Life Sciences, Faculty of Exact Sciences and Natural and Life Sciences, University Mohamed Khider of Biskra, BP 145 RP, 07000, Biskra, Algeria; Laboratory of Biotechnology, National Higher School of Biotechnology, Ville universitaire Ali Mendjeli, BP E66 25100, Constantine, Algeria.
| | - Luc Jasmin
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco, 707 Parnassus Ave Suite D-1201, San Francisco, CA, 94143, USA.
| | - Abdennacer Boudah
- Laboratory of Biotechnology, National Higher School of Biotechnology, Ville universitaire Ali Mendjeli, BP E66 25100, Constantine, Algeria.
| |
Collapse
|
48
|
Urban living in healthy Tanzanians is associated with an inflammatory status driven by dietary and metabolic changes. Nat Immunol 2021; 22:287-300. [PMID: 33574617 DOI: 10.1038/s41590-021-00867-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 12/29/2020] [Indexed: 01/28/2023]
Abstract
Sub-Saharan Africa currently experiences an unprecedented wave of urbanization, which has important consequences for health and disease patterns. This study aimed to investigate and integrate the immune and metabolic consequences of rural or urban lifestyles and the role of nutritional changes associated with urban living. In a cohort of 323 healthy Tanzanians, urban as compared to rural living was associated with a pro-inflammatory immune phenotype, both at the transcript and protein levels. We identified different food-derived and endogenous circulating metabolites accounting for these differences. Serum from urban dwellers induced reprogramming of innate immune cells with higher tumor necrosis factor production upon microbial re-stimulation in an in vitro model of trained immunity. These data demonstrate important shifts toward an inflammatory phenotype associated with an urban lifestyle and provide new insights into the underlying dietary and metabolic factors, which may affect disease epidemiology in sub-Sahara African countries.
Collapse
|
49
|
Marki A, Buscher K, Lorenzini C, Meyer M, Saigusa R, Fan Z, Yeh YT, Hartmann N, Dan JM, Kiosses WB, Golden GJ, Ganesan R, Winkels H, Orecchioni M, McArdle S, Mikulski Z, Altman Y, Bui J, Kronenberg M, Chien S, Esko JD, Nizet V, Smalley D, Roth J, Ley K. Elongated neutrophil-derived structures are blood-borne microparticles formed by rolling neutrophils during sepsis. J Exp Med 2021; 218:e20200551. [PMID: 33275138 PMCID: PMC7721910 DOI: 10.1084/jem.20200551] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 09/28/2020] [Accepted: 11/06/2020] [Indexed: 12/30/2022] Open
Abstract
Rolling neutrophils form tethers with submicron diameters. Here, we report that these tethers detach, forming elongated neutrophil-derived structures (ENDS) in the vessel lumen. We studied ENDS formation in mice and humans in vitro and in vivo. ENDS do not contain mitochondria, endoplasmic reticulum, or DNA, but are enriched for S100A8, S100A9, and 57 other proteins. Within hours of formation, ENDS round up, and some of them begin to present phosphatidylserine on their surface (detected by annexin-5 binding) and release S100A8-S100A9 complex, a damage-associated molecular pattern protein that is a known biomarker of neutrophilic inflammation. ENDS appear in blood plasma of mice upon induction of septic shock. Compared with healthy donors, ENDS are 10-100-fold elevated in blood plasma of septic patients. Unlike neutrophil-derived extracellular vesicles, most ENDS are negative for the tetraspanins CD9, CD63, and CD81. We conclude that ENDS are a new class of bloodborne submicron particles with a formation mechanism linked to neutrophil rolling on the vessel wall.
Collapse
Affiliation(s)
- Alex Marki
- La Jolla Institute for Immunology, La Jolla, CA
| | - Konrad Buscher
- La Jolla Institute for Immunology, La Jolla, CA
- Division of General Internal Medicine, Nephrology, and Rheumatology, Department of Medicine D, University Hospital Muenster, Muenster, Germany
| | - Cristina Lorenzini
- La Jolla Institute for Immunology, La Jolla, CA
- Laboratory of Immunobiology, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | | | | | - Zhichao Fan
- La Jolla Institute for Immunology, La Jolla, CA
- Department of Immunology, University of Connecticut Health Center, Farmington, CT
| | - Yi-Ting Yeh
- Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA
| | | | - Jennifer M. Dan
- La Jolla Institute for Immunology, La Jolla, CA
- Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA
| | | | - Gregory J. Golden
- Department of Cellular and Molecular Medicine and Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA
| | - Rajee Ganesan
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | | | | | | | - Yoav Altman
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Jack Bui
- Department of Pathology, University of California, San Diego, La Jolla, CA
| | | | - Shu Chien
- Institute for Immunology, University of Muenster, Muenster, Germany
| | - Jeffrey D. Esko
- Department of Cellular and Molecular Medicine and Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA
| | - Victor Nizet
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA
| | - David Smalley
- Systems Mass Spectrometry Core, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
| | - Johannes Roth
- Institute for Immunology, University of Muenster, Muenster, Germany
| | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA
- Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA
| |
Collapse
|
50
|
He W, Xiao K, Xu J, Guan W, Xie S, Wang K, Yan P, Fang M, Xie L. Recurrent Sepsis Exacerbates CD4 + T Cell Exhaustion and Decreases Antiviral Immune Responses. Front Immunol 2021; 12:627435. [PMID: 33717146 PMCID: PMC7946831 DOI: 10.3389/fimmu.2021.627435] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/18/2021] [Indexed: 12/29/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to an infection. It is a disease with a high incidence, mortality, and recurrence rate and frequently results in its survivors requiring readmission into hospitals. The readmission is mainly due to recurrent sepsis. Patients with recurrent sepsis are more susceptible to secondary infections partly due to immune dysfunction, leading to a higher mortality in the long term. However, there remains a gap in the understanding of immunological characteristics and underlying mechanisms of recurrent sepsis. In this study, we used mouse models of acute and recurrent sepsis to investigate their different immunological characteristics. And then we subjected the two mouse models to a secondary influenza A virus (H1N1) infection and characterized the different immune responses. Here, we demonstrated that CD4+ T cells present an exacerbated exhaustion phenotype in response to recurrent sepsis as illustrated by the decreased frequency of CD4+ T cells, reduced co-stimulatory CD28 and increased inhibitory PD-1 and Tim-3 expression on CD4+ T cells, increased frequency of regulatory T cells, and reduced MHC-II expression on antigen-presenting cells. Moreover, we showed that antiviral immune responses decrease in the recurrent sepsis mouse model subjected to a secondary infection as illustrated by the reduced pathogen clearance and inflammatory response. This may be a consequence of the exacerbated CD4+ T cell exhaustion. In summary, recurrent sepsis exacerbates CD4+ T cell exhaustion and decreases antiviral immune responses, contributing to significant morbidity, increased late mortality, and increased health care burden in recurrent sepsis patients.
Collapse
Affiliation(s)
- Wanxue He
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Kun Xiao
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jiaruo Xu
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Wei Guan
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Sheling Xie
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Kaifei Wang
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Peng Yan
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Min Fang
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Lixin Xie
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|