1
|
Lee CC, Fan H, Tsopmo A, Regenstein JM, Ashaolu TJ. Plant-based antioxidant peptides: impact on oxidative stress and gut microbiota. Crit Rev Food Sci Nutr 2025:1-24. [PMID: 40219794 DOI: 10.1080/10408398.2025.2490270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
Plant-based peptides can be obtained from natural and climate-friendly sources. These peptides show various bioactivities including antioxidant activity. Oxidative stress has an impact on the gut microbiota causing inflammation, insulin resistance, osteoporosis, cancer, and several chronic diseases like type 2 diabetes, arthritis, hypertension, and atherosclerosis. Therefore, antioxidant peptides may significantly affect oxidative stress as a potential alternative to conventional medication. The production of antioxidant peptides from plant-based protein sources through conventional and innovative approaches may provide promising strategies to improve gut microbiota. Recent studies in plant-based antioxidant peptides (PBAP) focus on their advanced identification and characterization techniques, structure-activity relationship, improvement of extraction and purification, cellular and molecular mechanisms, specific health applications in preventing and managing conditions with gut microbiota, and commercial applications in nutraceuticals. Short-chain fatty acids and reactive sulfur species are specific gut-derived metabolites that can improve metabolic function by modulating oxidative stress and the immune system. This review highlights the influence of food oxidants on the gut microbiota and PBAP-induced modulation of gut microbiota. Moreover, the production of PBAP and the challenges in their application will be discussed.
Collapse
Affiliation(s)
- Chi Ching Lee
- Department of Food Engineering, Faculty of Engineering and Natural Sciences, Istanbul Sabahattin Zaim University, Istanbul, Turkey
- Department of Food Technology and Nutrition, Faculty of Technologies, Klaipeda State University of Applied Sciences, Klaipeda, Lithuania
| | - Hongbing Fan
- Department of Animal and Food Sciences, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, USA
| | - Apollinaire Tsopmo
- Food Science and Nutrition Program, Department of Chemistry, Carleton University, Ottawa, Canada
- Institute of Biochemistry, Carleton University, Ottawa, Canada
| | - Joe M Regenstein
- Department of Food Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| | - Tolulope Joshua Ashaolu
- Institute for Global Health Innovations, Duy Tan University, Da Nang, Vietnam
- Faculty of Medicine, Duy Tan University, Da Nang, Vietnam
| |
Collapse
|
2
|
Ogawa S, Hori H, Niwa M, Itoh M, Lin M, Yoshida F, Ino K, Kawanishi H, Narita M, Nakano W, Imai R, Matsui M, Kamo T, Kunugi H, Hattori K, Kim Y. Serum lipid and plasma fatty acid profiles in PTSD patients and healthy individuals: Associations with symptoms, cognitive function, and inflammatory markers. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111298. [PMID: 39988258 DOI: 10.1016/j.pnpbp.2025.111298] [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: 10/19/2024] [Revised: 01/22/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Increasing evidence suggests that posttraumatic stress disorder (PTSD), a serious mental health condition, is associated with physical health problems. Lipid-related molecules are crucial for central nervous system functions associated with PTSD symptoms; however, case-control studies exploring the relationship between PTSD and lipid-related molecules are scarce. We examined 68 civilian PTSD patients and 97 healthy controls, evaluating PTSD symptoms, childhood maltreatment history, suicidality, and cognitive functions. Cholesterol, triglycerides, and inflammation-related marker levels were analyzed in serum, while fatty acid levels were measured in plasma. Compared to controls, patients exhibited significantly lower high-density lipoprotein cholesterol and n-6 linoleic acid levels, alongside higher saturated palmitic acid levels and the triene-to-tetraene (T/T) ratio. PTSD symptoms, particularly hyperarousal, were significantly positively correlated with n-6 γ-linolenic, n-6 dihomo-γ-linolenic, and n-9 mead acid levels, and the T/T ratio. Cognitive functions were significantly positively correlated with n-3 docosahexaenoic acid and total n-3 fatty acid levels, and negatively correlated with saturated lauric, palmitic, and total saturated fatty acid levels. Suicidality was significantly positively correlated with dihomo-γ-linolenic acid, mead acid levels, and the T/T ratio, and negatively correlated with polyunsaturated fatty acid (PUFA) levels. Inflammation-related marker levels were significantly correlated with higher palmitic, n-9 oleic, and total n-9 fatty acid levels, and lower linoleic acid and PUFA levels. Latent profile analysis (LPA) revealed distinct subgroups associated with unique fatty acid profiles. These lipid-related alterations may improve the understanding of PTSD pathophysiology. Distinct fatty acid profiles identified by LPA may help subtype PTSD patients and guide nutrition-based personalized treatment strategies.
Collapse
Affiliation(s)
- Shintaro Ogawa
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.
| | - Hiroaki Hori
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.
| | - Madoka Niwa
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Mariko Itoh
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Mingming Lin
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Laboratory for Imagination and Executive Functions, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Fuyuko Yoshida
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Keiko Ino
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Psychiatry and Cognitive-Behavioral Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Hitomi Kawanishi
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Advanced Neuroimaging, Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Megumi Narita
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Wakako Nakano
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Risa Imai
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Psychiatry and Cognitive-Behavioral Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan; Risa Irinaka Mental Clinic, Nagoya, Aichi, Japan
| | - Mie Matsui
- Department of Clinical Cognitive Neuroscience, Institute of Liberal Arts and Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Toshiko Kamo
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Wakamatsu-cho Mental and Skin Clinic, Shinjuku, Tokyo, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Psychiatry, Teikyo University School of Medicine, Itabashi, Tokyo, Japan
| | - Kotaro Hattori
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan; Department of Bioresources, Medical Genome Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Yoshiharu Kim
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| |
Collapse
|
3
|
Bitterlich LM, Tunstead C, Hogan AE, Ankrum JA, English K. Mesenchymal stromal cells can block palmitate training of macrophages via cyclooxygenase-2 and interleukin-1 receptor antagonist. Cytotherapy 2025; 27:169-180. [PMID: 39580716 DOI: 10.1016/j.jcyt.2024.10.011] [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: 06/26/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024]
Abstract
Innate training of macrophages can be beneficial for the clearance of pathogens. However, for certain chronic conditions, innate training can have detrimental effects due to an excessive production of pro-inflammatory cytokines. Obesity is a condition that is associated with a range of increased pro-inflammatory training stimuli including the free fatty acid palmitate. Mesenchymal stromal cells (MSCs) are powerful immunomodulators and known to suppress inflammatory macrophages via a range of soluble factors. We show that palmitate training of murine bone-marrow-derived macrophages and human monocyte-derived macrophages (MDMs) results in an increased production of TNFα and IL-6 upon stimulation with lipopolysaccharide and is associated with epigenetic remodeling. Palmitate training led to metabolic changes, however, MSCs did not alter the metabolic profile of human MDMs. Using a transwell system, we demonstrated that human bone marrow MSCs block palmitate training in both murine and human macrophages suggesting the involvement of secreted factors. MSC disruption of the training process occurs through more than one pathway. Suppression of palmitate-enhanced TNFα production is associated with cyclooxygenase-2 activity in MSCs, while secretion of interleukin-1 receptor antagonist by MSCs is required to suppress palmitate-enhanced IL-6 production in MDMs.
Collapse
Affiliation(s)
- Laura M Bitterlich
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland; Department of Biology, Maynooth University, Maynooth, Ireland
| | - Courteney Tunstead
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland; Department of Biology, Maynooth University, Maynooth, Ireland
| | - Andrew E Hogan
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland; Department of Biology, Maynooth University, Maynooth, Ireland
| | - James A Ankrum
- University of Iowa Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, USA; Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, USA
| | - Karen English
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland; Department of Biology, Maynooth University, Maynooth, Ireland.
| |
Collapse
|
4
|
Lin J, Sun W, Peng S, Hu Y, Zhang G, Song W, Jiang B, Liao Y, Pei C, Zhang J, Dai J, Wang X, Peng P, Bi X. Molecular characteristics of organic matters in PM 2.5 associated with upregulation of respiratory virus infection in vitro. JOURNAL OF HAZARDOUS MATERIALS 2025; 482:136583. [PMID: 39577291 DOI: 10.1016/j.jhazmat.2024.136583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024]
Abstract
The extent to which organic matters (OM) in PM2.5 affect virus infections and the key organic molecules involved in this process remain unclear. Herein, this study utilized ultra-high resolution mass spectrometry coupled with in vitro experiments to identify the organic molecules associated with respiratory virus infection for the first time. Water-soluble organic matters (WSOM) and water-insoluble organic matters (WIOM) were separated from PM2.5 samples collected at the urban area of Guangzhou, China. Their molecular compositions were analyzed using Fourier transform ion cyclotron resonance mass spectrometry. Subsequently, in vitro experiments were conducted to explore the impact of WSOM and WIOM exposure on the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pseudo-virus infection in A549 cells. Results revealed that WSOM and WIOM respectively promoted 1.7 to 2.1-fold and 1.9 to 3.5-fold upregulation of SARS-CoV-2 pseudo-virus infection in a concentration-dependent manner (at 25 to 100 μg mL-1) compared to the virus-only control group. Partial least squares model analysis indicated that the increased virus infection was likely related to phthalate ester and nitro-aromatic molecules in WSOM, as well as LipidC molecules with aliphatic and olefinic structures in WIOM. Interestingly, the molecules responsible for upregulating SARS-CoV-2 receptor angiotensin-converting enzyme 2 (ACE2) expression and virus infection differed. Thus, it was concluded that ACE2 upregulation alone may not fully elucidate the mechanisms underlying increased susceptibility to virus infection. The findings highlight the critical importance of aromatic and lipid molecules found in OM in relation to respiratory virus infection.
Collapse
Affiliation(s)
- Juying Lin
- State Key Laboratory of Organic Geochemistry and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Wei Sun
- State Key Laboratory of Organic Geochemistry and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, PR China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control, Guangzhou 510640, PR China
| | - Shuyi Peng
- State Key Laboratory of Organic Geochemistry and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yaohao Hu
- State Key Laboratory of Organic Geochemistry and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Guohua Zhang
- State Key Laboratory of Organic Geochemistry and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, PR China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control, Guangzhou 510640, PR China
| | - Wei Song
- State Key Laboratory of Organic Geochemistry and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, PR China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control, Guangzhou 510640, PR China
| | - Bin Jiang
- State Key Laboratory of Organic Geochemistry and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, PR China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control, Guangzhou 510640, PR China
| | - Yuhong Liao
- State Key Laboratory of Organic Geochemistry and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, PR China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control, Guangzhou 510640, PR China
| | - Chenglei Pei
- Guangzhou Sub-branch of Guangdong Ecological and Environmental Monitoring Center, Guangzhou 510006, China
| | - Jinpu Zhang
- Guangzhou Sub-branch of Guangdong Ecological and Environmental Monitoring Center, Guangzhou 510006, China
| | - Jianwei Dai
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou 510436, PR China
| | - Xinming Wang
- State Key Laboratory of Organic Geochemistry and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, PR China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control, Guangzhou 510640, PR China
| | - Ping'an Peng
- State Key Laboratory of Organic Geochemistry and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, PR China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control, Guangzhou 510640, PR China
| | - Xinhui Bi
- State Key Laboratory of Organic Geochemistry and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, PR China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control, Guangzhou 510640, PR China.
| |
Collapse
|
5
|
Guertler A, Neu K, Lill D, Clanner-Engelshofen B, French LE, Reinholz M. Exploring the potential of omega-3 fatty acids in acne patients: A prospective intervention study. J Cosmet Dermatol 2024; 23:3295-3304. [PMID: 38982829 DOI: 10.1111/jocd.16434] [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: 03/26/2024] [Revised: 05/31/2024] [Accepted: 06/11/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND Omega-3 fatty acids (ω-3 FA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), are essential nutrients known for their anti-inflammatory properties, which involve reducing pro-inflammatory cytokines, eicosanoids, and insulin-like growth factor-1. This suggests their potential to alleviate acne severity, especially when deficits are present. AIMS To elevate EPA/DHA levels in acne patients through dietary intervention and supplementation, observing subsequent clinical effects. METHODS Over 16 weeks, 60 patients without prescription medication (n = 23 acne comedonica [AC], n = 37 acne papulopustulosa [AP]) adhered to a Mediterranean diet, incorporating oral algae-derived ω-3 FA supplementation (600 mg DHA/300 mg EPA week 1-8, 800 mg DHA/400 mg EPA week 8-16). At four visits (V1-V4), blood EPA/DHA levels were tracked using the HS-omega 3 index® (EPA/DHA (%) of total identified fatty acids in erythrocytes; target 8%-11%, deficit <8%, severe deficit <4%), alongside clinical assessments and standardized questionnaires. RESULTS At baseline, 98.3% of patients had an EPA/DHA deficit, with the mean HS-omega 3 index® rising from 4.9% at V1 to 8.3% at V4 (p < 0.001). AC showed significantly higher indices than AP at V4 (p = 0.035). Objective improvements in both inflammatory and non-inflammatory lesions were observed (p < 0.001). While self-reported appearance worsened in four patients, overall quality of life improved (p < 0.001), particularly in AP. Dietary triggers were more clearly defined than beneficial foods. Intake of cow's milk and dairy products reduced (p < 0.001). Compliance was good; no adverse events were reported. CONCLUSION Many acne patients have a ω-3 FA deficit. The HS-omega 3 index® can be increased by a Mediterranean diet and oral supplementation with algae-derived ω-3 FA. Acne severity improved significantly in patients with target ω-3 FA levels.
Collapse
Affiliation(s)
- Anne Guertler
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
| | - Katharina Neu
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
| | - Diana Lill
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
| | | | - Lars E French
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
- Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, Miller School of Medicine, University of Miami, Coral Gables, USA
| | - Markus Reinholz
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
6
|
Luo L, Li M, Huang W, Zhang S, Sun J, Zhang B, Hu W, Yu H. Obesity aggravates the role of C-reactive protein on knee pain: A cross-sectional analysis with NHANES data. Immun Inflamm Dis 2024; 12:e1371. [PMID: 39222043 PMCID: PMC11367918 DOI: 10.1002/iid3.1371] [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: 03/04/2024] [Revised: 06/27/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE To examine the relationship between C-reactive protein (CRP) and knee pain, and further explore whether this association is mediated by obesity. METHODS The population was derived from 1999 to 2004 National Health and Nutrition Examination Survey. Logistic regression was used to analyze the relationship between CRP and knee pain in three different models, and the linear trend was analyzed. A restricted cubic spline model to assess the nonlinear dose-response relationship between CRP and knee pain. Mediation analyses were used to assess the potential mediating role of obesity. Subgroup analyses and sensitivity analyses were performed to ensure robustness. RESULTS Compared with adults with lower CRP (first quartile), those with higher CRP had higher risks of knee pain (odds ratio 1.39, 95% confidence interval 1.12-1.72 in third quartile; 1.56, 1.25-1.95 in fourth quartile) after adjusting for covariates (except body mass index [BMI]), and the proportion mediated by BMI was 76.10% (p < .001). BMI and CRP were linear dose-response correlated with knee pain. The odds ratio for those with obesity compared with normal to knee pain was 2.27 (1.42-3.65) in the first quartile of CRP, 1.99 (1.38-2.86) in the second, 2.15 (1.38-3.33) in the third, and 2.92 (1.72-4.97) in the fourth. CONCLUSION Obesity mediated the systemic inflammation results in knee pain in US adults. Moreover, higher BMI was associated with higher knee pain risk in different degree CRP subgroups, supporting an important role of weight loss in reducing knee pain caused by systemic inflammation.
Collapse
Affiliation(s)
- Ling Luo
- Department of Epidemiology and Medical Statistics, School of Public HealthGuangdong Medical UniversityDongguanGuangdongChina
- The First Dongguan Affiliated HospitalGuangdong Medical UniversityDongguanGuangdongChina
- Dongguan Key Laboratory of Chronic Noncommunicable Disease PreventionGuangdong Medical UniversityDongguanGuangdongChina
| | - Mingzi Li
- Department of Epidemiology and Medical Statistics, School of Public HealthGuangdong Medical UniversityDongguanGuangdongChina
| | - Wenlong Huang
- Department of Epidemiology and Medical Statistics, School of Public HealthGuangdong Medical UniversityDongguanGuangdongChina
| | - Siying Zhang
- Institute of Scientific and Technological InformationNanjing University of Aeronautics and AstronauticsNanjingJiangsuChina
| | - Jianbo Sun
- The First Dongguan Affiliated HospitalGuangdong Medical UniversityDongguanGuangdongChina
| | - Bingsong Zhang
- Department of Epidemiology and Medical Statistics, School of Public HealthGuangdong Medical UniversityDongguanGuangdongChina
| | - Wei Hu
- Department of Epidemiology, School of Public HealthSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Haibing Yu
- Department of Epidemiology and Medical Statistics, School of Public HealthGuangdong Medical UniversityDongguanGuangdongChina
- The First Dongguan Affiliated HospitalGuangdong Medical UniversityDongguanGuangdongChina
- Dongguan Key Laboratory of Chronic Noncommunicable Disease PreventionGuangdong Medical UniversityDongguanGuangdongChina
| |
Collapse
|
7
|
Ahamed F, Eppler N, Jones E, Zhang Y. Understanding Macrophage Complexity in Metabolic Dysfunction-Associated Steatotic Liver Disease: Transitioning from the M1/M2 Paradigm to Spatial Dynamics. LIVERS 2024; 4:455-478. [PMID: 39328386 PMCID: PMC11426415 DOI: 10.3390/livers4030033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses metabolic dysfunction-associated fatty liver (MASL) and metabolic dysfunction-associated steatohepatitis (MASH), with MASH posing a risk of progression to cirrhosis and hepatocellular carcinoma (HCC). The global prevalence of MASLD is estimated at approximately a quarter of the population, with significant healthcare costs and implications for liver transplantation. The pathogenesis of MASLD involves intrahepatic liver cells, extrahepatic components, and immunological aspects, particularly the involvement of macrophages. Hepatic macrophages are a crucial cellular component of the liver and play important roles in liver function, contributing significantly to tissue homeostasis and swift responses during pathophysiological conditions. Recent advancements in technology have revealed the remarkable heterogeneity and plasticity of hepatic macrophage populations and their activation states in MASLD, challenging traditional classification methods like the M1/M2 paradigm and highlighting the coexistence of harmful and beneficial macrophage phenotypes that are dynamically regulated during MASLD progression. This complexity underscores the importance of considering macrophage heterogeneity in therapeutic targeting strategies, including their distinct ontogeny and functional phenotypes. This review provides an overview of macrophage involvement in MASLD progression, combining traditional paradigms with recent insights from single-cell analysis and spatial dynamics. It also addresses unresolved questions and challenges in this area.
Collapse
Affiliation(s)
- Forkan Ahamed
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, MS 1018, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Natalie Eppler
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, MS 1018, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Elizabeth Jones
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, MS 1018, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, MS 1018, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| |
Collapse
|
8
|
Teder-Laving M, Kals M, Reigo A, Ehin R, Objärtel T, Vaht M, Nikopensius T, Metspalu A, Kingo K. Genome-wide meta-analysis identifies novel loci conferring risk of acne vulgaris. Eur J Hum Genet 2024; 32:1136-1143. [PMID: 36922633 PMCID: PMC11368920 DOI: 10.1038/s41431-023-01326-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 02/02/2023] [Accepted: 02/21/2023] [Indexed: 03/18/2023] Open
Abstract
Acne vulgaris is a common chronic skin disorder presenting with comedones, cystic structures forming within the distal hair follicle, and in most cases additionally with inflammatory skin lesions on the face and upper torso. We performed a genome-wide association study and meta-analysis of data from 34,422 individuals with acne and 364,991 controls from three independent European-ancestry cohorts. We replicated 19 previously implicated genome-wide significant risk loci and identified four novel loci [11q12.2 (FADS2), 12q21.1 (LGR5), 17q25.3 (FASN), and 22q12.1 (ZNRF3-KREMEN1)], bringing the total number of reported acne risk loci to 50. Our meta-analysis results explain 9.4% of the phenotypic variance of acne. A polygenic model of acne risk variants showed that individuals in the top 5% of the risk percentiles had a 1.62-fold (95% CI 1.47-1.78) increased acne risk relative to individuals with average risk (20-80% on the polygenic risk score distribution). Our findings highlight the Wnt and MAPK pathways as key factors in the genetic predisposition to acne vulgaris, together with the effects of genetic variation on the structure and maintenance of the hair follicle and pilosebaceous unit. Two novel loci, 11q12.2 and 17q25.3, contain genes encoding key enzymes involved in lipid biosynthesis pathways.
Collapse
Affiliation(s)
- Maris Teder-Laving
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia.
| | - Mart Kals
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Anu Reigo
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Riin Ehin
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Health Technologies, Tallinn University of Technology, Tallinn, Estonia
- BioCC Ltd, Tartu, Estonia
| | - Telver Objärtel
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Mariliis Vaht
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tiit Nikopensius
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Andres Metspalu
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Külli Kingo
- Faculty of Medicine, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Tartu University Hospital, Tartu, Estonia
| |
Collapse
|
9
|
Coles CA, Woodman KG, Gibbs EM, Crosbie RH, White JD, Lamandé SR. Benfotiamine improves dystrophic pathology and exercise capacity in mdx mice by reducing inflammation and fibrosis. Hum Mol Genet 2024; 33:1339-1355. [PMID: 38710523 PMCID: PMC11262745 DOI: 10.1093/hmg/ddae066] [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: 12/15/2023] [Revised: 03/26/2024] [Accepted: 03/29/2024] [Indexed: 05/08/2024] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is a progressive and fatal neuromuscular disease. Cycles of myofibre degeneration and regeneration are hallmarks of the disease where immune cells infiltrate to repair damaged skeletal muscle. Benfotiamine is a lipid soluble precursor to thiamine, shown clinically to reduce inflammation in diabetic related complications. We assessed whether benfotiamine administration could reduce inflammation related dystrophic pathology. Benfotiamine (10 mg/kg/day) was fed to male mdx mice (n = 7) for 15 weeks from 4 weeks of age. Treated mice had an increased growth weight (5-7 weeks) and myofibre size at treatment completion. Markers of dystrophic pathology (area of damaged necrotic tissue, central nuclei) were reduced in benfotiamine mdx quadriceps. Grip strength was increased and improved exercise capacity was found in mdx treated with benfotiamine for 12 weeks, before being placed into individual cages and allowed access to an exercise wheel for 3 weeks. Global gene expression profiling (RNAseq) in the gastrocnemius revealed benfotiamine regulated signalling pathways relevant to dystrophic pathology (Inflammatory Response, Myogenesis) and fibrotic gene markers (Col1a1, Col1a2, Col4a5, Col5a2, Col6a2, Col6a2, Col6a3, Lum) towards wildtype levels. In addition, we observed a reduction in gene expression of inflammatory gene markers in the quadriceps (Emr1, Cd163, Cd4, Cd8, Ifng). Overall, these data suggest that benfotiamine reduces dystrophic pathology by acting on inflammatory and fibrotic gene markers and signalling pathways. Given benfotiamine's excellent safety profile and current clinical use, it could be used in combination with glucocorticoids to treat DMD patients.
Collapse
MESH Headings
- Animals
- Mice
- Mice, Inbred mdx
- Fibrosis/drug therapy
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/physiopathology
- Muscular Dystrophy, Duchenne/metabolism
- Inflammation/drug therapy
- Inflammation/genetics
- Inflammation/pathology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Male
- Thiamine/analogs & derivatives
- Thiamine/pharmacology
- Physical Conditioning, Animal
- Disease Models, Animal
Collapse
Affiliation(s)
- Chantal A Coles
- Murdoch Childrens Research Institute, The Royal Children’s Hospital, 50 Flemington Road, Parkville, Victoria 3052, Australia
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia
| | - Keryn G Woodman
- Murdoch Childrens Research Institute, The Royal Children’s Hospital, 50 Flemington Road, Parkville, Victoria 3052, Australia
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia
- Department of Genetics, Yale Medical School, Yale University, 333 Cedar Street, New Haven, Connecticut 06520, USA
| | - Elizabeth M Gibbs
- Department of Integrative Biology and Physiology, University of California, 612 Charles E Young Dr S, Los Angeles 90095, California, USA
- Center for Duchenne Muscular Dystrophy, University of California, 615 Charles E Young Dr S, Los Angeles 90095, California, USA
| | - Rachelle H Crosbie
- Department of Integrative Biology and Physiology, University of California, 612 Charles E Young Dr S, Los Angeles 90095, California, USA
- Center for Duchenne Muscular Dystrophy, University of California, 615 Charles E Young Dr S, Los Angeles 90095, California, USA
- Department of Neurology, David Geffen School of Medicine, University of California, 610 Charles E Young Dr S, Los Angeles, California 90095, USA
| | - Jason D White
- Murdoch Childrens Research Institute, The Royal Children’s Hospital, 50 Flemington Road, Parkville, Victoria 3052, Australia
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia
- Charles Sturt University, Office of the Deputy Vice Chancellor Research, Boorooma Street, Wagga Wagga, NSW 2678, Australia
| | - Shireen R Lamandé
- Murdoch Childrens Research Institute, The Royal Children’s Hospital, 50 Flemington Road, Parkville, Victoria 3052, Australia
- Department of Paediatrics, University of Melbourne, 50 Flemington Road, Parkville, Victoria 3052, Australia
| |
Collapse
|
10
|
Choi D, Jang SJ, Choi S, Park S, Kim WK, Lee G, Lee C, Ko G. Oral Administration of Limosilactobacillus reuteri KBL346 Ameliorates Influenza Virus A/PR8 Infection in Mouse. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10301-8. [PMID: 38949757 DOI: 10.1007/s12602-024-10301-8] [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] [Accepted: 05/31/2024] [Indexed: 07/02/2024]
Abstract
Influenza virus infection is an important public-health concern because of its high transmissibility and potential for severe complications. To mitigate the severity and complications of influenza, probiotics containing Lactobacillus are used and generally recognized as safe. We evaluated the anti-influenza effect of Limosilactobacillus reuteri (L. reuteri) KBL346, isolated from the fecel sample of healthy South Koreans, in mice. BALB/c mice were orally administered live and heat-inactivated L. reuteri KBL346. After infection with influenza virus (A/Puerto Rico/8/34) 0.5 times the 50% lethal dose (LD50), body weight loss was improved and recovery was accelerated. Furthermore, L. reuteri KBL346 improved body weight loss and survival rate of mice infected with 4 times the LD50 of influenza virus. Heat-inactivated L. reuteri KBL346 reduced the viral titer in the lung and the plasma immunoglobulin G level. Expression levels of genes encoding inflammatory cytokines, such as interferon-γ and toll-like receptor 2 (Tlr2), were decreased in the lung tissues of mice administered L. reuteri KBL346. Live and heat-inactivated L. reuteri KBL346 increased the expression level of Adamts4, which promotes recovery after infection, and decreased that of Tlr2. The α-diversity of the gut microbiome was modulated by the administration of L. reuteri KBL346. In addition, the structure of the gut microbial community differed according to the degree of weight loss. L. reuteri KBL346 has the potential to alleviate disease severity and improve histopathological changes in mice infected with influenza A/PR8, suggesting its efficacy as a probiotic against influenza infection.
Collapse
Affiliation(s)
- Doseon Choi
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Sung Jae Jang
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- KoBioLabs, Inc, Seoul, Republic of Korea
| | - Sueun Choi
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - SungJun Park
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- KoBioLabs, Inc, Seoul, Republic of Korea
- N-Bio, Seoul National University, Seoul, Republic of Korea
| | - Woon-Ki Kim
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| | - Giljae Lee
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| | - Cheonghoon Lee
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea.
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea.
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH, USA.
| | - GwangPyo Ko
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea.
- KoBioLabs, Inc, Seoul, Republic of Korea.
- N-Bio, Seoul National University, Seoul, Republic of Korea.
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Zhang Z, Peng Z, Wang R, Guo X, Gao J. Metabolomic analysis reveals macrophage metabolic reprogramming and polarization under different nutritional cues. Clin Chim Acta 2024; 560:119735. [PMID: 38772523 DOI: 10.1016/j.cca.2024.119735] [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: 12/04/2023] [Revised: 04/05/2024] [Accepted: 05/14/2024] [Indexed: 05/23/2024]
Abstract
BACKGROUND AND AIMS Obesity-induced chronic inflammation and metabolic abnormalities are highly relevant to the functional dysregulation of macrophages, especially under obese conditions. Hyperglycemia and hyperlipidemia, central to obesity, directly alter macrophage activity. However, the impacts of different nutritional cues on the intricate metabolic networks in macrophages remain unclear. MATERIALS AND METHODS In this study, we employed metabolomic approaches to examine the metabolic responses of macrophages to high glucose, high fat and their coexistence, aiming to delineate the molecular mechanisms of nutritional factors on macrophage activation and obesity-related diseases from a metabolic perspective. RESULTS Our findings revealed that different nutritional conditions could reprogram key metabolism in macrophages. Additionally, we identified a metabolite derived from macrophages, Long-Chain Phosphatidylcholine (LPC), which exerts beneficial effects on obese mice. It ameliorates the obesity phenotype and improves glucose metabolism profiles. This discovery suggests that LPC has a significant therapeutic potential in the context of obesity-induced metabolic dysfunctions. Our study unveils the metabolic phenotype of macrophages in high-fat and high-sugar environments and uncovers a macrophage-derived metabolite that significantly ameliorates the obesity phenotype. CONCLUSION This finding reveals a potential dialogue mechanism between macrophages and adipocytes, shedding light on the complex interplay of immune and metabolic systems in obesity. This discovery not only enhances our understanding of obesity's underlying mechanisms but also opens up new avenues for therapeutic interventions targeting macrophage-adipocyte interactions.
Collapse
Affiliation(s)
- Zhongxiao Zhang
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhou Peng
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Wang
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xirong Guo
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jianfang Gao
- Endocrinology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
12
|
Lu L, Li J, Liu L, Wang C, Xie Y, Yu X, Tian L. Grape seed extract prevents oestrogen deficiency-induced bone loss by modulating the gut microbiota and metabolites. Microb Biotechnol 2024; 17:e14485. [PMID: 38850270 PMCID: PMC11162104 DOI: 10.1111/1751-7915.14485] [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: 10/23/2023] [Revised: 04/07/2024] [Accepted: 05/09/2024] [Indexed: 06/10/2024] Open
Abstract
Proanthocyanidin-rich grape seed extract (GSE) has been shown to have the potential to protect bones, although the underlying mechanism remains unknown. The current study aims to explore GSE's preventive and therapeutic impact on bone loss induced by oestrogen deficiency and the underlying mechanism through the gut microbiota (GM) and metabolomic responses. In oestrogen-deficient ovariectomized (OVX) mice, GSE ameliorated bone loss by inhibiting the expansion of bone marrow adipose tissue (BMAT), restoring BMAT lipolysis and promoting bone formation. GSE regulated OVX-induced GM dysbiosis by reducing the abundance of opportunistic pathogenic bacteria, such as Alistipes, Turicibacter and Romboutsia, while elevating the abundance of beneficial bacteria, such as Bifidobacterium. The modified GM primarily impacted lipid and amino acid metabolism. Furthermore, the serum metabolites of GSE exhibited a significant enrichment in lipid metabolism. In summary, GSE shows potential as a functional food for preventing oestrogen deficiency-induced bone loss by modulating GM and metabolite-mediated lipid metabolism.
Collapse
Affiliation(s)
- Lingyun Lu
- Division of Internal Medicine, Institute of Integrated Traditional Chinese and Western Medicine, West China HospitalSichuan UniversityChengduChina
| | - Jiao Li
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China HospitalSichuan UniversityChengduChina
| | - Lu Liu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, West China HospitalSichuan UniversityChengduChina
| | - Cui Wang
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, West China HospitalSichuan UniversityChengduChina
| | - Ying Xie
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, West China HospitalSichuan UniversityChengduChina
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, West China HospitalSichuan UniversityChengduChina
| | - Li Tian
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
13
|
Nguyen NTA, Jiang Y, McQuade JL. Eating away cancer: the potential of diet and the microbiome for shaping immunotherapy outcome. Front Immunol 2024; 15:1409414. [PMID: 38873602 PMCID: PMC11169628 DOI: 10.3389/fimmu.2024.1409414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024] Open
Abstract
The gut microbiome (GMB) plays a substantial role in human health and disease. From affecting gut barrier integrity to promoting immune cell differentiation, the GMB is capable of shaping host immunity and thus oncogenesis and anti-cancer therapeutic response, particularly with immunotherapy. Dietary patterns and components are key determinants of GMB composition, supporting the investigation of the diet-microbiome-immunity axis as a potential avenue to enhance immunotherapy response in cancer patients. As such, this review will discuss the role of the GMB and diet on anti-cancer immunity. We demonstrate that diet affects anti-cancer immunity through both GMB-independent and GMB-mediated mechanisms, and that different diet patterns mold the GMB's functional and taxonomic composition in distinctive ways. Dietary modulation therefore shows promise as an intervention for improving cancer outcome; however, further and more extensive research in human cancer populations is needed.
Collapse
Affiliation(s)
| | | | - Jennifer L. McQuade
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
14
|
Kang N, Ji Z, Li Y, Gao J, Wu X, Zhang X, Duan Q, Zhu C, Xu Y, Wen L, Shi X, Liu W. Metabolite-derived damage-associated molecular patterns in immunological diseases. FEBS J 2024; 291:2051-2067. [PMID: 37432883 DOI: 10.1111/febs.16902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 06/05/2023] [Accepted: 07/10/2023] [Indexed: 07/13/2023]
Abstract
Damage-associated molecular patterns (DAMPs) are typically derived from the endogenous elements of necrosis cells and can trigger inflammatory responses by activating DAMPs-sensing receptors on immune cells. Failure to clear DAMPs may lead to persistent inflammation, thereby contributing to the pathogenesis of immunological diseases. This review focuses on a newly recognized class of DAMPs derived from lipid, glucose, nucleotide, and amino acid metabolic pathways, which are then termed as metabolite-derived DAMPs. This review summarizes the reported molecular mechanisms of these metabolite-derived DAMPs in exacerbating inflammation responses, which may attribute to the pathology of certain types of immunological diseases. Additionally, this review also highlights both direct and indirect clinical interventions that have been explored to mitigate the pathological effects of these DAMPs. By summarizing our current understanding of metabolite-derived DAMPs, this review aims to inspire future thoughts and endeavors on targeted medicinal interventions and the development of therapies for immunological diseases.
Collapse
Affiliation(s)
- Na Kang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zhenglin Ji
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Yuxin Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Ji Gao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Xinfeng Wu
- Department of Rheumatology and Immunology, the First Affiliated Hospital, and College of Clinical Medical of Henan University of Science and Technology, Luoyang, China
| | - Xiaoyang Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Qinghui Duan
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Can Zhu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Yue Xu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Luyao Wen
- Department of Rheumatology and Immunology, the First Affiliated Hospital, and College of Clinical Medical of Henan University of Science and Technology, Luoyang, China
| | - Xiaofei Shi
- Department of Rheumatology and Immunology, the First Affiliated Hospital, and College of Clinical Medical of Henan University of Science and Technology, Luoyang, China
| | - Wanli Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| |
Collapse
|
15
|
Reese L, Niepmann ST, Düsing P, Hänschke L, Beiert T, Zimmer S, Nickenig G, Bauer R, Jansen F, Zietzer A. Loss of ceramide synthase 5 inhibits the development of experimentally induced aortic valve stenosis. Acta Physiol (Oxf) 2024; 240:e14140. [PMID: 38546351 DOI: 10.1111/apha.14140] [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: 09/20/2023] [Revised: 03/04/2024] [Accepted: 03/14/2024] [Indexed: 04/24/2024]
Abstract
AIM Inflammation and calcification are hallmarks in the development of aortic valve stenosis (AVS). Ceramides mediate inflammation and calcification in the vascular tissue. The highly abundant d18:1,16:0 ceramide (C16) has been linked to increased cardiovascular mortality and obesity. In this study, we investigate the role of ceramide synthase 5 (CerS5), a critical enzyme for C16 ceramide synthesis, in the development of AVS, particularly in conjunction with a high-fat/high-cholesterol diet (Western diet, WD). METHODS We used wild-type (WT) and CerS5-/- mice on WD or normal chow in a wire injury model. We measured the peak velocity to determine AVS development and performed histological analysis of the aortic valve area, immune cell infiltration (CD68 staining), and calcification (von Kossa). In vitro experiments involved measuring the calcification of human aortic valvular interstitial cells (VICs) and evaluating cytokine release from THP-1 cells, a human leukemia monocytic-like cell line, following CerS5 knockdown. RESULTS CerS5-/- mice showed a reduced peak velocity compared to WT only in the experiment with WD. Likewise, we observed reduced immune cell infiltration and calcification in the aortic valve of CerS5-/- mice, but only on WD. In vitro, calcification was reduced after knockdown of CerS5 in VICs, while THP-1 cells exhibited a decreased inflammatory response following CerS5 knockdown. CONCLUSION We conclude that CerS5 is an important mediator for the development of AVS in mice on WD and regulates critical pathophysiological hallmarks of AVS formation. CerS5 is therefore an interesting target for pharmacological therapy and merits further investigation.
Collapse
Affiliation(s)
- Laurine Reese
- Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Sven Thomas Niepmann
- Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Philip Düsing
- Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Lea Hänschke
- Life & Medical Sciences Institute (LIMES), Genetics & Molecular Physiology, University of Bonn, Bonn, Germany
| | - Thomas Beiert
- Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Sebastian Zimmer
- Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Georg Nickenig
- Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Reinhard Bauer
- Life & Medical Sciences Institute (LIMES), Genetics & Molecular Physiology, University of Bonn, Bonn, Germany
| | - Felix Jansen
- Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Andreas Zietzer
- Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
16
|
Otunla AA, Shanmugarajah K, Davies AH, Shalhoub J. Lipotoxicity and immunometabolism in ischemic acute kidney injury: current perspectives and future directions. Front Pharmacol 2024; 15:1355674. [PMID: 38464721 PMCID: PMC10924325 DOI: 10.3389/fphar.2024.1355674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/12/2024] [Indexed: 03/12/2024] Open
Abstract
Dysregulated lipid metabolism is implicated in the pathophysiology of a range of kidney diseases. The specific mechanisms through which lipotoxicity contributes to acute kidney injury (AKI) remain poorly understood. Herein we review the cardinal features of lipotoxic injury in ischemic kidney injury; lipid accumulation and mitochondrial lipotoxicity. We then explore a new mechanism of lipotoxicity, what we define as "immunometabolic" lipotoxicity, and discuss the potential therapeutic implications of targeting this lipotoxicity using lipid lowering medications.
Collapse
Affiliation(s)
- Afolarin A. Otunla
- Department of Surgical Biotechnology, University College London, London, United Kingdom
| | | | - Alun H. Davies
- UK and Imperial Vascular Unit, Section of Vascular Surgery, Department of Surgery and Cancer, Imperial College London, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Joseph Shalhoub
- UK and Imperial Vascular Unit, Section of Vascular Surgery, Department of Surgery and Cancer, Imperial College London, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
17
|
Liu S, He M, Jiang J, Duan X, Chai B, Zhang J, Tao Q, Chen H. Triggers for the onset and recurrence of psoriasis: a review and update. Cell Commun Signal 2024; 22:108. [PMID: 38347543 PMCID: PMC10860266 DOI: 10.1186/s12964-023-01381-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/02/2023] [Indexed: 02/15/2024] Open
Abstract
Psoriasis is an immune-mediated inflammatory skin disease, involving a complex interplay between genetic and environmental factors. Previous studies have demonstrated that genetic factors play a major role in the pathogenesis of psoriasis. However, non-genetic factors are also necessary to trigger the onset and recurrence of psoriasis in genetically predisposed individuals, which include infections, microbiota dysbiosis of the skin and gut, dysregulated lipid metabolism, dysregulated sex hormones, and mental illness. Psoriasis can also be induced by other environmental triggers, such as skin trauma, unhealthy lifestyles, and medications. Understanding how these triggers play a role in the onset and recurrence of psoriasis provides insights into psoriasis pathogenesis, as well as better clinical administration. In this review, we summarize the triggers for the onset and recurrence of psoriasis and update the current evidence on the underlying mechanism of how these factors elicit the disease. Video Abstract.
Collapse
Grants
- No.82173423, No.81974475, No.82103731 the National Natural Science Foundation of China
- No.82173423, No.81974475, No.82103731 the National Natural Science Foundation of China
- No.82173423, No.81974475, No.82103731 the National Natural Science Foundation of China
- Basic Research Project, No. JCYJ20190809103805589 Shenzhen Natural Science Foundation
- Basic Research Project, No. JCYJ20190809103805589 Shenzhen Natural Science Foundation
- Basic Research Project, No. JCYJ20190809103805589 Shenzhen Natural Science Foundation
- Key Project, No.2019003 Shenzhen Nanshan District Science and Technology Project
- Key Project, No.2019003 Shenzhen Nanshan District Science and Technology Project
- Key Project, No.2019003 Shenzhen Nanshan District Science and Technology Project
Collapse
Affiliation(s)
- Suwen Liu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mengwen He
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Jian Jiang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoru Duan
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bao Chai
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
- Department of Dermatology, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, 518052, China
| | - Jingyu Zhang
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
- Department of Dermatology, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, 518052, China
| | - Qingxiao Tao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hongxiang Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China.
| |
Collapse
|
18
|
Huang L, Yang S, Yu X, Fang F, Zhu L, Wang L, Zhang X, Yang C, Qian Q, Zhu T. Association of different cell types and inflammation in early acne vulgaris. Front Immunol 2024; 15:1275269. [PMID: 38357543 PMCID: PMC10864487 DOI: 10.3389/fimmu.2024.1275269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/17/2024] [Indexed: 02/16/2024] Open
Abstract
Acne vulgaris, one of the most common skin diseases, is a chronic cutaneous inflammation of the upper pilosebaceous unit (PSU) with complex pathogenesis. Inflammation plays a central role in the pathogenesis of acne vulgaris. During the inflammatory process, the innate and adaptive immune systems are coordinately activated to induce immune responses. Understanding the infiltration and cytokine secretion of differential cells in acne lesions, especially in the early stages of inflammation, will provide an insight into the pathogenesis of acne. The purpose of this review is to synthesize the association of different cell types with inflammation in early acne vulgaris and provide a comprehensive understanding of skin inflammation and immune responses.
Collapse
Affiliation(s)
- Lei Huang
- Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shuyun Yang
- Department of Dermatology, The People’s Hospital of Baoshan, Baoshan, Yunnan, China
| | - Xiuqin Yu
- Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fumin Fang
- Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liping Zhu
- Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lu Wang
- Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoping Zhang
- Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Changzhi Yang
- Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qihong Qian
- Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tingting Zhu
- Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
19
|
Cha SJ, Kim SS, Shin JH, Seo SR. Peiminine Exerts Its Anti-Acne Effects by Regulating the NF-κB Pathway. Antioxidants (Basel) 2024; 13:131. [PMID: 38275656 PMCID: PMC10812726 DOI: 10.3390/antiox13010131] [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: 12/11/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Peiminine is the main natural alkaloid compound extracted from the Chinese herb Fritillaria. Although peiminine is known for its antioxidant and anti-inflammatory effects in conditions such as mastitis and arthritis, its impact on inflammation induced by Cutibacterisum acnes (C. acnes) has not been explored. The aim of this study was to investigate the effect of peiminine on C. acnes-induced inflammatory responses in the skin and to identify the underlying mechanism involved. We discovered that peiminine inhibits the C. acnes-induced expression of inflammatory mediators such as pro-interleukin-1β (pro-IL-1β), cyclooxygenase-2 (COX-2), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) in mouse bone marrow-derived macrophages (BMDMs). Peiminine suppressed the activation of nuclear factor-kappa B (NF-κB) without affecting the activation of mitogen-activated protein kinase (MAPK) pathways such as JNK, ERK, and p38 MAPK. In addition, we found that peiminine suppressed inflammatory cytokine expression and ameliorated histological symptoms in C. acnes-induced mouse skin. Our study is the first to provide evidence that peiminine has an inhibitory effect on acne, and it points toward the potential of incorporating peiminine into cosmetic and pharmaceutical formulations for acne treatment.
Collapse
Affiliation(s)
- So Jin Cha
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea; (S.J.C.); (J.H.S.)
| | - Seon Sook Kim
- Institute of Life Science, Kangwon National University, Chuncheon 24341, Republic of Korea;
| | - Jin Hak Shin
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea; (S.J.C.); (J.H.S.)
| | - Su Ryeon Seo
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea; (S.J.C.); (J.H.S.)
- Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
20
|
Wang W, Kou J, Long J, Wang T, Zhang M, Wei M, Xie Q. GC/MS and LC/MS serum metabolomic analysis of Chinese LN patients. Sci Rep 2024; 14:1523. [PMID: 38233574 PMCID: PMC10794181 DOI: 10.1038/s41598-024-52137-w] [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: 05/30/2023] [Accepted: 01/14/2024] [Indexed: 01/19/2024] Open
Abstract
China, being a densely populated nation, faces a substantial economic burden due to a high incidence of lupus nephritis (LN) cases. The concealed onset of LN has resulted in many individuals have missed the optimal timing for treatment. The aim of the research is to study the serum metabolomics of Chinese LN patients using gas chromatography (GC)/mass spectrometry (MS) and liquid chromatography (LC)/MS to identify potential diagnostic markers. Fifty LN patients and fifty normal controls, matched for Body Mass Index (BMI) and age, were selected. Serum analysis was conducted using GC/MS and LC/MS, followed by multivariate statistical analysis. Various multidimensional analyses, including principal component analysis, partial least squares discrimination analysis, and orthogonal partial least squares discrimination analysis, along with one-dimensional analyses such as t-tests, were performed. Metabolites with variable importance in projection value > 1 and a p-value < 0.05 were considered critical biomarkers for LN. Furthermore, identified biomarkers delineated relevant metabolic pathways, and a metabolic pathway map was obtained from the database. Forty-one metabolites were identified as potential LN biomarkers, primarily associated with immune regulation, energy metabolism, intestinal microbial metabolism, renal damage, and oxidative stress. The potential for diagnosing LN and other diseases through metabolomics is demonstrated. Future research should explore larger sample sizes, metabolomic comparisons across different diseases and health states, and integration of metabolomics with clinical diagnostics. Such studies will enhance the understanding of metabolomics in medical diagnosis and provide robust support for its practical application.
Collapse
Affiliation(s)
- Wei Wang
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000, People's Republic of China
| | - Jun Kou
- Department of Ultrasound Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders (Chongqing Key Laboratory of Pediatrics), Chongqing, 400010, China
| | - Jie Long
- Department of Nephrology, Honghui Hospital, Xi'an Jiaotong University College of Medicine, No.555 Youyi East Road, Beilin District, Xi'an, 710054, Shaanxi, People's Republic of China
| | - Tao Wang
- Department of Rheumatism and Immunology, The General Hospital of Western Theater Command, Tianhui Road 270, Chengdu, 610000, People's Republic of China
| | - Mingmei Zhang
- Department of Rheumatism and Immunology, The General Hospital of Western Theater Command, Tianhui Road 270, Chengdu, 610000, People's Republic of China
| | - Meng Wei
- Department of Rheumatism and Immunology, The General Hospital of Western Theater Command, Tianhui Road 270, Chengdu, 610000, People's Republic of China.
| | - Qingyun Xie
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000, People's Republic of China.
| |
Collapse
|
21
|
Seliga AK, Zabłocki K, Bandorowicz-Pikuła J. Palmitate Stimulates Expression of the von Willebrand Factor and Modulates Toll-like Receptors Level and Activity in Human Umbilical Vein Endothelial Cells (HUVECs). Int J Mol Sci 2023; 25:254. [PMID: 38203423 PMCID: PMC10779284 DOI: 10.3390/ijms25010254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/12/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
An increased concentration of palmitate in circulation is one of the most harmful factors in obesity. The von Willebrand factor (vWF), a protein involved in haemostasis, is produced and secreted by the vascular endothelium. An increased level of vWF in obese patients is associated with thrombosis and cardiovascular disease. The aim of this study was to investigate a palmitate effect on vWF in endothelial cells and understand the mechanisms of palmitate-activated signalling. Human umbilical vein endothelial cells (HUVECs) incubated in the presence of palmitate, exhibited an increased VWF gene expression, vWF protein maturation, and stimulated vWF secretion. Cardamonin, a Nuclear Factor kappa B (NF-κB) inhibitor, abolished the palmitate effect on VWF expression. The inhibition of Toll-like receptor (TLR) 2 with C29 resulted in the TLR4 overactivation in palmitate-treated cells. Palmitate, in the presence of TLR4 inhibitor TAK-242, leads to a higher expression of TLR6, CD36, and TIRAP. The silencing of TLR4 resulted in an increase in TLR2 level and vice versa. The obtained results indicate a potential mechanism of obesity-induced thrombotic complication caused by fatty acid activation of NF-κB signalling and vWF upregulation and help to identify various compensatory mechanisms related to TLR4 signal transduction.
Collapse
Affiliation(s)
| | | | - Joanna Bandorowicz-Pikuła
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology PAS, 3 Pasteur Str., 02-093 Warsaw, Poland; (A.K.S.); (K.Z.)
| |
Collapse
|
22
|
Pham PT, Bavuu O, Kim‐Kaneyama J, Lei X, Yamamoto T, Otsuka K, Suto K, Kusunose K, Yagi S, Yamada H, Soeki T, Shimabukuro M, Barber GN, Sata M, Fukuda D. Innate Immune System Regulated by Stimulator of Interferon Genes, a Cytosolic DNA Sensor, Regulates Endothelial Function. J Am Heart Assoc 2023; 12:e030084. [PMID: 37947148 PMCID: PMC10727293 DOI: 10.1161/jaha.123.030084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 09/08/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Sterile inflammation caused by metabolic disorders impairs endothelial function; however, the underlying mechanism by which hyperglycemia induces inflammation remains obscure. Recent studies have suggested that stimulator of interferon genes (STING), a key cytosolic DNA sensor in the innate immune system, contributes to the pathogenesis of inflammatory diseases. This study examines the role of the STING in endothelial dysfunction in streptozotocin-induced diabetic mice. METHODS AND RESULTS Injection of streptozotocin promoted the expression of STING and DNA damage markers in the aorta of wild-type mice. Streptozotocin elevated blood glucose and lipid levels in both wild-type and STING-deficient mice, which showed no statistical differences. Genetic deletion of STING ameliorated endothelial dysfunction as determined by the vascular relaxation in response to acetylcholine (P<0.001) and increased endothelial nitric oxide synthase phosphorylation in the aorta (P<0.05) in STZ-injected mice. Endothelium-independent vascular response to sodium nitroprusside did not differ. Treatment with a direct STING agonist, cyclic GMP-AMP, or mitochondrial DNA increased inflammatory molecule expression (eg, VCAM1 and IFNB) and decreased endothelial nitric oxide synthase phosphorylation in human umbilical vein endothelial cells, partially through the STING pathway. Cyclic GMP-AMP significantly impaired endothelial function of aortic segments obtained from wild-type mice, which was ameliorated in the presence of C-176, a STING inhibitor, or a neutralizing interferon-β antibody. Furthermore, the administration of C-176 ameliorated endothelial dysfunction in STZ-induced diabetic mice (P<0.01). CONCLUSIONS The DNA damage response regulated by STING impairs endothelial function. STING signaling may be a potential therapeutic target of endothelial dysfunction caused by hyperglycemia.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Experimental/metabolism
- Membrane Proteins/metabolism
- Membrane Proteins/genetics
- Nitric Oxide Synthase Type III/metabolism
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/physiopathology
- Human Umbilical Vein Endothelial Cells/metabolism
- Human Umbilical Vein Endothelial Cells/drug effects
- Immunity, Innate
- Humans
- Mice, Inbred C57BL
- Male
- Mice, Knockout
- Signal Transduction
- Phosphorylation
- Vasodilation/drug effects
- Mice
- DNA Damage
- DNA, Mitochondrial/genetics
- DNA, Mitochondrial/metabolism
- Nucleotides, Cyclic/metabolism
- Nucleotides, Cyclic/pharmacology
Collapse
Affiliation(s)
- Phuong Tran Pham
- Department of Cardiovascular MedicineTokushima University Graduate School of Biomedical SciencesTokushimaJapan
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Oyunbileg Bavuu
- Department of Cardiovascular MedicineTokushima University Graduate School of Biomedical SciencesTokushimaJapan
| | | | - Xiao‐Feng Lei
- Department of BiochemistryShowa University School of MedicineTokyoJapan
| | - Takayuki Yamamoto
- Department of Cardiovascular MedicineOsaka Metropolitan University Graduate School of MedicineOsakaJapan
| | - Kenichiro Otsuka
- Department of Cardiovascular MedicineOsaka Metropolitan University Graduate School of MedicineOsakaJapan
| | - Kumiko Suto
- Department of Cardiovascular MedicineTokushima University Graduate School of Biomedical SciencesTokushimaJapan
| | - Kenya Kusunose
- Department of Cardiovascular MedicineTokushima University Graduate School of Biomedical SciencesTokushimaJapan
- Department of Cardiovascular Medicine, Nephrology, and Neurology, Graduate School of MedicineUniversity of the RyukyusOkinawaJapan
| | - Shusuke Yagi
- Department of Cardiovascular MedicineTokushima University Graduate School of Biomedical SciencesTokushimaJapan
| | - Hirotsugu Yamada
- Department of Community Medicine for CardiologyTokushima University Graduate School of Biomedical SciencesTokushimaJapan
| | - Takeshi Soeki
- Department of Community Medicine and Medical ScienceTokushima University Graduate School of Biomedical SciencesTokushimaJapan
| | - Michio Shimabukuro
- Department of Diabetes, Endocrinology and MetabolismFukushima Medical University School of MedicineFukushimaJapan
| | - Glen N. Barber
- Department of Cell BiologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Masataka Sata
- Department of Cardiovascular MedicineTokushima University Graduate School of Biomedical SciencesTokushimaJapan
| | - Daiju Fukuda
- Department of Cardiovascular MedicineTokushima University Graduate School of Biomedical SciencesTokushimaJapan
- Department of Cardiovascular MedicineOsaka Metropolitan University Graduate School of MedicineOsakaJapan
| |
Collapse
|
23
|
Ellis RJ, Marquine MJ, Kaul M, Fields JA, Schlachetzki JCM. Mechanisms underlying HIV-associated cognitive impairment and emerging therapies for its management. Nat Rev Neurol 2023; 19:668-687. [PMID: 37816937 PMCID: PMC11052664 DOI: 10.1038/s41582-023-00879-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2023] [Indexed: 10/12/2023]
Abstract
People living with HIV are affected by the chronic consequences of neurocognitive impairment (NCI) despite antiretroviral therapies that suppress viral replication, improve health and extend life. Furthermore, viral suppression does not eliminate the virus, and remaining infected cells may continue to produce viral proteins that trigger neurodegeneration. Comorbidities such as diabetes mellitus are likely to contribute substantially to CNS injury in people living with HIV, and some components of antiretroviral therapy exert undesirable side effects on the nervous system. No treatment for HIV-associated NCI has been approved by the European Medicines Agency or the US Food and Drug Administration. Historically, roadblocks to developing effective treatments have included a limited understanding of the pathophysiology of HIV-associated NCI and heterogeneity in its clinical manifestations. This heterogeneity might reflect multiple underlying causes that differ among individuals, rather than a single unifying neuropathogenesis. Despite these complexities, accelerating discoveries in HIV neuropathogenesis are yielding potentially druggable targets, including excessive immune activation, metabolic alterations culminating in mitochondrial dysfunction, dysregulation of metal ion homeostasis and lysosomal function, and microbiome alterations. In addition to drug treatments, we also highlight the importance of non-pharmacological interventions. By revisiting mechanisms implicated in NCI and potential interventions addressing these mechanisms, we hope to supply reasons for optimism in people living with HIV affected by NCI and their care providers.
Collapse
Affiliation(s)
- Ronald J Ellis
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.
| | - María J Marquine
- Department of Medicine, Duke University, Durham, NC, USA
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Marcus Kaul
- School of Medicine, Division of Biomedical Sciences, University of California Riverside, Riverside, CA, USA
| | - Jerel Adam Fields
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
24
|
Unterberger S, Mullen L, Flint MS, Sacre S. Multiple TLRs elicit alternative NLRP3 inflammasome activation in primary human monocytes independent of RIPK1 kinase activity. Front Immunol 2023; 14:1092799. [PMID: 37954581 PMCID: PMC10639122 DOI: 10.3389/fimmu.2023.1092799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
The canonical NOD-like receptor family pyrin domain containing 3 (NLRP3) pathway involves a priming step to induce pro-IL-1β followed by a secondary signal such as K+ efflux to activate inflammasome formation. This then leads to the maturation of IL-1β and the formation of gasdermin D (GSDMD) pores that initiate pyroptosis and mediate IL-1β release. In contrast, primary human monocytes also engage an alternative pathway in response to toll-like receptor (TLR) 4 activation, without the need for a secondary signal. Data from a monocyte-like cell line suggest that the alternative pathway functions via the TLR adaptor protein TIR-domain-containing adapter-inducing interferon-β (TRIF), receptor-interacting protein kinase 1 (RIPK1), FAS-associated death domain (FADD) and caspase-8 upstream of NLRP3 activation, but in the absence of K+ efflux or pyroptosis. Usage of the alternative pathway by other members of the TLR family that induce IL-1β but do not signal through TRIF, has yet to be explored in primary human monocytes. Furthermore, the mechanism by which IL-1β is released from monocytes remains unclear. Therefore, this study investigated if the alternative NLRP3 inflammasome pathway is initiated following activation of TLRs other than TLR4, and if GSDMD was necessary for the release of IL-1β. Monocytes were stimulated with ligands that activate TLR1/2, TLR2/6, TLR4 and TLR7 and/or TLR8 (using a dual ligand). Similar to TLR4, all of the TLRs investigated induced IL-1β release in a NLRP3 and caspase-1 dependent manner, indicating that TRIF may not be an essential upstream component of the alternative pathway. Furthermore, inhibition of RIPK1 kinase activity had no effect on IL-1β release. Although IL-1β was released independently of K+ efflux and pyroptosis, it was significantly reduced by an inhibitor of GSDMD. Therefore, it is feasible that low level GSDMD pore formation may facilitate the release of IL-1β from the cell, but not be present in sufficient quantities to initiate pyroptosis. Together these data suggest that the alternative pathway operates independently of RIPK1 kinase activity, downstream of diverse TLRs including TLR4 in primary human monocytes and supports the potential for IL-1β release via GSDMD pores alongside other unconventional secretory pathways.
Collapse
Affiliation(s)
- Sarah Unterberger
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| | - Lisa Mullen
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| | - Melanie S. Flint
- Centre for Stress and Age-related Disease, School of Applied Sciences, University of Brighton, Brighton, United Kingdom
| | - Sandra Sacre
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| |
Collapse
|
25
|
Zhang W, Lang R. Macrophage metabolism in nonalcoholic fatty liver disease. Front Immunol 2023; 14:1257596. [PMID: 37868954 PMCID: PMC10586316 DOI: 10.3389/fimmu.2023.1257596] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) and its inflammatory and often progressive subtype nonalcoholic steatohepatitis (NASH), have emerged as significant contributors to hepatic morbidity worldwide. The pathophysiology of NAFLD/NASH is multifaceted, variable, and remains incompletely understood. The pivotal role of liver-resident and recruited macrophages in the pathogenesis of NAFLD and NASH is widely acknowledged as a crucial factor in innate immunity. The remarkable plasticity of macrophages enables them to assume diverse activation and polarization states, dictated by their immunometabolism microenvironment and functional requirements. Recent studies in the field of immunometabolism have elucidated that alterations in the metabolic profile of macrophages can profoundly influence their activation state and functionality, thereby influencing various pathological processes. This review primarily focuses on elucidating the polarization and activation states of macrophages, highlighting the correlation between their metabolic characteristics and the transition from pro-inflammatory to anti-inflammatory phenotypes. Additionally, we explore the potential of targeting macrophage metabolism as a promising therapeutic approach for the management of NAFLD/NASH.
Collapse
Affiliation(s)
| | - Ren Lang
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
26
|
Sim J, Park J, Moon JS, Lim J. Dysregulation of inflammasome activation in glioma. Cell Commun Signal 2023; 21:239. [PMID: 37723542 PMCID: PMC10506313 DOI: 10.1186/s12964-023-01255-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/01/2023] [Indexed: 09/20/2023] Open
Abstract
Gliomas are the most common brain tumors characterized by complicated heterogeneity. The genetic, molecular, and histological pathology of gliomas is characterized by high neuro-inflammation. The inflammatory microenvironment in the central nervous system (CNS) has been closely linked with inflammasomes that control the inflammatory response and coordinate innate host defenses. Dysregulation of the inflammasome causes an abnormal inflammatory response, leading to carcinogenesis in glioma. Because of the clinical importance of the various physiological properties of the inflammasome in glioma, the inflammasome has been suggested as a promising treatment target for glioma management. Here, we summarize the current knowledge on the contribution of the inflammasomes in glioma and therapeutic insights. Video Abstract.
Collapse
Affiliation(s)
- JeongMin Sim
- Department of Biomedical Science, College of Life Science, CHA University, Pocheon, 11160, Republic of Korea
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University College of Medicine, 59 Yatap-Ro, Bundang-Gu, Seongnam, 13496, Republic of Korea
| | - JeongMan Park
- Department of Biomedical Science, College of Life Science, CHA University, Pocheon, 11160, Republic of Korea
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University College of Medicine, 59 Yatap-Ro, Bundang-Gu, Seongnam, 13496, Republic of Korea
| | - Jong-Seok Moon
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan, 31151, Republic of Korea.
| | - Jaejoon Lim
- Department of Biomedical Science, College of Life Science, CHA University, Pocheon, 11160, Republic of Korea.
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University College of Medicine, 59 Yatap-Ro, Bundang-Gu, Seongnam, 13496, Republic of Korea.
| |
Collapse
|
27
|
Ma Y, Yu N, Lu H, Shi J, Zhang Y, Chen Z, Jia G. Titanium dioxide nanoparticles: revealing the mechanisms underlying hepatotoxicity and effects in the gut microbiota. Arch Toxicol 2023; 97:2051-2067. [PMID: 37344693 DOI: 10.1007/s00204-023-03536-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/06/2023] [Indexed: 06/23/2023]
Abstract
Numerous studies in recent years have questioned the safety of oral exposure to titanium dioxide nanoparticles (TiO2 NPs). TiO2 NPs are not only likely to accumulate in the gastrointestinal tract, but they are also found to penetrate the body circulation and reach distant organs. The liver, which is considered to be a target organ for nanoparticles, is of particular concern. TiO2 NPs accumulate in the liver and cause oxidative stress and inflammatory reactions, resulting in pathological damage. The impact of TiO2 NPs on liver aspartate aminotransferase (AST) and alanine aminotransferase (ALT) was studied using a meta-analysis. According to the findings, TiO2 NPs exposure can cause an elevation in AST and ALT levels in the blood. Furthermore, TiO2 NPs are eliminated mostly through feces, and their lengthy residence in the gut exposes them to microbiota. The gut microbiota is also dysbiotic due to titanium dioxide's antibacterial capabilities. This further leads to changes in the amount of microbiota metabolites, which can reach the liver with blood circulation and trigger hepatotoxicity through the gut-liver axis. This review examines the gut-liver axis to assess the effects of gut microbiota dysbiosis on the liver to provide suggestions for assessing the gut-hepatotoxicity of TiO2 NPs.
Collapse
Affiliation(s)
- Ying Ma
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, People's Republic of China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100191, People's Republic of China
| | - Nairui Yu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, People's Republic of China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100191, People's Republic of China
| | - Huaye Lu
- Jiangsu Prov Ctr Dis Control and Prevent, 172 Jiangsu Rd, Nanjing, 210009, People's Republic of China
| | - Jiaqi Shi
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, People's Republic of China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100191, People's Republic of China
| | - Yi Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, People's Republic of China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100191, People's Republic of China
| | - Zhangjian Chen
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, People's Republic of China.
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100191, People's Republic of China.
| | - Guang Jia
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, People's Republic of China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100191, People's Republic of China
| |
Collapse
|
28
|
Ueland T, Waagsbø B, Berge RK, Trøseid M, Aukrust P, Damås JK. Fatty Acids Composition and HIV Infection: Altered Levels of n-6 Polyunsaturated Fatty Acids Are Associated with Disease Progression. Viruses 2023; 15:1613. [PMID: 37515299 PMCID: PMC10385810 DOI: 10.3390/v15071613] [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: 05/18/2023] [Revised: 07/11/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Fatty acids (FAs) are important regulators of immune responses and innate defense mechanisms. We hypothesized that disturbed FA metabolism could contribute to the progression of HIV infection. Plasma levels of 45 FAs were analyzed with gas chromatography in healthy controls and HIV-infected patients with regard to Mycobacterium avium complex (MAC) infection. In vitro, we assessed MAC-PPD-induced release of inflammatory cytokines in peripheral and bone marrow mononuclear cells (PBMC and BMMC) according to levels of n-6 polyunsaturated fatty acids (PUFAs). While plasma saturated FAs were higher in HIV infection, PUFAs, and in particular the n-6 PUFA arachidonic acid (AA), were lower in patients with advanced disease. The ratio between AA and precursor dihomo-γ-linolenic acid, reflecting Δ5-desaturase activity, was markedly lower and inversely correlated with plasma HIV RNA levels in these patients. Depletion of AA was observed prior to MAC infection, and MAC-PPD-induced release of TNF and IL-6 in PBMC and BMMC was lower in patients with low plasma AA. Our findings suggest that dysregulated metabolism of n-6 PUFAs may play a role in the progression of HIV infection. While high AA may contribute to chronic inflammation in asymptomatic HIV-infected patients, low AA seems to increase the susceptibility to MAC infection in patients with advanced disease.
Collapse
Affiliation(s)
- Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, NO-0420 Oslo, Norway
- Faculty of Medicine, University of Oslo, NO-0318 Oslo, Norway
- Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, NO-9038 Tromsø, Norway
| | - Bjørn Waagsbø
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
- Department of Infectious Diseases, St Olav's Hospital, NO-7006 Trondheim, Norway
| | - Rolf K Berge
- Department of Clinical Science, University of Bergen, NO-5021 Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, NO-5021 Bergen, Norway
| | - Marius Trøseid
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, NO-0420 Oslo, Norway
- Faculty of Medicine, University of Oslo, NO-0318 Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, NO-0424 Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, NO-0420 Oslo, Norway
- Faculty of Medicine, University of Oslo, NO-0318 Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, NO-0424 Oslo, Norway
| | - Jan K Damås
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
- Department of Infectious Diseases, St Olav's Hospital, NO-7006 Trondheim, Norway
| |
Collapse
|
29
|
Warmink K, Rios JL, van Valkengoed DR, Vinod P, Korthagen NM, Weinans H. Effects of different obesogenic diets on joint integrity, inflammation and intermediate monocyte levels in a rat groove model of osteoarthritis. Front Physiol 2023; 14:1211972. [PMID: 37520829 PMCID: PMC10372350 DOI: 10.3389/fphys.2023.1211972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/05/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction: Obesogenic diets aggravate osteoarthritis (OA) by inducing low-grade systemic inflammation, and diet composition may affect OA severity. Here, we investigated the effect of diet on joint damage and inflammation in an OA rat model. Methods: Wistar-Han rats (n = 24) were fed a chow, a high-fat (HF) diet, or a high-fat/high-sucrose (HFS) for 24 weeks. OA was induced unilaterally 12 weeks after the diet onset by groove surgery, and compared to sham surgery or no surgical intervention (contralateral limb). Knee OA severity was determined by OARSI histopathology scoring system. At several timepoints monocyte populations were measured using flow cytometry, and joint macrophage response was determined via CD68 immunohistochemistry staining. Results: Groove surgery combined with HF or HFS diet resulted in higher OARSI scores, and both HF and HFS diet showed increased circulating intermediate monocytes compared to chow fed rats. Additionally, in the HFS group, minimal damage by sham surgery resulted in an increased OARSI score. HFS diet resulted in the largest metabolic dysregulation, synovial inflammation and increased CD68 staining in tibia epiphysis bone marrow. Conclusion: Obesogenic diets resulted in aggravated OA development, even with very minimal joint damage when combined with the sucrose/fat-rich diet. We hypothesize that diet-induced low-grade inflammation primes monocytes and macrophages in the blood, bone marrow, and synovium, resulting in joint damage when triggered by groove OA inducing surgery. When the metabolic dysregulation is larger, as observed here for the HFS diet, the surgical trigger required to induce joint damage may be smaller, or even redundant.
Collapse
Affiliation(s)
- K. Warmink
- Department of Orthopedics, University Medical Center Utrecht (UMCU), Utrecht, Netherlands
| | - J. L. Rios
- Department of Orthopedics, University Medical Center Utrecht (UMCU), Utrecht, Netherlands
| | - D. R. van Valkengoed
- Department of Orthopedics, University Medical Center Utrecht (UMCU), Utrecht, Netherlands
| | - P. Vinod
- Department of Orthopedics, University Medical Center Utrecht (UMCU), Utrecht, Netherlands
| | - N. M. Korthagen
- Department of Orthopedics, University Medical Center Utrecht (UMCU), Utrecht, Netherlands
- Department of Equine Sciences, Utrecht University, Utrecht, Netherlands
| | - H. Weinans
- Department of Orthopedics, University Medical Center Utrecht (UMCU), Utrecht, Netherlands
- Department of Biomechanical Engineering, TU Delft, Delft, Netherlands
| |
Collapse
|
30
|
Dang Y, Ma C, Chen K, Chen Y, Jiang M, Hu K, Li L, Zeng Z, Zhang H. The Effects of a High-Fat Diet on Inflammatory Bowel Disease. Biomolecules 2023; 13:905. [PMID: 37371485 PMCID: PMC10296751 DOI: 10.3390/biom13060905] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
The interactions among diet, intestinal immunity, and microbiota are complex and play contradictory roles in inflammatory bowel disease (IBD). An increasing number of studies has shed light on this field. The intestinal immune balance is disrupted by a high-fat diet (HFD) in several ways, such as impairing the intestinal barrier, influencing immune cells, and altering the gut microbiota. In contrast, a rational diet is thought to maintain intestinal immunity by regulating gut microbiota. In this review, we emphasize the crucial contributions made by an HFD to the gut immune system and microbiota.
Collapse
Affiliation(s)
- Yuan Dang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Inflammatory Bowel Disease, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chunxiang Ma
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Inflammatory Bowel Disease, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kexin Chen
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Inflammatory Bowel Disease, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yiding Chen
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mingshan Jiang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Inflammatory Bowel Disease, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kehan Hu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Inflammatory Bowel Disease, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lili Li
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Inflammatory Bowel Disease, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhen Zeng
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Inflammatory Bowel Disease, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hu Zhang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Inflammatory Bowel Disease, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
31
|
Bauer S, Hezinger L, Rexhepi F, Ramanathan S, Kufer TA. NOD-like Receptors-Emerging Links to Obesity and Associated Morbidities. Int J Mol Sci 2023; 24:ijms24108595. [PMID: 37239938 DOI: 10.3390/ijms24108595] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Obesity and its associated metabolic morbidities have been and still are on the rise, posing a major challenge to health care systems worldwide. It has become evident over the last decades that a low-grade inflammatory response, primarily proceeding from the adipose tissue (AT), essentially contributes to adiposity-associated comorbidities, most prominently insulin resistance (IR), atherosclerosis and liver diseases. In mouse models, the release of pro-inflammatory cytokines such as TNF-alpha (TNF-α) and interleukin (IL)-1β and the imprinting of immune cells to a pro-inflammatory phenotype in AT play an important role. However, the underlying genetic and molecular determinants are not yet understood in detail. Recent evidence demonstrates that nucleotide-binding and oligomerization domain (NOD)-like receptor (NLR) family proteins, a group of cytosolic pattern recognition receptors (PRR), contribute to the development and control of obesity and obesity-associated inflammatory responses. In this article, we review the current state of research on the role of NLR proteins in obesity and discuss the possible mechanisms leading to and the outcomes of NLR activation in the obesity-associated morbidities IR, type 2 diabetes mellitus (T2DM), atherosclerosis and non-alcoholic fatty liver disease (NAFLD) and discuss emerging ideas about possibilities for NLR-based therapeutic interventions of metabolic diseases.
Collapse
Affiliation(s)
- Sarah Bauer
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, 70593 Stuttgart, Germany
| | - Lucy Hezinger
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, 70593 Stuttgart, Germany
| | - Fjolla Rexhepi
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Sheela Ramanathan
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Thomas A Kufer
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, 70593 Stuttgart, Germany
| |
Collapse
|
32
|
Zhang Y, Ma XZ, Zhao XY, Li JJ, Ma S, Pang ZD, Xu J, Du XJ, Deng XL, Wang JH. AGEs-RAGE-KCa3.1 pathway mediates palmitic acid-induced migration of PBMCs from patients with type 2 diabetes. Heliyon 2023; 9:e14823. [PMID: 37025887 PMCID: PMC10070889 DOI: 10.1016/j.heliyon.2023.e14823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 03/08/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by chronic low-grade systemic inflammation. Tissue infiltration by monocyte migration contributes to the pathogenesis of vascular complications in T2DM. We studied the role of intermediate-conductance Ca2+-activated K+ (KCa3.1) channels in the palmitic acid (PA)-induced migration of peripheral blood mononuclear cells (PBMCs) from T2DM patients and the influence of advanced glycation endproducts (AGEs). A total of 49 T2DM patients and 33 healthy subjects was recruited into this study. Using flow cytometry and Western blotting analysis as well as cell migration assay, we found that there was a significant decrease in frequency of T lymphocytes and monocytes in CD45+ leukocyte population. PA at 100 μM stimulated migration of PBMCs from T2DM individuals, which was inhibited by the specific KCa3.1 channel blocker TRAM-34 (1 μM). The PBMC migration was positively correlated with glycosylated hemoglobin A1 chain (HbA1c) level of T2DM patients, an indicator of AGEs, and PBMCs with higher level of HbA1c showed upregulated expression of toll-like receptor (TLR) 2/4 and KCa3.1 channels. In THP-1 cells, AGEs at 200 μg/ml increased protein expression of TLR 2/4 and KCa3.1 channels, and were synergistically involved in PA-induced migration through receptors of AGEs (RAGE)-mediated KCa3.1 upregulation. In conclusion, in PBMCs of T2DM patients, AGEs promotes PA-induced migration via upregulation of TLR2/4 and KCa3.1 channels.
Collapse
|
33
|
Jacquet A. The HDM allergen orchestra and its cysteine protease maestro: Stimulators of kaleidoscopic innate immune responses. Mol Immunol 2023; 156:48-60. [PMID: 36889186 DOI: 10.1016/j.molimm.2023.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/29/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023]
Abstract
House dust mite (HDM) encloses an explosive cocktail of allergenic proteins sensitizing hundreds of millions of people worldwide. To date, the innate cellular and molecular mechanism(s) orchestrating the HDM-induced allergic inflammation remains partially deciphered. Understanding the kaleidoscope of HDM-induced innate immune responses is hampered by (1) the large complexity of the HDM allergome with very diverse functional bioreactivities, (2) the perpetual presence of microbial compounds (at least LPS, β-glucan, chitin) promoting as well pro-Th2 innate signaling pathways and (3) multiple cross-talks involving structural, neuronal and immune cells. The present review provides an update on the innate immune properties, identified so far, of multiple HDM allergen groups. Experimental evidence highlights the importance of HDM allergens displaying protease or lipid-binding activities on the initiation of the allergic responses. Specifically, group 1 HDM cysteine proteases are considered as the key initiators of the allergic response through their capacities to impair the epithelial barrier integrity, to stimulate the release of pro-Th2 danger-associated molecular patterns (DAMPs) in epithelial cells, to produce super-active forms of IL-33 alarmin and to mature thrombin leading to Toll-like receptor 4 (TLR4) activation. Remarkably, the recently evidenced primary sensing of cysteine protease allergens by nociceptive neurons confirms the critical role of this HDM allergen group in the early events leading to Th2 differentiation.
Collapse
Affiliation(s)
- Alain Jacquet
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
34
|
Sinha RA. Autophagy: A Cellular Guardian against Hepatic Lipotoxicity. Genes (Basel) 2023; 14:553. [PMID: 36874473 PMCID: PMC7614268 DOI: 10.3390/genes14030553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Lipotoxicity is a phenomenon of lipid-induced cellular injury in nonadipose tissue. Excess of free saturated fatty acids (SFAs) contributes to hepatic injury in nonalcoholic fatty liver disease (NAFLD), which has been growing at an unprecedented rate in recent years. SFAs and their derivatives such as ceramides and membrane phospholipids have been shown to induce intrahepatic oxidative damage and ER stress. Autophagy represents a cellular housekeeping mechanism to counter the perturbation in organelle function and activation of stress signals within the cell. Several aspects of autophagy, including lipid droplet assembly, lipophagy, mitophagy, redox signaling and ER-phagy, play a critical role in mounting a strong defense against lipotoxic lipid species within the hepatic cells. This review provides a succinct overview of our current understanding of autophagy-lipotoxicity interaction and its pharmacological and nonpharmacological modulation in treating NAFLD.
Collapse
Affiliation(s)
- Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| |
Collapse
|
35
|
Finney AC, Das S, Kumar D, McKinney MP, Cai B, Yurdagul A, Rom O. The interplay between nonalcoholic fatty liver disease and atherosclerotic cardiovascular disease. Front Cardiovasc Med 2023; 10:1116861. [PMID: 37200978 PMCID: PMC10185914 DOI: 10.3389/fcvm.2023.1116861] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/23/2023] [Indexed: 05/20/2023] Open
Abstract
Therapeutic approaches that lower circulating low-density lipoprotein (LDL)-cholesterol significantly reduced the burden of cardiovascular disease over the last decades. However, the persistent rise in the obesity epidemic is beginning to reverse this decline. Alongside obesity, the incidence of nonalcoholic fatty liver disease (NAFLD) has substantially increased in the last three decades. Currently, approximately one third of world population is affected by NAFLD. Notably, the presence of NAFLD and particularly its more severe form, nonalcoholic steatohepatitis (NASH), serves as an independent risk factor for atherosclerotic cardiovascular disease (ASCVD), thus, raising interest in the relationship between these two diseases. Importantly, ASCVD is the major cause of death in patients with NASH independent of traditional risk factors. Nevertheless, the pathophysiology linking NAFLD/NASH with ASCVD remains poorly understood. While dyslipidemia is a common risk factor underlying both diseases, therapies that lower circulating LDL-cholesterol are largely ineffective against NASH. While there are no approved pharmacological therapies for NASH, some of the most advanced drug candidates exacerbate atherogenic dyslipidemia, raising concerns regarding their adverse cardiovascular consequences. In this review, we address current gaps in our understanding of the mechanisms linking NAFLD/NASH and ASCVD, explore strategies to simultaneously model these diseases, evaluate emerging biomarkers that may be useful to diagnose the presence of both diseases, and discuss investigational approaches and ongoing clinical trials that potentially target both diseases.
Collapse
Affiliation(s)
- Alexandra C. Finney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Sandeep Das
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Dhananjay Kumar
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - M. Peyton McKinney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, NY, United States
| | - Arif Yurdagul
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Correspondence: Arif Yurdagul Oren Rom
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Correspondence: Arif Yurdagul Oren Rom
| |
Collapse
|
36
|
Zhu H, Wang H, Cheng Y, Liu D, Zhang A, Wen Z, Gao J. Hadh deficiency induced oligoasthenoteratozoospermia through the TNF-α/Bcl-2 pathway in male mice. FASEB J 2022; 36:e22661. [PMID: 36398584 DOI: 10.1096/fj.202201144r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/11/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022]
Abstract
The process of spermatogenesis is a complex and delicate process that is still not fully understood. In this study, we examined the role of fatty acid oxidase 3-hydroxy acyl CoA dehydrogenase (HADH) in maintaining normal spermatogenesis in mice. In male mice, ablation of the Hadh gene using CRISPR/Cas9 technology arrested spermatocyte meiosis, increased multinucleated giant germ cells and vacuoles in seminiferous tubules, and accompanied with acrosomal dysplasia. Hadh-/- male mice showed the typical features of oligoasthenoteratozoospermia (OAT), including decreased sperm concentration and motility and increased sperm abnormalities. Next, we explored the molecular events in the testes of the mutant mice. We found fatty acids accumulated in the testis of Hadh-/- mice. And also, inflammatory factors TNF-α, IL-1β, and IL-6 were significantly increased, apoptosis-related protein Bcl-2 was decreased, and Bax and cleaved-Caspase3 were increased in Hadh-/- male mice testis. After using etanercept, a specific inhibitor of TNF-α, testis injury caused by Hadh knockout was significantly alleviated, the sperm quality and motility were improved, and germ cell apoptosis was reduced. So our study demonstrated that Hadh deletion caused an increase in fatty acids. The accumulated fatty acids further induced testicular inflammation and germ cell apoptosis through the TNF-α/Bcl-2 signaling pathway, finally resulting in OAT in the Hadh-/- mice. Inhibiting TNF-α may be used as a new treatment approach for testicular inflammation and OAT.
Collapse
Affiliation(s)
- Haixia Zhu
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Hongxiang Wang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Yin Cheng
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Dongyue Liu
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Aizhen Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Zongzhuang Wen
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Jiangang Gao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China.,Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| |
Collapse
|
37
|
Peng L, Xiang L, Xu Z, Gu H, Zhu Z, Tang Y, Jiang Y, He H, Wang Y, Zhao X. Association between low-fat diet and liver cancer risk in 98,455 participants: Results from a prospective study. Front Nutr 2022; 9:1013643. [PMID: 36466389 PMCID: PMC9716652 DOI: 10.3389/fnut.2022.1013643] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/26/2022] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Low-fat diet reduces the risk of chronic metabolic diseases such as obesity and diabetes, which exhibit overlapping mechanisms with liver cancer. However, the association between low-fat diet and liver cancer risk remains unclear. AIM To investigate whether adherence to low-fat diet is associated with a reduced risk of liver cancer in a prospective study. MATERIALS AND METHODS Data of participants in this study were collected from the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial. A low-fat diet score was calculated to reflect adherence to low-fat dietary pattern, with higher scores indicating greater adherence. Cox regression was used to calculate hazard ratios (HRs) and 95% confidence intervals (CIs) for liver cancer incidence with adjustment for potential covariates. Restricted cubic spline model was used to characterize liver cancer risk across the full range of the low-fat diet score. Prespecified subgroup analyses were used to identify potential impact modifiers. Sensitivity analyses were performed to test the robustness of this association. RESULTS A total of 98,455 participants were included in the present analysis. The mean (standard deviation) age, low-fat diet score, and follow-up time were 65.52 (5.73) years, 14.99 (6.27) points, and 8.86 (1.90) years, respectively. During 872639.5 person-years of follow-up, 91 liver cancers occurred, with an overall incidence rate of 0.01 cases per 100 person-years. In the fully adjusted Cox model, the highest versus the lowest quartile of low-fat diet score was found to be associated with a reduced risk of liver cancer (HR Q4 vs. Q1: 0.458; 95% CI: 0.218, 0.964; P = 0.035 for trend), which remained associated through a series of sensitivity analyses. The restricted cubic spline model showed a linear dose-response association between low-fat diet score and liver cancer incidence (p = 0.482 for non-linear). Subgroup analyses did not show significant interaction between low-fat diet score and potential impact modifiers in the incidence of liver cancer. CONCLUSION In this study, low-fat diet score is associated with reduced liver cancer risk in the US population, indicating that adherence to low-fat diet may be helpful for liver cancer prevention. Future studies should validate our findings in other populations.
Collapse
Affiliation(s)
- Linglong Peng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Xiang
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiquan Xu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haitao Gu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiyong Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunhao Tang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yahui Jiang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongmei He
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yaxu Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaodong Zhao
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
38
|
Luo J, Chen H, Zhang Q, Huang X, Qin X, Li J, Chen S, Xiao Y, Sun L, Sun B. Metabolism Characteristics of Mycoplasma pneumoniae Infection in Asthmatic Children. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2022; 14:713-729. [PMID: 36426399 PMCID: PMC9709688 DOI: 10.4168/aair.2022.14.6.713] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 08/03/2022] [Accepted: 08/09/2022] [Indexed: 07/30/2023]
Abstract
PURPOSE Studies have shown that Mycoplasma pneumoniae (Mp) infection can aggravate symptoms in asthmatics. However, the mechanism by which Mp infection exacerbates asthma remains unclear. Metabolomics can help identify the mechanism of Mp aggravating asthma in children, thereby providing more a potential target for improving clinical treatment programs. In this article, we analyzed the metabolic level of patients to explain how Mp aggravates asthma in children. METHODS We divided the subjects into the asthma, Mp infection, asthma combined with Mp infection and healthy groups. Patients' peripheral blood was collected for metabolic and interaction analysis. Cytokine levels were measured via serum and exhaled breath condensate (EBC). RESULTS A total of 150 participating subjects were divided into four groups after exclusion. We found out that there were different metabolic pathways between the healthy and disease groups. The major pathways of both asthma and asthma combined with Mp infection were valine, leucine and isoleucine biosynthesis; malate-aspartate shuttle was the main differential pathway for Mp infection. Moreover, even though three disease groups involved 81 metabolites at the same time, compared with asthma combined with Mp infection, 2 single disease groups still involved different amino acid pathways (phenylalanine, tyrosine and tryptophan biosynthesis; valine, leucine and isoleucine biosynthesis). Interaction analysis showed that Mp infection in asthmatic patients not only activated cytokines, but also activated Toll-like receptors (TLRs) 2 and 6. Finally, the levels of interleukin (IL)-4, IL-8, IL-13 and tumor necrosis factor-α in EBC with asthma combined with Mp infection were significantly higher than the 2 single disease groups. CONCLUSIONS Mp infection in asthmatic children can cause changes in the levels of various amino acids in the body, which were enriched in the pathways such as valine, leucine and isoleucine biosynthesis. Palmitic acid can activate TLR2, and iloprost reduces IL-10 levels, ultimately leading to the increased airway inflammation.
Collapse
Affiliation(s)
- Jiaying Luo
- State Key Lab of Respiratory Disease, National Clinical Research Center of Respiratory Disease, and Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huian Chen
- State Key Lab of Respiratory Disease, National Clinical Research Center of Respiratory Disease, and Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qiyong Zhang
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xinyun Huang
- State Key Lab of Respiratory Disease, National Clinical Research Center of Respiratory Disease, and Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xu Qin
- State Key Lab of Respiratory Disease, National Clinical Research Center of Respiratory Disease, and Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Li
- Institute of Integrated Traditional Chinese and Western Medicine, Guangzhou Medical University, Guangzhou, China
| | - Siyi Chen
- State Key Lab of Respiratory Disease, National Clinical Research Center of Respiratory Disease, and Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongxin Xiao
- State Key Lab of Respiratory Disease, National Clinical Research Center of Respiratory Disease, and Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lihong Sun
- State Key Lab of Respiratory Disease, National Clinical Research Center of Respiratory Disease, and Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Baoqing Sun
- State Key Lab of Respiratory Disease, National Clinical Research Center of Respiratory Disease, and Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
39
|
Nishimoto S, Sata M, Fukuda D. Expanding role of deoxyribonucleic acid-sensing mechanism in the development of lifestyle-related diseases. Front Cardiovasc Med 2022; 9:881181. [PMID: 36176986 PMCID: PMC9513035 DOI: 10.3389/fcvm.2022.881181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/15/2022] [Indexed: 11/14/2022] Open
Abstract
In lifestyle-related diseases, such as cardiovascular, metabolic, respiratory, and kidney diseases, chronic inflammation plays a causal role in their pathogenesis; however, underlying mechanisms of sterile chronic inflammation are not well-understood. Previous studies have confirmed the damage of cells in these organs in the presence of various risk factors such as diabetes, dyslipidemia, and cigarette smoking, releasing various endogenous ligands for pattern recognition receptors. These studies suggested that nucleic acids released from damaged tissues accumulate in these tissues, acting as an endogenous ligand. Undamaged DNA is an integral factor for the sustenance of life, whereas, DNA fragments, especially those from pathogens, are potent activators of the inflammatory response. Recent studies have indicated that inflammatory responses such as the production of type I interferon (IFN) induced by DNA-sensing mechanisms which contributes to self-defense system in innate immunity participates in the progression of inflammatory diseases by the recognition of nucleic acids derived from the host, including mitochondrial DNA (mtDNA). The body possesses several types of DNA sensors. Toll-like receptor 9 (TLR9) recognizes DNA fragments in the endosomes. In addition, the binding of DNA fragments in the cytosol activates cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) synthase (cGAS), resulting in the synthesis of the second messenger cyclic GMP-AMP (cGAMP). The binding of cGAMP to stimulator of interferon genes (STING) activates NF-κB and TBK-1 signaling and consequently the production of many inflammatory cytokines including IFNs. Numerous previous studies have demonstrated the role of DNA sensors in self-defense through the recognition of DNA fragments derived from pathogens. Beyond the canonical role of TLR9 and cGAS-STING, this review describes the role of these DNA-sensing mechanism in the inflammatory responses caused by endogenous DNA fragments, and in the pathogenesis of lifestyle-related diseases.
Collapse
Affiliation(s)
- Sachiko Nishimoto
- Faculty of Clinical Nutrition and Dietetics, Konan Women’s University, Kobe, Japan
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Daiju Fukuda
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
- Department of Cardiovascular Medicine, Osaka Metropolitan University, Osaka, Japan
- *Correspondence: Daiju Fukuda, ,
| |
Collapse
|
40
|
Abhishek A, Fuller A, Nakafero G, Zhang W, Dumbleton J, Hawkey C, Coupland C, Terkeltaub R, Doherty M. Feasibility of conducting a randomized, placebo-controlled study assessing whether omega-3 fatty acids prevent gout flares when starting urate-lowering treatment. Rheumatol Adv Pract 2022; 6:rkac086. [DOI: 10.1093/rap/rkac086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
Abstract
Objective
The aim was to test the feasibility of a randomized controlled trial exploring whether omega-3 fatty acid supplementation limits gout flares during treat-to-target urate-lowering treatment (T2T-ULT).
Methods
Adults with at least one gout flare in the past 12 months and serum urate (SU) ≥360 μmol/l were recruited from general practices (primary method) and randomly assigned 1:1 to receive omega-3 fatty acid supplementation (4 g/day) or placebo for 28 weeks. At week 5, participants began T2T-ULT. The primary outcome was drop-out rate. Secondary outcomes were recruitment rate, outcome data completeness, the number, severity and duration of gout flares between weeks 5 and 28, and study drug compliance.
Results
Ninety-five per cent of randomized participants (n = 60) completed all study visits. The primary method recruitment rate was 2.2%. Fifty and 42 participants achieved SU < 360 and 300 μmol/l (6 and 5 mg/dl), respectively. The number of gout flares [median (interquartile range): active 1 (0–2) and placebo 1 (0–2)], flare duration [mean (s.d.): active 7.00 (4.52) days and placebo 7.06 (8.14) days] and time to first flare [hazard ratio (95% CI) 0.97 (0.50, 1.86)] were comparable between both arms. Study drug compliance was high and comparable in both arms [median (interquartile range) returned capsule count: active 57 (26–100) and placebo 58 (27–154)]; red blood cell omega-3 fatty acid index increased twofold in the active arm and remained unchanged in the control arm.
Conclusion
The study demonstrated feasibility and provided useful metrics for conducting a community-based gout flare prophylaxis trial.
Study registration
ISRCTN; https://www.isrctn.com/; ISRCTN79392964.
Collapse
Affiliation(s)
- Abhishek Abhishek
- Academic Rheumatology, The University of Nottingham , Nottingham, UK
| | - Amy Fuller
- Academic Rheumatology, The University of Nottingham , Nottingham, UK
| | - Georgina Nakafero
- Academic Rheumatology, The University of Nottingham , Nottingham, UK
| | - Weiya Zhang
- Academic Rheumatology, The University of Nottingham , Nottingham, UK
| | - Jennifer Dumbleton
- Nottingham Digestive Diseases Centre, The University of Nottingham , Nottingham, UK
| | - Christopher Hawkey
- Nottingham Digestive Diseases Centre, The University of Nottingham , Nottingham, UK
| | - Carol Coupland
- Division of Primary Care, The University of Nottingham , Nottingham, UK
| | - Robert Terkeltaub
- Veterans Affairs San Diego Healthcare System , San Diego, CA, USA
- Department of Medicine, University of California , San Diego, CA, USA
| | - Michael Doherty
- Academic Rheumatology, The University of Nottingham , Nottingham, UK
| |
Collapse
|
41
|
Zhu S, Liu Q, Xiang X, Cui K, Zhao F, Mai K, Ai Q. Docosahexaenoic Acid Ameliorates the Toll-Like Receptor 22-Triggered Inflammation in Fish by Disrupting Lipid Raft Formation. J Nutr 2022; 152:1991-2002. [PMID: 35679100 DOI: 10.1093/jn/nxac125] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/11/2022] [Accepted: 06/13/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Although dietary DHA alleviates Toll-like receptor (TLR)-associated chronic inflammation in fish, the underlying mechanism is not well understood. OBJECTIVES This study aimed to explore the role of Tlr22 in the innate immunity of large yellow croaker and investigate the anti-inflammatory effects of DHA on Tlr22-triggered inflammation. METHODS Head kidney-derived macrophages of croaker and HEK293T cells were or were not pretreated with 100 μM DHA for 10 h prior to polyinosinic-polycytidylic acid (poly I:C) stimulation. We executed qRT-PCR, immunoblotting, and lipidomic analysis to examine the impact of DHA on Tlr22-triggered inflammation and membrane lipid composition. In vivo, croakers (12.03 ± 0.05 g) were fed diets containing 0.2% [control (Ctrl)], 0.8%, and 1.6% DHA for 8 wk before injection with poly I:C. Inflammatory genes expression and rafts-related lipids and protein expression were measured in the head kidney. Data were analyzed by ANOVA or Student t test. RESULTS The activation of Tlr22 by poly I:C induced inflammation, and DHA diminished Tlr22-targeted inflammatory gene expression by 56-73% (P ≤ 0.05). DHA reduced membrane sphingomyelin (SM) and SFA-containing phosphatidylcholine (SFA-PC) contents, as well as lipid raft marker caveolin 1 amounts. Furthermore, lipid raft disruption suppressed Tlr22-induced Nf-κb and interferon h activation and p65 nuclear translocation. In vivo, expression of Tlr22 target inflammatory genes was 32-64% lower in the 1.6% DHA group than in the Ctrl group upon poly I:C injection (P ≤ 0.05). Also, the 1.6% DHA group showed a reduction in membrane SM and SFA-PC contents, accompanied by a decrease in caveolin 1 amounts, compared with the Ctrl group. CONCLUSIONS The activation of Tlr22 signaling depends on lipid rafts, and DHA ameliorates the Tlr22-triggered inflammation in both head kidney and head kidney-derived macrophages of croaker partially by altering membrane SMs and SFA-PCs that are required for lipid raft organization.
Collapse
Affiliation(s)
- Si Zhu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China.,Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Qiangde Liu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Xiaojun Xiang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Kun Cui
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Fang Zhao
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong, China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong, China
| |
Collapse
|
42
|
Acne, Microbiome, and Probiotics: The Gut–Skin Axis. Microorganisms 2022; 10:microorganisms10071303. [PMID: 35889022 PMCID: PMC9318165 DOI: 10.3390/microorganisms10071303] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/20/2022] [Accepted: 06/25/2022] [Indexed: 11/17/2022] Open
Abstract
The objective of this narrative review was to check the influence of the human microbiota in the pathogenesis of acne and how the treatment with probiotics as adjuvant or alternative therapy affects the evolution of acne vulgaris. Acne is a chronic inflammatory skin disease involving the pilosebaceous units. The pathogenesis of acne is complex and multifactorial involving genetic, metabolic, and hormonal factors in which both skin and gut microbiota are implicated. Numerous studies have shown the bidirectionality between the intestinal microbiota and skin homeostasis, a communication mainly established by modifying the immune system. Increased data on the mechanisms of action regarding the relevance of Cutibacterium acnes, as well as the importance of the gut–skin axis, are becoming known. Diverse and varied in vitro studies have shown the potential beneficial effects of probiotics in this context. Clinical trials with both topical and oral probiotics are scarce, although they have shown positive results, especially with oral probiotics through the modulation of the intestinal microbiota, generating an anti-inflammatory response and restoring intestinal integrity, or through metabolic pathways involving insulin-like growth factor I (IGF-1). Given the aggressiveness of some standard acne treatments, probiotics should continue to be investigated as an alternative or adjuvant therapy.
Collapse
|
43
|
Conway J, Certo M, Lord JM, Mauro C, Duggal NA. Understanding the role of host metabolites in the induction of immune senescence: Future strategies for keeping the ageing population healthy. Br J Pharmacol 2022; 179:1808-1824. [PMID: 34435354 DOI: 10.1111/bph.15671] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/12/2021] [Accepted: 08/19/2021] [Indexed: 12/24/2022] Open
Abstract
Advancing age is accompanied by significant remodelling of the immune system, termed immune senescence, and increased systemic inflammation, termed inflammageing, both of which contribute towards an increased risk of developing chronic diseases in old age. Age-associated alterations in metabolic homeostasis have been linked with changes in a range of physiological functions, but their effects on immune senescence remains poorly understood. In this article, we review the recent literature to formulate hypotheses as to how an age-associated dysfunctional metabolism, driven by an accumulation of key host metabolites (saturated fatty acids, cholesterol, ceramides and lactate) and loss of other metabolites (glutamine, tryptophan and short-chain fatty acids), might play a role in driving immune senescence and inflammageing, ultimately leading to diseases of old age. We also highlight the potential use of metabolic immunotherapeutic strategies targeting these processes in counteracting immune senescence and restoring immune homeostasis in older adults. LINKED ARTICLES: This article is part of a themed issue on Inflammation, Repair and Ageing. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.9/issuetoc.
Collapse
Affiliation(s)
- Jessica Conway
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| | - Michelangelo Certo
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Janet M Lord
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham and University of Birmingham, Birmingham, UK
| | - Claudio Mauro
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| | - Niharika A Duggal
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
44
|
Jiang M, Fan X, Jiang Z, Chen H, Liu Y, Yu T, Huang Q, Ma Y. Comparative Proteomic Analysis of Membrane Vesicles from Clinical C. acnes Isolates with Differential Antibiotic Resistance. CLINICAL, COSMETIC AND INVESTIGATIONAL DERMATOLOGY 2022; 15:703-712. [PMID: 35463830 PMCID: PMC9022742 DOI: 10.2147/ccid.s363537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/12/2022] [Indexed: 12/13/2022]
Abstract
Purpose Cutibacterium acnes (C. acnes) is closely associated with the pathogenesis of acne, and antibiotics targeting C. acnes have been widely used for decades. However, antibiotic resistance has been increasing rapidly. Membrane vesicles (MVs) have been found to play important roles in antibiotic resistance in some bacteria. We aimed to explore the mechanism of antibiotic resistance and the virulence components within C. acnes-derived MVs. Materials and Methods We isolated clinical C. acnes strains from the lesions of acne patients who were sensitive or resistant to the antibiotics erythromycin and clindamycin. We analyzed the proteome of MVs from four sensitive C. acnes isolates and three resistant isolates by LC-MS/MS. Results We identified 543 proteins within the MVs of clinical C. acnes strains. Several lipases, NlpC/P60, CAMP factor, and Hta domain protein were detected as virulence factors in the C. acnes-derived MVs. The levels of two lipases and FtsZ were significantly higher in resistant C. acnes-derived MVs compared with sensitive strains (p < 0.05). Conclusion According to the implications of this study, improper antibiotic use might not only increase antibiotic resistance in C. acnes but could also further alter the cutaneous lipid composition and aggravate host inflammation, thus resulting in worse clinical manifestations in acne patients. This study re-emphasizes that the improper use of antibiotics should be treated more seriously in clinical practice. Furthermore, to combat multidrug resistance in C. acnes, this study suggests that FtsZ inhibitors could be useful.
Collapse
Affiliation(s)
- Min Jiang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Xiaoyao Fan
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Ziqi Jiang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Huyan Chen
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Ye Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Tianze Yu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Qiong Huang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Ying Ma
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| |
Collapse
|
45
|
Divella R, Gadaleta Caldarola G, Mazzocca A. Chronic Inflammation in Obesity and Cancer Cachexia. J Clin Med 2022; 11:2191. [PMID: 35456284 PMCID: PMC9027625 DOI: 10.3390/jcm11082191] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 12/14/2022] Open
Abstract
Chronic inflammation has long been linked to obesity and related conditions such as type 2 diabetes and metabolic syndrome. According to current research, the increased risk of cancer in people with certain metabolic diseases may be due to chronic inflammation. Adipocytokines, which are pro-inflammatory cytokines secreted in excess, are elevated in many chronic metabolic diseases. Cytokines and inflammatory mediators, which are not directly linked to DNA, are important in tumorigenesis. Cachexia, a type of metabolic syndrome linked to the disease, is associated with a dysregulation of metabolic pathways. Obesity and cachexia have distinct metabolic characteristics, such as insulin resistance, increased lipolysis, elevated free fatty acids (FFA), and ceramide levels, which are discussed in this section. The goal of this research project is to create a framework for bringing together our knowledge of inflammation-mediated insulin resistance.
Collapse
Affiliation(s)
- Rosa Divella
- ASD Nordic Walking Apulia Lifestyle, Corso Giuseppe Di Vittorio 14, 70024 Gravina in Puglia, Italy
| | | | - Antonio Mazzocca
- Interdisciplinary Department of Medicine, University of Bari School of Medicine, Piazza G. Cesare, 11, 70124 Bari, Italy
| |
Collapse
|
46
|
Shayo SC, Ogiso K, Kawade S, Hashiguchi H, Deguchi T, Nishio Y. Dietary obesity and glycemic excursions cause a parallel increase in STEAP4 and pro-inflammatory gene expression in murine PBMCs. Diabetol Int 2022; 13:358-371. [PMID: 35463853 PMCID: PMC8980188 DOI: 10.1007/s13340-021-00542-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/05/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND The balance between pro-atherogenic and anti-atherogenic factors is very crucial in the development of atherosclerotic lesions. Although the expression of the six-transmembrane epithelial antigen of the prostate 4 (STEAP4) in myeloid cells is known to be atheroprotective, there is not a single study reporting on the status of STEAP4 expression in circulating monocytes in the early stages of diet-induced obesity or in events of glycemic excursions. METHODS We induced glycemic spikes twice daily for a 1-week duration to rats fed on regular chow and western diet, and analyzed gene expression changes in the peripheral blood mononuclear cells (PBMCs). We also conducted experiments on RAW 264.7 cells to gain insight into some of our in vivo findings. RESULTS Diet-induced obesity and glycemic excursions independently caused a significant increase in STEAP4 mRNA expression in PBMCs. This was also accompanied by an induction of a substantial number of pro-inflammatory cytokines, chemokines, and chemokine receptors. However, the combined effect of western diet and hyperglycemic spikes was subtle and non-additive. In the in vitro setting, either glucose spikes, persistent hyperglycemia, or a combination of palmitic acid and insulin resulted in a parallel increase in expression of STEAP4 and pro-inflammatory genes. This was, however, significantly abrogated with 4-octyl itaconate or attenuated by inhibitors of p38MAPK and NF-kB. CONCLUSIONS STEAP4 expression in mononuclear cells is induced by increasing inflammation or oxidative stress. The observed increase in STEAP4 expression in circulating monocytes due to visceral obesity or glycemic excursions is a compensatory response. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s13340-021-00542-1.
Collapse
Affiliation(s)
- Sigfrid Casmir Shayo
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Science, 8‑35‑1 Sakuragaoka, Kagoshima, 890‑8520 Japan
- Muhimbili University of Health and Allied Sciences, P.O.BOX 65001, Dar es Salaam, Tanzania
| | - Kazuma Ogiso
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Science, 8‑35‑1 Sakuragaoka, Kagoshima, 890‑8520 Japan
| | - Shigeru Kawade
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Science, 8‑35‑1 Sakuragaoka, Kagoshima, 890‑8520 Japan
| | - Hiroshi Hashiguchi
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Science, 8‑35‑1 Sakuragaoka, Kagoshima, 890‑8520 Japan
| | - Takahisa Deguchi
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Science, 8‑35‑1 Sakuragaoka, Kagoshima, 890‑8520 Japan
| | - Yoshihiko Nishio
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Science, 8‑35‑1 Sakuragaoka, Kagoshima, 890‑8520 Japan
| |
Collapse
|
47
|
Hu C, Lin Z, Liu Z, Tang X, Song J, Lin J, Chen Y, Hu Z. Dietary fatty acid patterns and risk of oesophageal squamous cell carcinoma. PeerJ 2022; 10:e13036. [PMID: 35382006 PMCID: PMC8977065 DOI: 10.7717/peerj.13036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 02/09/2022] [Indexed: 01/11/2023] Open
Abstract
Background To characterize and examine the associations between dietary fatty acid intake patterns and the risk of oesophageal squamous cell carcinoma (ESCC). Methods A total of 422 patients and 423 controls were recruited. Dietary fatty acids were entered into a factor analysis. Multivariable logistic regression and restricted cubic spline were used to evaluate the risk of ESCC specific for different dietary fatty acid patterns (FAPs). A forest plot was applied to show the association between FAPs and ESCC risk after stratification by lifestyle exposure factors (tobacco smoking, alcohol drinking, pickled food, fried food, hot food, hard food). Results The factor analysis generated four major fatty acid patterns: a medium- and long-chain SFA (MLC-SFA) pattern; an even-chain unsaturated fatty acid (EC-UFA) pattern, a saturated fatty acid (SFA) pattern and an n-3 long-chain polyunsaturated fatty acid (n-3 LC-PUFA) pattern. In the multivariate-adjusted model, the odds ratios (ORs) with 95% confidence intervals (CIs) of ESCC were 2.07 (1.31, 3.26) and 0.53 (0.34, 0.81) for the highest versus the lowest tertiles of the EC-UFA pattern and n-3 LC-PUFA pattern, respectively. The MLC-SFA and SFA patterns were not associated with ESCC. An association between FAPs and ESCC risk after stratification by lifestyle exposure factors was also observed. Conclusions Our study indicates that the EC-UFA pattern and n-3 LC-PUFA pattern intake are associated with ESCC, providing a potential dietary intervention for ESCC prevention.
Collapse
Affiliation(s)
- Chanchan Hu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Zheng Lin
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhiqiang Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Xuwei Tang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Jianyu Song
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Jianbo Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Fujian Medical University, FuZhou, Fujian, China
| | - Yuanmei Chen
- Department of Thoracic Surgery, Fujian Medical University Cancer Hospital, FuZhou, Fujian, China
| | - Zhijian Hu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, FuZhou, Fujian, China
| |
Collapse
|
48
|
Lipke K, Kubis-Kubiak A, Piwowar A. Molecular Mechanism of Lipotoxicity as an Interesting Aspect in the Development of Pathological States-Current View of Knowledge. Cells 2022; 11:cells11050844. [PMID: 35269467 PMCID: PMC8909283 DOI: 10.3390/cells11050844] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
Free fatty acids (FFAs) play numerous vital roles in the organism, such as contribution to energy generation and reserve, serving as an essential component of the cell membrane, or as ligands for nuclear receptors. However, the disturbance in fatty acid homeostasis, such as inefficient metabolism or intensified release from the site of storage, may result in increased serum FFA levels and eventually result in ectopic fat deposition, which is unfavorable for the organism. The cells are adjusted for the accumulation of FFA to a limited extent and so prolonged exposure to elevated FFA levels results in deleterious effects referred to as lipotoxicity. Lipotoxicity contributes to the development of diseases such as insulin resistance, diabetes, cardiovascular diseases, metabolic syndrome, and inflammation. The nonobvious organs recognized as the main lipotoxic goal of action are the pancreas, liver, skeletal muscles, cardiac muscle, and kidneys. However, lipotoxic effects to a significant extent are not organ-specific but affect fundamental cellular processes occurring in most cells. Therefore, the wider perception of cellular lipotoxic mechanisms and their interrelation may be beneficial for a better understanding of various diseases’ pathogenesis and seeking new pharmacological treatment approaches.
Collapse
|
49
|
Williams EJ, Guilleminault L, Berthon BS, Eslick S, Wright T, Karihaloo C, Gately M, Baines KJ, Wood LG. Sulforaphane Reduces Pro-Inflammatory Response To Palmitic Acid In Monocytes And Adipose Tissue Macrophages. J Nutr Biochem 2022; 104:108978. [PMID: 35271969 DOI: 10.1016/j.jnutbio.2022.108978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/03/2021] [Accepted: 02/09/2022] [Indexed: 10/18/2022]
|
50
|
Quintanilha BJ, Chaves DF, Brasili E, Corrêa TA, Capetini VC, Ferreira FM, Castro IA, Hassimotto NM, Rogero MM, Lajolo FM. Ingestion of orange juice prevents hyperglycemia and increases plasma miR-375 expression. Clin Nutr ESPEN 2022; 47:240-245. [DOI: 10.1016/j.clnesp.2021.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/23/2021] [Accepted: 12/01/2021] [Indexed: 10/19/2022]
|