101
|
Rodrigues MOM, Evangelista-Silva PH, Neves NN, Moreno LG, Santos CS, Rocha KLS, Ottone VO, Batista-da-Silva B, Dias-Peixoto MF, Magalhães FC, Esteves EA. Caloric restriction-induced weight loss with a high-fat diet does not fully recover visceral adipose tissue inflammation in previously obese C57BL/6 mice. Appl Physiol Nutr Metab 2020; 45:1353-1359. [PMID: 32574503 DOI: 10.1139/apnm-2020-0220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Caloric restriction (CR) reduces body weight and systemic inflammation, but the effects on adipose tissue under dietary lipid overload are controversial. We evaluated the effects of CR-induced weight loss with a high-fat diet on adipose tissue inflammation of obese mice. Male mice were assigned into low-fat diet (LF) and high-fat diet (HF) groups. After 8 weeks, the mice in the HF group were reassigned for another 7 weeks into the following 3 conditions: (i) kept in the HF condition; (ii) changed to low-fat diet ad libitum (LFAL); and (iii) changed to high-fat calorie-restricted (RHF) diet to reach LFAL body weight. Serum markers, adipocytokines, morphology, and inflammatory infiltrates in retroperitoneal adipose tissue (RAT) were accessed. The body weights of the LFAL and RHF groups were reduced, equaling the body weights of the LF group. The LFAL mice had restored almost all inflammatory markers as the LF mice, except tumor necrosis factor-alpha (TNF-α), monocyte chemoattractant protein-1 (MCP-1), and adiponectin. Compared with the HF group, the RHF group had lowered visceral adiposity, retroperitoneal adipocyte sizes, and RAT inflammatory cell infiltration, as well as TNF-α, interleukin-6, and hepatic and serum C-reactive protein, which were higher than that of the LFAL group; adiponectin and MCP-1 did not change. CR with high-fat diet reduced body weight and attenuated visceral adiposity but did not fully recover visceral tissue inflammation. Novelty Caloric restriction in a high-fat diet ameliorated visceral adiposity. Caloric restriction in a high-fat diet did not recover visceral adipose tissue inflammation.
Collapse
Affiliation(s)
- M O M Rodrigues
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, MG 39100-000, Brazil
| | - P H Evangelista-Silva
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, MG 39100-000, Brazil
| | - N N Neves
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, MG 39100-000, Brazil
| | - L G Moreno
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, MG 39100-000, Brazil
| | - C S Santos
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, MG 39100-000, Brazil
| | - K L S Rocha
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, MG 39100-000, Brazil
| | - V O Ottone
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, MG 39100-000, Brazil
| | - B Batista-da-Silva
- Instituto de Biotecnologia, Universidade Federal de Uberlândia, Uberlândia, MG 38408-100, Brazil
| | - M F Dias-Peixoto
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, MG 39100-000, Brazil
| | - F C Magalhães
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, MG 39100-000, Brazil
| | - E A Esteves
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, MG 39100-000, Brazil
| |
Collapse
|
102
|
Lechner K, McKenzie AL, Kränkel N, Von Schacky C, Worm N, Nixdorff U, Lechner B, Scherr J, Weingärtner O, Krauss RM. High-Risk Atherosclerosis and Metabolic Phenotype: The Roles of Ectopic Adiposity, Atherogenic Dyslipidemia, and Inflammation. Metab Syndr Relat Disord 2020; 18:176-185. [PMID: 32119801 PMCID: PMC7196362 DOI: 10.1089/met.2019.0115] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Current algorithms for assessing risk of atherosclerotic cardiovascular disease (ASCVD) and, in particular, the reliance on low-density lipoprotein (LDL) cholesterol in conditions where this measurement is discordant with apoB and LDL-particle concentrations fail to identify a sizeable part of the population at high risk for adverse cardiovascular events. This results in missed opportunities for ASCVD prevention, most notably in those with metabolic syndrome, prediabetes, and diabetes. There is substantial evidence that accumulation of ectopic fat and associated metabolic traits are markers for and pathogenic components of high-risk atherosclerosis. Conceptually, the subset of advanced lesions in high-risk atherosclerosis that triggers vascular complications is closely related to a set of coordinated high-risk traits clustering around a distinct metabolic phenotype. A key feature of this phenotype is accumulation of ectopic fat, which, coupled with age-related muscle loss, creates a milieu conducive for the development of ASCVD: atherogenic dyslipidemia, nonresolving inflammation, endothelial dysfunction, hyperinsulinemia, and impaired fibrinolysis. Sustained vascular inflammation, a hallmark of high-risk atherosclerosis, impairs plaque stabilization in this phenotype. This review describes how metabolic and inflammatory processes that are promoted in large measure by ectopic adiposity, as opposed to subcutaneous adipose tissue, relate to the pathogenesis of high-risk atherosclerosis. Clinical biomarkers indicative of these processes provide incremental information to standard risk factor algorithms and advanced lipid testing identifies atherogenic lipoprotein patterns that are below the discrimination level of standard lipid testing. This has the potential to enable improved identification of high-risk patients who are candidates for therapeutic interventions aimed at prevention of ASCVD.
Collapse
Affiliation(s)
- Katharina Lechner
- Department of Prevention, Rehabilitation and Sports Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | | | - Nicolle Kränkel
- Klinik Für Kardiologie, Campus Benjamin Steglitz, Charité—Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Clemens Von Schacky
- Preventive Cardiology, Ludwig-Maximilians University, Munich, Germany
- Omegametrix, Martinsried, Germany
| | - Nicolai Worm
- German University for Prevention and Health Care Management, Saarbrücken, Germany
| | | | - Benjamin Lechner
- Department of Internal Medicine IV, Ludwig-Maximilians University, Munich, Germany
| | - Johannes Scherr
- Department of Prevention, Rehabilitation and Sports Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- University Center for Prevention and Sports Medicine, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | | | - Ronald M. Krauss
- University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
103
|
Mo Z, Huang S, Burnett DJ, Rutledge JC, Hwang DH. Endotoxin May Not Be the Major Cause of Postprandial Inflammation in Adults Who Consume a Single High-Fat or Moderately High-Fat Meal. J Nutr 2020; 150:1303-1312. [PMID: 32040591 DOI: 10.1093/jn/nxaa003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/30/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Metabolic endotoxemia is considered a cause for high-fat diet (HFD)-induced inflammation. However, convincing experimental evidence in humans is scant. OBJECTIVE We determined whether a HFD or moderately HFD increases LPS and LPS-mediated cytokine production in the postprandial blood (PPB). METHODS Ninety-eight volunteers (age: 37.3 ± 1.5 y) from the cross-sectional phenotyping study (PS) and 62 volunteers (age: 26.8 ± 1.2 y) from the intervention study (IS) consumed a breakfast containing 60% kcal fat (HF) and 36% kcal fat (moderately HF), respectively. For the IS, only the results from the placebo group are presented. Blood samples were probed for LPS-mediated cytokine production by incubating them with LPS inhibitor polymyxin B (PMB) for 24 h at 37°C besides the Limulus amebocyte lysate (LAL) assay. Repeated-measures ANOVA was used to compare the temporal changes of metabolic profiles and treatment outcomes. RESULTS At least 87.5% of the plasma LPS measurements in 32 PS volunteers from each time point were below the LAL assay sensitivity (0.002 EU/mL). PMB suppressed IL-1β (P = 0.035) and IL-6 (P = 0.0487) production in the 3 h PPB of the PS after 24 h incubation at 37°C compared to the vehicle control, suggesting the presence of LPS. However, the amount of LPS did not increase the cytokine concentrations in the 3 h PPB above the fasting concentrations. Such suppression was not detected in the PPB of the IS. Treating whole blood with lipoprotein lipase (LPL) significantly (P < 0.05) increased FFA and cytokine (IL-1β, IL-6, TNF-α) concentrations in both studies. CONCLUSION LPS may not be the major cause of postprandial inflammation in healthy adults consuming a moderately HF meal (36% kcal fat, similar to the typical American diet) or a HF meal (60% kcal fat). Plasma FFAs may modulate postprandial inflammation. The prevailing concept of HFD-induced metabolic endotoxemia requires careful re-evaluation. The PS was registered at clinicaltrials.gov as NCT02367287 and the IS as NCT02472171.
Collapse
Affiliation(s)
- Zhenzhen Mo
- USDA-ARS Western Human Nutrition Research Center, 430 West Health Sciences Dr., Davis, CA 95616, USA.,Department of Nutrition, University of California-Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Shurong Huang
- USDA-ARS Western Human Nutrition Research Center, 430 West Health Sciences Dr., Davis, CA 95616, USA
| | - Dustin J Burnett
- USDA-ARS Western Human Nutrition Research Center, 430 West Health Sciences Dr., Davis, CA 95616, USA.,Department of Nutrition, University of California-Davis, One Shields Avenue, Davis, CA 95616, USA
| | - John C Rutledge
- Department of Internal Medicine, School of Medicine, University of California-Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Daniel H Hwang
- USDA-ARS Western Human Nutrition Research Center, 430 West Health Sciences Dr., Davis, CA 95616, USA.,Department of Nutrition, University of California-Davis, One Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|
104
|
Interactions of dietary fat with the gut microbiota: Evaluation of mechanisms and metabolic consequences. Clin Nutr 2020; 39:994-1018. [DOI: 10.1016/j.clnu.2019.05.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/25/2019] [Accepted: 05/01/2019] [Indexed: 12/12/2022]
|
105
|
Ma Y, Wang J, Li Q, Cao B. The Effect of Omega-3 Polyunsaturated Fatty Acid Supplementations on anti-Tumor Drugs in Triple Negative Breast Cancer. Nutr Cancer 2020; 73:196-205. [PMID: 32223441 DOI: 10.1080/01635581.2020.1743873] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Triple-negative breast cancer (TNBC) comprises about 10-20% of all diagnosed breast cancers. Increasing evidence shows that the omega-3 polyunsaturated fatty acids (ω-3PUFAs), docosahexaenoic acid and eicosapentaenoic acid, can influence the development, progression, and prognosis of TNBC In Vivo and In Vitro; however, clinical evidence supporting the effect of ω-3PUFAs on TNBC is lacking. Research has demonstrated that ω-3PUFAs can induce apoptosis in breast cancer cells by inhibiting the PI3K/AKT signal transduction pathway, and that ω-3PUFAs can improve the effectiveness of chemotherapy drugs. Using ω-3PUFA supplementation in addition to pharmacotherapy in the treatment of breast cancer may result in enhanced anti-tumor effects that will be particularly applicable to difficult to treat phenotypes such as TNBC. The aim of the current review was to summarize the evidence-base supporting the antitumor effects of omega-3 PUFAs in TNBC.
Collapse
Affiliation(s)
- Yingjie Ma
- Department of Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Jing Wang
- Department of Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Qin Li
- Department of Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Bangwei Cao
- Department of Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| |
Collapse
|
106
|
Abstract
Many studies highlighted that a bidirectional communication between the gut and the central nervous system (CNS) exists. A vigorous immune response to antigens must be avoided, and pathogenic organisms crossing the gut barrier must be detected and killed. For this reason, the immune system developed fine mechanisms able to maintain this delicate balance. The microbiota is beneficial to its host, providing protection against pathogenic bacteria. It is intimately involved in numerous aspects of host physiology, from nutritional status to behavior and stress response. In the last few years, the implication of the gut microbiota and its bioactive microbiota-derived molecules in the progression of multiple diseases, as well as in the development of neurodegenerative disorders, gained increasing attention. The purpose of this review is to provide an overview of the gut microbiota with particular attention toward neurological disorders and mast cells. Relevant roles are played by the mast cells in neuroimmune communication, such as sensors and effectors of cytokines and neurotransmitters. In this context, the intake of beneficial bacterial strains as probiotics could represent a valuable therapeutic approach to adopt in combination with classical therapies. Further studies need to be performed to understand if the gut bacteria are responsible for neurological disorders or if neurological disorders influence the bacterial profile.
Collapse
|
107
|
Martínez-García MÁ, Ojeda-Ojeda M, Rodríguez-Martín E, Insenser M, Moncayo S, Álvarez-Blasco F, Luque-Ramírez M, Escobar-Morreale HF. TLR2 and TLR4 Surface and Gene Expression in White Blood Cells after Fasting and Oral Glucose, Lipid and Protein Challenges: Influence of Obesity and Sex Hormones. Biomolecules 2020; 10:biom10010111. [PMID: 31936430 PMCID: PMC7023426 DOI: 10.3390/biom10010111] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 12/20/2019] [Accepted: 01/04/2020] [Indexed: 12/19/2022] Open
Abstract
We studied if macronutrients of the diet have different effects on leukocyte activation, and if these effects are influenced by sex hormones or obesity. We analyzed leukocyte cell surface and gene expression of toll-like receptors 2 and 4 (TLR2 and TLR4) during fasting and after macronutrient loads in women with polycystic ovary syndrome and female and male controls. Fasting TLR2 surface expression in neutrophils was higher in men than in women. Obese subjects presented higher TLR2 gene expression than nonobese individuals, particularly in men. In contrast, surface TLR4 expression was lower in men and in obese individuals. Postprandial cell-surface expression decreased similarly after all macronutrient loads. Neutrophil TLR2 decreased only in obese subjects whereas TLR4 showed a greater decrease in nonobese individuals. However, TLR2 gene expression increased after glucose ingestion and decreased during the lipid load, while TLR4 was induced in response to lipids and mostly to glucose. Postprandial TLR gene expression was not influenced by group of subjects or obesity. Both cell-surface and gene postprandial expression inversely correlated with their fasting levels. These responses suggest a transient compensatory response aiming to prevent postprandial inflammation. However, obesity and sex hormones showed opposite influences on surface expression of TLR2 and TLR4, but not on their gene expression, pointing to regulatory posttranscriptional mechanisms.
Collapse
Affiliation(s)
- M. Ángeles Martínez-García
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal &Universidad de Alcalá &Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain; (M.Á.M.-G.); (M.O.-O.); (M.I.); (S.M.); (F.Á.-B.); (M.L.-R.)
| | - Miriam Ojeda-Ojeda
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal &Universidad de Alcalá &Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain; (M.Á.M.-G.); (M.O.-O.); (M.I.); (S.M.); (F.Á.-B.); (M.L.-R.)
| | | | - María Insenser
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal &Universidad de Alcalá &Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain; (M.Á.M.-G.); (M.O.-O.); (M.I.); (S.M.); (F.Á.-B.); (M.L.-R.)
| | - Samuel Moncayo
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal &Universidad de Alcalá &Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain; (M.Á.M.-G.); (M.O.-O.); (M.I.); (S.M.); (F.Á.-B.); (M.L.-R.)
| | - Francisco Álvarez-Blasco
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal &Universidad de Alcalá &Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain; (M.Á.M.-G.); (M.O.-O.); (M.I.); (S.M.); (F.Á.-B.); (M.L.-R.)
| | - Manuel Luque-Ramírez
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal &Universidad de Alcalá &Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain; (M.Á.M.-G.); (M.O.-O.); (M.I.); (S.M.); (F.Á.-B.); (M.L.-R.)
| | - Héctor F. Escobar-Morreale
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal &Universidad de Alcalá &Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain; (M.Á.M.-G.); (M.O.-O.); (M.I.); (S.M.); (F.Á.-B.); (M.L.-R.)
- Correspondence: ; Tel.: +34-91-3369164
| |
Collapse
|
108
|
Zhou H, Urso CJ, Jadeja V. Saturated Fatty Acids in Obesity-Associated Inflammation. J Inflamm Res 2020; 13:1-14. [PMID: 32021375 PMCID: PMC6954080 DOI: 10.2147/jir.s229691] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/11/2019] [Indexed: 01/14/2023] Open
Abstract
Obesity is a major risk factor for the development of various pathological conditions including insulin resistance, diabetes, cardiovascular diseases, and non-alcoholic fatty liver disease (NAFLD). Central to these conditions is obesity-associated chronic low-grade inflammation in many tissues including adipose, liver, muscle, kidney, pancreas, and brain. There is increasing evidence that saturated fatty acids (SFAs) increase the phosphorylation of MAPKs, enhance the activation of transcription factors such as nuclear factor (NF)-κB, and elevate the expression of inflammatory genes. This paper focuses on the mechanisms by which SFAs induce inflammation. SFAs may induce the expression inflammatory genes via different pathways including toll-like receptor (TLR), protein kinase C (PKC), reactive oxygen species (ROS), NOD-like receptors (NLRs), and endoplasmic reticulum (ER) stress. These findings suggest that SFAs act as an important link between obesity and inflammation.
Collapse
Affiliation(s)
- Heping Zhou
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA
| | - C J Urso
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA
| | - Viren Jadeja
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA
| |
Collapse
|
109
|
Naderi N, Souri M, Nasr Esfahani MH, Hajian M, Tanhaei Vash N. Ferulago angulata extract ameliorates epididymal sperm toxicity in mice induced by lead and diazinon. Andrology 2020; 8:706-718. [PMID: 31747138 DOI: 10.1111/andr.12730] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/30/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND The potential toxicity that results from environmental xenobiotics is not completely known. Increasing levels of heavy metals and the use of organophosphate pesticides (OPs) and their co-existence in the environment could be associated with an increasing incidence of male reproductive system disorders in humans and animals. Ferulago angulata is a dietary source of phenolic compounds with reported health benefits. OBJECTIVE This study was conducted to investigate whether an extract of Ferulago angulata could protect adult male NMRI mice against reproductive toxicity induced by lead acetate (PbAc), diazinon (DZN), or PbAc + DZN. MATERIALS AND METHODS Adult male NMRI mice were exposed to either 0.5% PbAc in drinking water, DZN (3 mg/kg/day, intraperitoneal [i.p.] injection), or PbAc + DZN in the presence or absence of 400 mg/kg/day Ferulago angulata hydroalcoholic extract (FAE) that was administered via gavage for 6 weeks. RESULTS Chronic exposure to PbAc, DZN, and PbAc + DZN decreased sperm quality, sperm chromatin maturity and integrity, increased oxidative stress and lipid peroxidation, and could reduce male fertility indices. Co-administration of FAE could reduce these negative effects. CONCLUSION The Ferulago angulata extract should be considered as a useful natural extract for the treatment of male infertility, especially in males exposed to conditions which induce reproductive toxicity.
Collapse
Affiliation(s)
- Nushin Naderi
- Department of Animal Science, College of Agriculture, Razi University, Kermanshah, Iran.,Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Manouchehr Souri
- Department of Animal Science, College of Agriculture, Razi University, Kermanshah, Iran
| | - Mohammad Hossein Nasr Esfahani
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mehdi Hajian
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Nima Tanhaei Vash
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
110
|
Hidalgo-Lanussa O, Baez-Jurado E, Echeverria V, Ashraf GM, Sahebkar A, Garcia-Segura LM, Melcangi RC, Barreto GE. Lipotoxicity, neuroinflammation, glial cells and oestrogenic compounds. J Neuroendocrinol 2020; 32:e12776. [PMID: 31334878 DOI: 10.1111/jne.12776] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023]
Abstract
The high concentrations of free fatty acids as a consequence of obesity and being overweight have become risk factors for the development of different diseases, including neurodegenerative ailments. Free fatty acids are strongly related to inflammatory events, causing cellular and tissue alterations in the brain, including cell death, deficits in neurogenesis and gliogenesis, and cognitive decline. It has been reported that people with a high body mass index have a higher risk of suffering from Alzheimer's disease. Hormones such as oestradiol not only have beneficial effects on brain tissue, but also exert some adverse effects on peripheral tissues, including the ovary and breast. For this reason, some studies have evaluated the protective effect of oestrogen receptor (ER) agonists with more specific tissue activities, such as the neuroactive steroid tibolone. Activation of ERs positively affects the expression of pro-survival factors and cell signalling pathways, thus promoting cell survival. This review aims to discuss the relationship between lipotoxicity and the development of neurodegenerative diseases. We also elaborate on the cellular and molecular mechanisms involved in neuroprotection induced by oestrogens.
Collapse
Affiliation(s)
- Oscar Hidalgo-Lanussa
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Eliana Baez-Jurado
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Valentina Echeverria
- Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile
- Bay Pines VA Healthcare System, Research and Development, Bay Pines, FL, USA
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Roberto C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| |
Collapse
|
111
|
Tamer F, Ulug E, Akyol A, Nergiz-Unal R. The potential efficacy of dietary fatty acids and fructose induced inflammation and oxidative stress on the insulin signaling and fat accumulation in mice. Food Chem Toxicol 2020; 135:110914. [DOI: 10.1016/j.fct.2019.110914] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 10/17/2019] [Accepted: 10/23/2019] [Indexed: 02/09/2023]
|
112
|
Denisenko YK, Kytikova OY, Novgorodtseva TP, Antonyuk MV, Gvozdenko TA, Kantur TA. Lipid-Induced Mechanisms of Metabolic Syndrome. J Obes 2020; 2020:5762395. [PMID: 32963827 PMCID: PMC7491450 DOI: 10.1155/2020/5762395] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/23/2020] [Accepted: 07/30/2020] [Indexed: 12/23/2022] Open
Abstract
Metabolic syndrome (MetS) has a worldwide tendency to increase and depends on many components, which explains the complexity of diagnosis, approaches to the prevention, and treatment of this pathology. Insulin resistance (IR) is the crucial cause of the MetS pathogenesis, which develops against the background of abdominal obesity. In light of recent evidence, it has been shown that lipids, especially fatty acids (FAs), are important signaling molecules that regulate the signaling pathways of insulin and inflammatory mediators. On the one hand, the lack of n-3 polyunsaturated fatty acids (PUFAs) in the body leads to impaired molecular mechanisms of glucose transport, the formation of unresolved inflammation. On the other hand, excessive formation of free fatty acids (FFAs) underlies the development of oxidative stress and mitochondrial dysfunction in MetS. Understanding the molecular mechanisms of the participation of FAs and their metabolites in the pathogenesis of MetS will contribute to the development of new diagnostic methods and targeted therapy for this disease. The purpose of this review is to highlight recent advances in the study of the effect of fatty acids as modulators of insulin response and inflammatory process in the pathogenesis and treatment for MetS.
Collapse
Affiliation(s)
- Yulia K. Denisenko
- Vladivostok Branch of the Far Eastern Scientific Centre of Physiology and Pathology of Respiration, Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok 690105, Russia
| | - Oxana Yu Kytikova
- Vladivostok Branch of the Far Eastern Scientific Centre of Physiology and Pathology of Respiration, Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok 690105, Russia
| | - Tatyana P. Novgorodtseva
- Vladivostok Branch of the Far Eastern Scientific Centre of Physiology and Pathology of Respiration, Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok 690105, Russia
| | - Marina V. Antonyuk
- Vladivostok Branch of the Far Eastern Scientific Centre of Physiology and Pathology of Respiration, Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok 690105, Russia
| | - Tatyana A. Gvozdenko
- Vladivostok Branch of the Far Eastern Scientific Centre of Physiology and Pathology of Respiration, Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok 690105, Russia
| | | |
Collapse
|
113
|
Jacquet A, Robinson C. Proteolytic, lipidergic and polysaccharide molecular recognition shape innate responses to house dust mite allergens. Allergy 2020; 75:33-53. [PMID: 31166610 DOI: 10.1111/all.13940] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 04/05/2019] [Accepted: 05/23/2019] [Indexed: 02/06/2023]
Abstract
House dust mites (HDMs) are sources of an extensive repertoire of allergens responsible for a range of allergic conditions. Technological advances have accelerated the identification of these allergens and characterized their putative roles within HDMs. Understanding their functional bioactivities is illuminating how they interact with the immune system to cause disease and how interrelations between them are essential to maximize allergic responses. Two types of allergen bioactivity, namely proteolysis and peptidolipid/lipid binding, elicit IgE and stimulate bystander responses to unrelated allergens. Much of this influence arises from Toll-like receptor (TLR) 4 or TLR2 signalling and, in the case of protease allergens, the activation of additional pleiotropic effectors with strong disease linkage. Of related interest is the interaction of HDM allergens with common components of the house dust matrix, through either their binding to allergens or their autonomous modulation of immune receptors. Herein, we provide a contemporary view of how proteolysis, lipid-binding activity and interactions with polysaccharides and polysaccharide molecular recognition systems coordinate the principal responses which underlie allergy. The power of the catalytically competent group 1 HDM protease allergen component is demonstrated by a review of disclosures surrounding the efficacy of novel inhibitors produced by structure-based design.
Collapse
Affiliation(s)
- Alain Jacquet
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center-Chula VRC) Chulalongkorn University Bangkok Thailand
| | - Clive Robinson
- Institute for Infection and Immunity St George's, University of London London UK
| |
Collapse
|
114
|
A global perspective on the crosstalk between saturated fatty acids and Toll-like receptor 4 in the etiology of inflammation and insulin resistance. Prog Lipid Res 2019; 77:101020. [PMID: 31870728 DOI: 10.1016/j.plipres.2019.101020] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 12/16/2022]
Abstract
Obesity is featured by chronic systemic low-grade inflammation that eventually contributes to the development of insulin resistance. Toll-like receptor 4 (TLR4) is an important mediator that triggers the innate immune response by activating inflammatory signaling cascades. Human, animal and cell culture studies identified saturated fatty acids (SFAs), the dominant non-esterified fatty acid (NEFA) in the circulation of obese subjects, as non-microbial agonists that trigger the inflammatory response via activating TLR4 signaling, which acts as an important causative link between fatty acid overload, chronic low-grade inflammation and the related metabolic aberrations. The interaction between SFAs and TLR4 may be modulated through the myeloid differentiation primary response gene 88-dependent and independent signaling pathway. Greater understanding of the crosstalk between dietary SFAs and TLR4 signaling in the pathogenesis of metabolic imbalance may facilitate the design of a more efficient pharmacological strategy to alleviate the risk of developing chronic diseases elicited in part by fatty acid overload. The current review discusses recent advances in the impact of crosstalk between SFAs and TLR4 on inflammation and insulin resistance in multiple cell types, tissues and organs in the context of metabolic dysregulation.
Collapse
|
115
|
Marrone MC, Coccurello R. Dietary Fatty Acids and Microbiota-Brain Communication in Neuropsychiatric Diseases. Biomolecules 2019; 10:E12. [PMID: 31861745 PMCID: PMC7022659 DOI: 10.3390/biom10010012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 12/13/2022] Open
Abstract
The gut-brain axis is a multimodal communication system along which immune, metabolic, autonomic, endocrine and enteric nervous signals can shape host physiology and determine liability, development and progression of a vast number of human diseases. Here, we broadly discussed the current knowledge about the either beneficial or deleterious impact of dietary fatty acids on microbiota-brain communication (MBC), and the multiple mechanisms by which different types of lipids can modify gut microbial ecosystem and contribute to the pathophysiology of major neuropsychiatric diseases (NPDs), such as schizophrenia (SCZ), depression and autism spectrum disorders (ASD).
Collapse
Affiliation(s)
- Maria Cristina Marrone
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, 00161 Rome, Italy;
| | - Roberto Coccurello
- National Research Council (CNR), Institute for Complex System (ISC), 00185 Rome, Italy
- IRCCS–S. Lucia Foundation (FSL), 00143 Rome, Italy
| |
Collapse
|
116
|
van Niekerk G, Christowitz C, Conradie D, Engelbrecht AM. Insulin as an immunomodulatory hormone. Cytokine Growth Factor Rev 2019; 52:34-44. [PMID: 31831339 DOI: 10.1016/j.cytogfr.2019.11.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 12/11/2022]
Abstract
Insulin plays an indispensable role in the management of hyperglycaemia that arises in a variety of settings, including Type I and II diabetes, gestational diabetes, as well as is in hyperglycaemia following a severe inflammatory insult. However, insulin receptors are also expressed on a range of cells that are not canonically implicated in glucose homeostasis. This includes immune cells, where the anti-inflammatory effects of insulin have been repeatedly reported. However, recent findings have also implicated a more involved role for insulin in shaping the immune response during an infection. This includes the ability of insulin to modulate immune cell differentiation and polarisation as well as the modulation of effector functions such as biocidal ROS production. Finally, inflammatory mediators can through both direct and indirect mechanisms also regulate serum insulin levels, suggesting that insulin may be co-opted by the immune system during an infection to direct immunological operations. Collectively, these observations implicate insulin as a bona fide immune-modulating hormone and suggest that a better understanding of insulin's immunological function may aid in optimising insulin therapy in a range of clinical settings.
Collapse
Affiliation(s)
- Gustav van Niekerk
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa.
| | - Claudia Christowitz
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Daleen Conradie
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
117
|
Effects of Fat Supplementation in Dairy Goats on Lipid Metabolism and Health Status. Animals (Basel) 2019; 9:ani9110917. [PMID: 31689973 PMCID: PMC6912558 DOI: 10.3390/ani9110917] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/24/2019] [Accepted: 10/27/2019] [Indexed: 12/13/2022] Open
Abstract
Simple Summary There is an increasing demand for information on the nutraceutical properties of food. Due to its bioactive components and high digestibility, goat milk is an excellent functional food. Dietary fat supplementation can further enrich the value of goat milk by modifying its acidic profile. Nevertheless, animal health can also benefit from lipids supplied with rations. In this review, the relationships between dietary fats and goat health status are summarized. Particular attention is paid to describing the effects of specific fatty acids on lipid metabolism and immune functionality. Abstract Fat supplementation has long been used in dairy ruminant nutrition to increase the fat content of milk and supply energy during particularly challenging production phases. Throughout the years, advances have been made in the knowledge of metabolic pathways and technological treatments of dietary fatty acids (FAs), resulting in safer and more widely available lipid supplements. There is an awareness of the positive nutraceutical effects of the addition of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) to fat supplementation, which provides consumers with healthier animal products through manipulation of their characteristics. If it is true that benefits to human health can be derived from the consumption of animal products rich in bioactive fatty acids (FAs), then it is reasonable to think that the same effect can occur in the animals to which the supplements are administered. Therefore, recent advances in fat supplementation of dairy goats with reference to the effect on health status have been summarized. In vivo trials and in vitro analysis on cultured cells, as well as histological and transcriptomic analyses of hepatic and adipose tissue, have been reviewed in order to assess documented relationships between specific FAs, lipid metabolism, and immunity.
Collapse
|
118
|
Wotzka SY, Kreuzer M, Maier L, Arnoldini M, Nguyen BD, Brachmann AO, Berthold DL, Zünd M, Hausmann A, Bakkeren E, Hoces D, Gül E, Beutler M, Dolowschiak T, Zimmermann M, Fuhrer T, Moor K, Sauer U, Typas A, Piel J, Diard M, Macpherson AJ, Stecher B, Sunagawa S, Slack E, Hardt WD. Escherichia coli limits Salmonella Typhimurium infections after diet shifts and fat-mediated microbiota perturbation in mice. Nat Microbiol 2019; 4:2164-2174. [PMID: 31591555 PMCID: PMC6881180 DOI: 10.1038/s41564-019-0568-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 08/23/2019] [Indexed: 12/16/2022]
Abstract
The microbiota confers colonization resistance, which blocks Salmonella gut colonization1. As diet affects microbiota composition, we studied whether food composition shifts enhance susceptibility to infection. Shifting mice to diets with reduced fibre or elevated fat content for 24 h boosted Salmonella Typhimurium or Escherichia coli gut colonization and plasmid transfer. Here, we studied the effect of dietary fat. Colonization resistance was restored within 48 h of return to maintenance diet. Salmonella gut colonization was also boosted by two oral doses of oleic acid or bile salts. These pathogen blooms required Salmonella's AcrAB/TolC-dependent bile resistance. Our data indicate that fat-elicited bile promoted Salmonella gut colonization. Both E. coli and Salmonella show much higher bile resistance than the microbiota. Correspondingly, competitive E. coli can be protective in the fat-challenged gut. Diet shifts and fat-elicited bile promote S. Typhimurium gut infections in mice lacking E. coli in their microbiota. This mouse model may be useful for studying pathogen-microbiota-host interactions, the protective effect of E. coli, to analyse the spread of resistance plasmids and assess the impact of food components on the infection process.
Collapse
Affiliation(s)
- Sandra Y Wotzka
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | - Markus Kreuzer
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | - Lisa Maier
- European Molecular Biology Laboratory, Heidelberg, Heidelberg, Germany
| | - Markus Arnoldini
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | - Bidong D Nguyen
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | | | | | - Mirjam Zünd
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | - Annika Hausmann
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | - Erik Bakkeren
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | - Daniel Hoces
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | - Ersin Gül
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | - Markus Beutler
- Max von Pettenkofer Institute, Faculty of Medicine, LMU Munich, Munich, Germany
| | | | - Michael Zimmermann
- Institute of Molecular Systems Biology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | - Tobias Fuhrer
- Institute of Molecular Systems Biology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | - Kathrin Moor
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | - Uwe Sauer
- Institute of Molecular Systems Biology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | - Athanasios Typas
- European Molecular Biology Laboratory, Heidelberg, Heidelberg, Germany
| | - Jörn Piel
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | - Médéric Diard
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | - Andrew J Macpherson
- Maurice Müller Laboratories, University Clinic for Visceral Surgery and Medicine, University of Bern, Bern, Switzerland
| | - Bärbel Stecher
- Max von Pettenkofer Institute, Faculty of Medicine, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Munich, Germany
| | | | - Emma Slack
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | | |
Collapse
|
119
|
Changes in Plasma Free Fatty Acids Associated with Type-2 Diabetes. Nutrients 2019; 11:nu11092022. [PMID: 31466350 PMCID: PMC6770316 DOI: 10.3390/nu11092022] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/22/2019] [Accepted: 08/24/2019] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is associated with increased total plasma free fatty acid (FFA) concentrations and an elevated risk of cardiovascular disease. The exact mechanisms by which the plasma FFA profile of subjects with T2DM changes is unclear, but it is thought that dietary fats and changes to lipid metabolism are likely to contribute. Therefore, establishing the changes in concentrations of specific FFAs in an individual’s plasma is important. Each type of FFA has different effects on physiological processes, including the regulation of lipolysis and lipogenesis in adipose tissue, inflammation, endocrine signalling and the composition and properties of cellular membranes. Alterations in such processes due to altered plasma FFA concentrations/profiles can potentially result in the development of insulin resistance and coagulatory defects. Finally, fibrates and statins, lipid-regulating drugs prescribed to subjects with T2DM, are also thought to exert part of their beneficial effects by impacting on plasma FFA concentrations. Thus, it is also interesting to consider their effects on the concentration of FFAs in plasma. Collectively, we review how FFAs are altered in T2DM and explore the likely downstream physiological and pathological implications of such changes.
Collapse
|
120
|
Cao D, Wang W, Li S, Lai W, Huang X, Zhou J, Chen X, Li X. TLR2-Deficiency Promotes Prenatal LPS Exposure-Induced Offspring Hyperlipidemia. Front Physiol 2019; 10:1102. [PMID: 31507457 PMCID: PMC6713936 DOI: 10.3389/fphys.2019.01102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/08/2019] [Indexed: 12/15/2022] Open
Abstract
Toll-like receptor 2 (TLR2), which recognizes several lipopeptides and transduces inflammatory signaling, promotes the pathogenesis of diet-induced dyslipidemia and obesity. TLR2-deficient mice were shown to have improved insulin sensitivity and reduced diet-induced metabolic syndrome. Previous studies demonstrated that prenatal lipopolysaccharide (LPS) exposure causes dyslipidemia accompanied by increased body weight and insulin resistance in offspring. To determine whether TLRs are involved in this complex abnormal phenotype, we analyzed TLR2 and TLR4 expression levels in adipose tissues from offspring with prenatal LPS-exposure (offspring-pLPS) and compared these levels to those of control offspring with prenatal saline-exposure (offspring-pSaline). TLR2 expression was specifically upregulated in the adipose tissue of offspring-pLPS mice. However, unexpectedly, TLR2-deficient offspring-pLPS mice not only presented with an abnormal phenotype comparable to that of wild-type offspring-pLPS mice but also exhibited significantly more severe hyperlipidemia. Our further analyses revealed a dramatic upregulation of TLR4 expression and overactivation of the TLR4/Myd88 signaling pathway in TLR2-deficient offspring-pLPS adipose tissue. Our finding suggests a compensatory genetic interaction between TLR2 and TLR4 in the context of prenatal inflammatory stimulation, and this interaction likely contributes to the prenatal inflammation-induced hyperlipidemia and lipid overload-induced obesity, thus providing a potential mechanism for the fetal origin of adult metabolic diseases.
Collapse
Affiliation(s)
- Dayan Cao
- Institute of Materia Medica, Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| | - Wenjia Wang
- Institute of Materia Medica, Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| | - Shuhui Li
- Department of Clinical Biochemistry, College of Pharmacy, Army Medical University, Chongqing, China
| | - Wenjing Lai
- Department of Pharmacy, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiaoyong Huang
- Institute of Immunology, PLA, Army Medical University, Chongqing, China
| | - Jianzhi Zhou
- Institute of Materia Medica, Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| | - Xin Chen
- Institute of Materia Medica, Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| | - Xiaohui Li
- Institute of Materia Medica, Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| |
Collapse
|
121
|
Di Cara F, Andreoletti P, Trompier D, Vejux A, Bülow MH, Sellin J, Lizard G, Cherkaoui-Malki M, Savary S. Peroxisomes in Immune Response and Inflammation. Int J Mol Sci 2019; 20:ijms20163877. [PMID: 31398943 PMCID: PMC6721249 DOI: 10.3390/ijms20163877] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/24/2019] [Accepted: 08/05/2019] [Indexed: 12/11/2022] Open
Abstract
The immune response is essential to protect organisms from infection and an altered self. An organism’s overall metabolic status is now recognized as an important and long-overlooked mediator of immunity and has spurred new explorations of immune-related metabolic abnormalities. Peroxisomes are essential metabolic organelles with a central role in the synthesis and turnover of complex lipids and reactive species. Peroxisomes have recently been identified as pivotal regulators of immune functions and inflammation in the development and during infection, defining a new branch of immunometabolism. This review summarizes the current evidence that has helped to identify peroxisomes as central regulators of immunity and highlights the peroxisomal proteins and metabolites that have acquired relevance in human pathologies for their link to the development of inflammation, neuropathies, aging and cancer. This review then describes how peroxisomes govern immune signaling strategies such as phagocytosis and cytokine production and their relevance in fighting bacterial and viral infections. The mechanisms by which peroxisomes either control the activation of the immune response or trigger cellular metabolic changes that activate and resolve immune responses are also described.
Collapse
Affiliation(s)
- Francesca Di Cara
- Department of Microbiology and Immunology, Dalhousie University, IWK Health Centre, Halifax, NS B3K 6R8, Canada
| | - Pierre Andreoletti
- Lab. Bio-PeroxIL EA7270, University of Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France
| | - Doriane Trompier
- Lab. Bio-PeroxIL EA7270, University of Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France
| | - Anne Vejux
- Lab. Bio-PeroxIL EA7270, University of Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France
| | - Margret H Bülow
- Molecular Developmental Biology, Life & Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Julia Sellin
- Molecular Developmental Biology, Life & Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Gérard Lizard
- Lab. Bio-PeroxIL EA7270, University of Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France
| | - Mustapha Cherkaoui-Malki
- Lab. Bio-PeroxIL EA7270, University of Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France
| | - Stéphane Savary
- Lab. Bio-PeroxIL EA7270, University of Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France.
| |
Collapse
|
122
|
The Lipid Status in Patients with Ulcerative Colitis: Sphingolipids are Disease-Dependent Regulated. J Clin Med 2019; 8:jcm8070971. [PMID: 31277430 PMCID: PMC6678307 DOI: 10.3390/jcm8070971] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/13/2019] [Accepted: 07/02/2019] [Indexed: 02/06/2023] Open
Abstract
The factors that contribute to the development of ulcerative colitis (UC), are still not fully identified. Disruption of the colon barrier is one of the first events leading to invasion of bacteria and activation of the immune system. The colon barrier is strongly influenced by sphingolipids. Sphingolipids impact cell-cell contacts and function as second messengers. We collected blood and colon tissue samples from UC patients and healthy controls and investigated the sphingolipids and other lipids by LC-MS/MS or LC-QTOFMS. The expression of enzymes of the sphingolipid pathway were determined by RT-PCR and immunohistochemistry. In inflamed colon tissue, the de novo-synthesis of sphingolipids is reduced, whereas lactosylceramides are increased. Reduction of dihydroceramides was due to posttranslational inhibition rather than altered serine palmitoyl transferase or ceramide synthase expression in inflamed colon tissue. Furthermore, in human plasma from UC-patients, several sphinglipids change significantly in comparison to healthy controls. Beside sphingolipids free fatty acids, lysophosphatidylcholines and triglycerides changed significantly in the blood of colitis patients dependent on the disease severity. Our data indicate that detraction of the sphingolipid de novo synthesis in colon tissue might be an important trigger for UC. Several lipids changed significantly in the blood, which might be used as biomarkers for disease control; however, diet-related variabilities need to be considered.
Collapse
|
123
|
Lemay DG, Huang S, Huang L, Alkan Z, Kirschke C, Burnett DJ, Wang YE, Hwang DH. Temporal changes in postprandial blood transcriptomes reveal subject-specific pattern of expression of innate immunity genes after a high-fat meal. J Nutr Biochem 2019; 72:108209. [PMID: 31473510 DOI: 10.1016/j.jnutbio.2019.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 12/31/2022]
Abstract
White blood cells are among the first responders to dietary components and their metabolites absorbed from the gut. The objective of this study was to determine the whole blood transcriptome response to high-fat challenge meals. A total of 45 fasting and postprandial (3-h and 6-h) whole blood transcriptomes from 5 subjects in a crossover intervention trial of a high-fat meal supplemented with placebo, blueberry powder or docosahexaenoic acid (DHA) were analyzed using RNA sequencing. Select target genes were validated by quantitative reverse-transcription polymerase chain reaction in 180 samples from 20 subjects. The largest contributor to variance was the subject (13,856 genes differentially expressed), followed by the subject on a specific day (2276 genes), followed by the subject's postprandial response (651 genes). After determining the nonsignificance of individual dietary treatments (blueberry, DHA, placebo), treatments were used as replicates to examine postprandial responses to a high-fat meal. The universal postprandial response (95 genes) was associated with lipid utilization, fatty acid beta-oxidation and circadian rhythms. Subject-specific postprandial responses were enriched for genes involved in the innate immune response, particularly those of pattern recognition receptors and their downstream signaling components. Genes involved in innate immune responses are differentially expressed in a subject-specific and time-dependent manner in response to the high-fat meals. These genes can serve as biomarkers to assess individual responsiveness to a high-fat diet in inducing postprandial inflammation. Furthermore, the dynamic temporal change in gene expression in postprandial blood suggests that monitoring these genes at multiple time points is necessary to reveal responders to dietary intervention.
Collapse
Affiliation(s)
- Danielle G Lemay
- USDA-ARS Western Human Nutrition Research Center, 430 West Health Sciences Dr., Davis, CA 95616; Department of Nutrition, University of California-Davis, One Shields Avenue, Davis, CA, 95616; Genome Center, University of California-Davis, 451 Health Sciences Dr., Davis, CA 95616.
| | - Shurong Huang
- USDA-ARS Western Human Nutrition Research Center, 430 West Health Sciences Dr., Davis, CA 95616.
| | - Liping Huang
- USDA-ARS Western Human Nutrition Research Center, 430 West Health Sciences Dr., Davis, CA 95616; Department of Nutrition, University of California-Davis, One Shields Avenue, Davis, CA, 95616.
| | - Zeynep Alkan
- USDA-ARS Western Human Nutrition Research Center, 430 West Health Sciences Dr., Davis, CA 95616.
| | - Catherine Kirschke
- USDA-ARS Western Human Nutrition Research Center, 430 West Health Sciences Dr., Davis, CA 95616.
| | - Dustin J Burnett
- USDA-ARS Western Human Nutrition Research Center, 430 West Health Sciences Dr., Davis, CA 95616; Department of Nutrition, University of California-Davis, One Shields Avenue, Davis, CA, 95616.
| | - Yining E Wang
- USDA-ARS Western Human Nutrition Research Center, 430 West Health Sciences Dr., Davis, CA 95616.
| | - Daniel H Hwang
- USDA-ARS Western Human Nutrition Research Center, 430 West Health Sciences Dr., Davis, CA 95616; Department of Nutrition, University of California-Davis, One Shields Avenue, Davis, CA, 95616.
| |
Collapse
|
124
|
Zhang X, Xue C, Xu Q, Zhang Y, Li H, Li F, Liu Y, Guo C. Caprylic acid suppresses inflammation via TLR4/NF-κB signaling and improves atherosclerosis in ApoE-deficient mice. Nutr Metab (Lond) 2019; 16:40. [PMID: 31182969 PMCID: PMC6555760 DOI: 10.1186/s12986-019-0359-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 05/01/2019] [Indexed: 12/23/2022] Open
Abstract
Background As reported previously by our group, medium-chain triglycerides can ameliorate atherosclerosis. Given that TLR4 is closely related to atherosclerosis, we hypothesized herein that caprylic acid (C8:0) would suppress inflammation via TLR4/NF-κB signaling and further promote the amelioration of atherosclerosis in apoE- deficient (apoE-/-) mice. Methods Fifty 6-week male apoE-/- mice were randomly allocated into five diet groups: a high-fat diet (HFD) without or with 2% caprylic acid (C8:0), capric acid (C10:0), stearic acid (C18:0), or linolenic acid (C18:3). RAW246.7 cells were treated with caprylic acid (C8:0), docosahexenoic acid (DHA), palmitic acid (C16:0), and lipopolysaccharide (LPS) with or without TLR4 knock-down (TLR4-KD). The serum lipid profiles, inflammatory biomolecules, and mRNA and protein expression levels were measured. Atherosclerotic lesions that occurred in the aorta and aortic sinuses were evaluated and quantified. Results Our results indicated that C8:0 reduced body fat, improved the lipid profiles, suppressed inflammatory cytokine production, downregulated aortic TLR4, MyD88, NF-κB, TNF-α, IKKα, and IKKβ mRNA expression, and alleviated atherosclerosis in the apoE-/- mice (P < 0.05). In RAW 264.7 cells, C8:0 diminished the inflammatory response and both mRNA and protein expression of TLR4, MyD88, NF-κB, and TNF-α compared to those in the LPS and C16:0 groups (P < 0.05). However, in the TLR4-KD RAW 264.7 cells, C8:0 significantly upregulated NF-κB mRNA and protein expression compared to those in the C16:0 and DHA groups. Conclusions These results suggest that C8:0 functions via TLR4/NF-κB signaling to improve the outcomes of apoE-/- mice through suppressing inflammation and ameliorating atherosclerosis. Thus, C8:0 may represent as a promising nutrient against chronic inflammatory diseases.
Collapse
Affiliation(s)
- Xinsheng Zhang
- Department of Nutrition, Tianjin Institute of Environmental & Operational Medicine, Tianjin, 300050 China.,2Department of Nutrition, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853 China
| | - Changyong Xue
- 2Department of Nutrition, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853 China
| | - Qing Xu
- 2Department of Nutrition, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853 China
| | - Yong Zhang
- 2Department of Nutrition, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853 China
| | - Huizi Li
- 3Department of Nutrition, PLA Rocket Force Characteristic Medical Center, Beijing, 100088 China
| | - Feng Li
- 4Department of Nutrition, Air Force Medical Center, PLA, Beijing, 100142 China
| | - Yinghua Liu
- 2Department of Nutrition, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853 China
| | - Changjiang Guo
- Department of Nutrition, Tianjin Institute of Environmental & Operational Medicine, Tianjin, 300050 China
| |
Collapse
|
125
|
Wang W, Wu L, Du X, Zhang F, Ullah SH, Lei T, Li D, Yan X. Anti-Toll-like receptor 2 antibody inhibits nuclear factor kappa B activation and attenuates cardiac damage in high-fat-feeding rats. Acta Biochim Biophys Sin (Shanghai) 2019; 51:347-355. [PMID: 30877771 DOI: 10.1093/abbs/gmz009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 01/10/2019] [Accepted: 01/15/2019] [Indexed: 01/07/2023] Open
Abstract
Long-time consumption of high-fat food is a direct cause of cardiovascular diseases, and high-fat-related inflammation plays an important role in it. Toll-like receptors (TLRs), especially TLR2 and TLR4, play important roles in high-fat-related inflammation. However, the impact of TLR2 on high-fat-associated cardiovascular complications is still unknown. In this study, we try to investigate the relationship between TLR2 and high-fat-related cardiac injury. SD rats were allocated to either a control group which were fed with normal diet or a high-fat group which were fed with high-fat diet for 5 months. At the last month, rats fed with high-fat diet were intraperitoneally injected with control normal mouse IgG or anti-TLR2 antibody. Heart tissues were collected for further analysis. RT-qPCR and western blot analysis results revealed that TLR2 expression was increased in the heart tissues from rats fed with high-fat diet and anti-TLR2 antibody had no effect on TLR2 expression. However, anti-TLR2 antibody alleviated masson staining area, levels of TGF-β1 and Collagen I mRNA, and decreased TUNEL-positive myocardial cells and caspase-3 activity, suggesting that anti-TLR2 antibody protected cardiac cells against high-fat-induced cardiac fibrosis and cell apoptosis. By using immunohistochemistry, RT-qPCR and ELISA, we found that anti-TLR2 antibody blocked NF-κB activation, inhibited the expression of inflammatory factors such as TNF-α, IL-1β, IL-6 and IL-18 in the heart tissues from rats fed with high-fat diet. These results hinted that anti-TLR2 antibody might exert its protective effect via inhibition of the TLR2/NF-κB/inflammation pathway. Our findings suggest that anti-TLR2 antibody has a preventive function against high-fat-induced deleterious effects in the heart, and anti-TLR2 antibody may be used as an attractive therapeutic option for high-fat-induced cardiac injury.
Collapse
Affiliation(s)
- Wanzheng Wang
- Department of Pathology, Medical College of Xi’an Jiaotong University, Xi’an, China
- The Hanjiang River Hospital Affiliated of Xi’an Medical University, Hanzhong, China
| | - Litao Wu
- Department of Biochemistry and Molecular Biology, Medical College of Xi’an Jiaotong University, Xi’an, China
| | - Xiaojuan Du
- Department of Biochemistry and Molecular Biology, Medical College of Xi’an Jiaotong University, Xi’an, China
| | - Fujun Zhang
- Department of Biochemistry and Molecular Biology, Medical College of Xi’an Jiaotong University, Xi’an, China
| | - Sayyed Hanif Ullah
- Department of Biochemistry and Molecular Biology, Medical College of Xi’an Jiaotong University, Xi’an, China
| | - Ting Lei
- The Hanjiang River Hospital Affiliated of Xi’an Medical University, Hanzhong, China
| | - Dongming Li
- Department of Biochemistry and Molecular Biology, Medical College of Xi’an Jiaotong University, Xi’an, China
| | - Xiaofei Yan
- Department of Biochemistry and Molecular Biology, Medical College of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
126
|
Nematollahi A, Kazeminasab F, Tavalaee M, Marandi SM, Ghaedi K, Nazem MN, Nasr-Esfahani MH. Effect of aerobic exercise, low-fat and high-fat diet on the testis tissue and sperm parameters in obese and nonobese mice model. Andrologia 2019; 51:e13273. [PMID: 30920027 DOI: 10.1111/and.13273] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/16/2019] [Accepted: 02/19/2019] [Indexed: 12/14/2022] Open
Abstract
Semen quality and male fertility depend on numerous factors such as age, environment, lifestyle, physical activity, genetic background and occupation. We aimed to access the effect of aerobic exercise, low- and high-fat diet on mice testis tissue, and sperm function. Obese and nonobese male mice C57BL/6 were exposed to high fat (Hf) or low fat (Lf) and/or activity (Exe: exercise or Sed: sedentary). Finally, testicular morphometric characteristics, sperm concentration and motility (light microscopy), sperm morphology (eosin/nigrosin dye), lipid peroxidation (BODIPY C11 Probe), chromatin (acridine orange and chromomycin A3 staining) were compared within obese groups (Hf/Exe, Lf/Exe, Lf/Sed, Hf/Sed) and nonobese groups (Hf/Exe, Lf/Exe, Lf/Sed, Hf/Sed). Both exercise and diet interventions did not show any alteration in testicular morphological characteristics, sperm morphology and DNA fragmentation within both obese and nonobese groups (p > 0.05). Exercise and/or diet resulted in a significant increase in sperm concentration and motility within both groups (p < 0.05). Exercise in both groups leads to high percentage of lipid peroxidation (p < 0.05). Exercise intervention significantly improved sperm protamine deficiency within obese group (p < 0.05). We concluded that exercise intervention was more effective than diet in improvement of sperm function within obese groups.
Collapse
Affiliation(s)
- Arezou Nematollahi
- ACECR Institute of Higher Education, Isfahan, Iran.,Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Fatemeh Kazeminasab
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Isfahan, Isfahan, Iran
| | - Marziyeh Tavalaee
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Sayed M Marandi
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Isfahan, Isfahan, Iran
| | - Kamran Ghaedi
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.,Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Mohammad N Nazem
- Department of Basic Sciences, School of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
127
|
Mužáková V, Meloun M, Jindrová A, Čegan A. The effect of fatty acids in red blood cell membranes on the dynamics of inflammatory markers following the coronary stent implantation. J Pharm Biomed Anal 2019; 166:310-325. [PMID: 30690246 DOI: 10.1016/j.jpba.2019.01.002] [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: 12/20/2018] [Revised: 01/02/2019] [Accepted: 01/03/2019] [Indexed: 10/27/2022]
Abstract
The effect of 20 fatty acids in erythrocyte cell membranes on the extent of inflammatory response and cell oxidative stress was evaluated using multidimensional statistical data analysis in 54 patients suffering from ischemic heart disease undergoing percutaneous coronary intervention with coronary stent implantation using multidimensional statistical data analysis. A systemic inflammatory response was indicated by an increase of C-reactive protein (CRP), serum amyloid A (SAA) and ceruloplasmin 48 h after stent implantation and by an increase of interleukin-6 (IL-6) 24 h after intervention. The increase of malondialdehyde (MDA) after 48 h was used as a marker of cell damage by oxidative stress. Multiple linear regression revealed statistically significant relationships between concentration of some fatty acids and the magnitude of inflammatory response, or oxidative stress, after stent implantation. The most significant relationship with an increase of plasma CRP was found for myristic acid and, to a lesser extent, for oleic acid. Trans octadecenoic acid, and to a lesser extent palmitooleic and nervonic fatty acids were found in inverse correlation with the CRP increase. The increase of IL-6 showed a statistically significant correlation with myristic acid, to a lesser extent with cis-9-eicosenoic acid and to the least extent with docosahexaenoic acid, inversely with pentadecanoic, γ-linolenic and stearic acids. An increase of oxidative stress (MDA) significantly correlated only with γ-linolenic acid. Other studied markers of inflammatory response to coronary stenting were SAA and ceruloplasmin (Cp). Statistical evaluation revealed that SAA and Cp are not suitable markers for assessment relationships between inflammation and erythrocyte membrane fatty acids.
Collapse
Affiliation(s)
- Vladimíra Mužáková
- Department of Biological and Biochemical Sciences, University of Pardubice, 532 10 Pardubice, Czech Republic
| | - Milan Meloun
- Department of Analytical Chemistry, University of Pardubice, 532 10 Pardubice, Czech Republic.
| | - Andrea Jindrová
- Department of Biological and Biochemical Sciences, University of Pardubice, 532 10 Pardubice, Czech Republic
| | - Alexander Čegan
- Department of Biological and Biochemical Sciences, University of Pardubice, 532 10 Pardubice, Czech Republic
| |
Collapse
|
128
|
Walker ME, Matthan NR, Solano-Aguilar G, Jang S, Lakshman S, Molokin A, Faits T, Urban JF, Johnson WE, Lamon-Fava S, Lichtenstein AH. A Western-type dietary pattern and atorvastatin induce epicardial adipose tissue interferon signaling in the Ossabaw pig. J Nutr Biochem 2019; 67:212-218. [PMID: 30981985 DOI: 10.1016/j.jnutbio.2019.02.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/17/2019] [Accepted: 02/11/2019] [Indexed: 12/17/2022]
Abstract
Epicardial adipose tissue (EAT) inflammation is thought to potentiate the development of coronary artery disease (CAD). Overall diet quality and statin therapy are important modulators of inflammation and CAD progression. Our objective was to examine the effects and interaction of dietary patterns and statin therapy on EAT gene expression in the Ossabaw pig. Pigs were randomized to 1 of 4 groups; Heart Healthy diet (high in unsaturated fat, unrefined grain, fruits/vegetables [HHD]) or Western diet (high in saturated fat, cholesterol, refined grain [WD]), with or without atorvastatin. Diets were fed in isocaloric amounts for 6 months. A two-factor edge R analysis identified the differential expression of 21 genes. Relative to the HHD, the WD resulted in a significant 12-fold increase of radical s-adenosyl methionine domain containing 2 (RSAD2), a gene induced by interferon signaling. Atorvastatin led to the significant differential expression of 17 genes predominately involved in interferon signaling. Results were similar using the Porcine Translational Research Database. Pathway analysis confirmed the up-regulation of interferon signaling in response to the WD and atorvastatin independently. An expression signature of the largely interferon related differentially expressed genes had no predictive capability on a histological assessment of atherosclerosis in the underlying coronary artery. These results suggest that a WD and atorvastatin evoke an interferon mediated immune response in EAT of the Ossabaw pig, which is not associated with the presence of atherosclerosis.
Collapse
Affiliation(s)
- Maura E Walker
- Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Nirupa R Matthan
- Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Gloria Solano-Aguilar
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Saebyeol Jang
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Sukla Lakshman
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Aleksey Molokin
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Tyler Faits
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Joseph F Urban
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - W Evan Johnson
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Stefania Lamon-Fava
- Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Alice H Lichtenstein
- Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA.
| |
Collapse
|
129
|
Abekura F, Park J, Kwak CH, Ha SH, Cho SH, Chang YC, Ha KT, Chang HW, Lee YC, Chung TW, Kim CH. Esculentoside B inhibits inflammatory response through JNK and downstream NF-κB signaling pathway in LPS-triggered murine macrophage RAW 264.7 cells. Int Immunopharmacol 2019; 68:156-163. [PMID: 30639961 DOI: 10.1016/j.intimp.2019.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 12/24/2018] [Accepted: 01/03/2019] [Indexed: 01/02/2023]
Abstract
Natural compound esculentoside B (EsB), (2S,4aR,6aR,6aS,6bR,8aR,9R,10R,11S,12aR,14bS)-11-hydroxy-9-(hydroxymethyl)-2 methoxycarbonyl-2,6a,6b,9,12a-pentamethyl-10-[(2S,3R,4S,5R)-3,4,5-trihydroxyoxan-2-yl]oxy-1,3,4,5,6,6a,7,8,8a,10,11,12,13,14b-tetradecahydropicene-4a-carboxylic acid with molecular weight of 664.833, isolated from roots of Phytolacca acinosa Roxb has been widely used as a constituent of traditional Chinese medicine (TCM). However, the anti-inflammatory capacity of EsB has not been reported yet. Therefore, the objective of this study was to investigate anti-inflammatory activities of EsB in LPS-treated macrophage RAW 264.7 cells. EsB could inhibit nitric oxide (NO) production. EsB also suppressed gene and protein expression levels of inducible isoform of NO synthase (NOS) and cyclooxygenase-2 in a dose-dependent manner. In addition, EsB decreased gene expression and protein secretion levels of pro-inflammatory cytokines such as IL-1β, TNF-α, and IL-6. EsB remarkably suppressed nuclear translocation of nuclear factor kappa-B (NF-κB) from cytosolic space. Phosphorylation of IκB was also inhibited by EsB. Moreover, EsB specifically down-regulated phospho-c-Jun N-terminal kinase (p-JNK), but not p-p38 or phospho-extracellular signal-regulated kinase 1/2 (p-ERK1/2). Taken together, these results suggest that EsB has inhibitory effect on inflammatory response by inactivating NF-κB and p-JNK. It could be used as a new modulatory drug for effective treatment of inflammation-related diseases.
Collapse
Affiliation(s)
- Fukushi Abekura
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, Sungkyunkwan University, Seoburo 2066, Jangan-Gu, Suwon, Gyunggi-Do 16419, Republic of Korea
| | - Junyoung Park
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, Sungkyunkwan University, Seoburo 2066, Jangan-Gu, Suwon, Gyunggi-Do 16419, Republic of Korea
| | - Choong-Hwan Kwak
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, Sungkyunkwan University, Seoburo 2066, Jangan-Gu, Suwon, Gyunggi-Do 16419, Republic of Korea; Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan City, Gyeongsangnam-Do, Republic of Korea
| | - Sun-Hyung Ha
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, Sungkyunkwan University, Seoburo 2066, Jangan-Gu, Suwon, Gyunggi-Do 16419, Republic of Korea
| | - Seung-Hak Cho
- Division of Enteric Diseases, Center for Infectious Diseases Research, Korea National Institute of Health, Heungdeok-gu, Cheongju 363-951, Republic of Korea
| | - Young-Chae Chang
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea.
| | - Ki-Tae Ha
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan City, Gyeongsangnam-Do, Republic of Korea.
| | - Hyeun-Wook Chang
- College of Pharmacy, Yeungnam University, Gyeongsan 701-947, Republic of Korea
| | - Young-Choon Lee
- Faculty of Medicinal Biotechnology, Dong-A University, Saha-Gu, Busan, Republic of Korea.
| | - Tae-Wook Chung
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan City, Gyeongsangnam-Do, Republic of Korea
| | - Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, Sungkyunkwan University, Seoburo 2066, Jangan-Gu, Suwon, Gyunggi-Do 16419, Republic of Korea.
| |
Collapse
|
130
|
Alí A, Boutjdir M, Aromolaran AS. Cardiolipotoxicity, Inflammation, and Arrhythmias: Role for Interleukin-6 Molecular Mechanisms. Front Physiol 2019; 9:1866. [PMID: 30666212 PMCID: PMC6330352 DOI: 10.3389/fphys.2018.01866] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/11/2018] [Indexed: 12/12/2022] Open
Abstract
Fatty acid infiltration of the myocardium, acquired in metabolic disorders (obesity, type-2 diabetes, insulin resistance, and hyperglycemia) is critically associated with the development of lipotoxic cardiomyopathy. According to a recent Presidential Advisory from the American Heart Association published in 2017, the current average dietary intake of saturated free-fatty acid (SFFA) in the US is 11–12%, which is significantly above the recommended <10%. Increased levels of circulating SFFAs (or lipotoxicity) may represent an unappreciated link that underlies increased vulnerability to cardiac dysfunction. Thus, an important objective is to identify novel targets that will inform pharmacological and genetic interventions for cardiomyopathies acquired through excessive consumption of diets rich in SFFAs. However, the molecular mechanisms involved are poorly understood. The increasing epidemic of metabolic disorders strongly implies an undeniable and critical need to further investigate SFFA mechanisms. A rapidly emerging and promising target for modulation by lipotoxicity is cytokine secretion and activation of pro-inflammatory signaling pathways. This objective can be advanced through fundamental mechanisms of cardiac electrical remodeling. In this review, we discuss cardiac ion channel modulation by SFFAs. We further highlight the contribution of downstream signaling pathways involving toll-like receptors and pathological increases in pro-inflammatory cytokines. Our expectation is that if we understand pathological remodeling of major cardiac ion channels from a perspective of lipotoxicity and inflammation, we may be able to develop safer and more effective therapies that will be beneficial to patients.
Collapse
Affiliation(s)
- Alessandra Alí
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States.,Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Mohamed Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States.,Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Ademuyiwa S Aromolaran
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States.,Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY, United States
| |
Collapse
|
131
|
Kiewiet MBG, Dekkers R, van Gool MP, Ulfman LH, Groeneveld A, Faas MM, de Vos P. Identification of a TLR2 Inhibiting Wheat Hydrolysate. Mol Nutr Food Res 2018; 62:e1800716. [PMID: 30354027 PMCID: PMC6646915 DOI: 10.1002/mnfr.201800716] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/01/2018] [Indexed: 12/25/2022]
Abstract
SCOPE Wheat hydrolysates are used in medical nutrition to provide undernourished patients a readily digestible protein source, for instance to recover from chemotherapy-induced intestinal mucosal inflammation. Since many hydrolysates of different sources can modulate the immune system, likely via Toll-like receptors (TLRs), it is hypothesized that also wheat hydrolysates might interact with TLR signaling, which could be a way to prevent intestinal inflammation and damage. METHODS AND RESULTS The capacity of three wheat hydrolysates to modulate immunity by interfering with TLR signaling is determined. All wheat hydrolysates have TLR modulating effects but only one has strong TLR2 inhibiting effects, attenuating both TLR2/1 and TLR2/6 signaling in a reporter cell system. This is likely induced by direct TLR2-ectodomain binding, as confirmed by ELISA. Furthermore, this TLR2 blocking hydrolysate reduces IL-6 production in human dendritic cells. Application of reversed-phase-ultra HPLC combined with MS reveals that the presence of peptide WQIPEQSR is associated with the observed TLR2 inhibiting capacity. CONCLUSION The study demonstrates TLR2-inhibiting capacities of a wheat hydrolysate. The findings provide a good start for further research to investigate whether this hydrolysate might contribute to the management of intestinal mucosal inflammation in cancer patients receiving chemotherapy.
Collapse
Affiliation(s)
- Mensiena B. G. Kiewiet
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of GroningenUniversity Medical Center GroningenHanzeplein 19700RBGroningenThe Netherlands
| | - Renske Dekkers
- FrieslandCampinaStationsplein 43818LEAmersfoortThe Netherlands
| | | | | | | | - Marijke M. Faas
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of GroningenUniversity Medical Center GroningenHanzeplein 19700RBGroningenThe Netherlands
- Department of Obstetrics and GynecologyUniversity of GroningenUniversity Medical Center Groningen9713GZGroningenThe Netherlands
| | - Paul de Vos
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of GroningenUniversity Medical Center GroningenHanzeplein 19700RBGroningenThe Netherlands
| |
Collapse
|
132
|
Nicoli F, Paul S, Appay V. Harnessing the Induction of CD8 + T-Cell Responses Through Metabolic Regulation by Pathogen-Recognition-Receptor Triggering in Antigen Presenting Cells. Front Immunol 2018; 9:2372. [PMID: 30410483 PMCID: PMC6209652 DOI: 10.3389/fimmu.2018.02372] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 09/24/2018] [Indexed: 01/03/2023] Open
Abstract
Cytotoxic CD8+ T-cells are key players of the immune responses against viruses. During the priming of a CD8+ T-cell response, the activation of a naïve T-cell by a professional antigen presenting cell (APC) involves the induction of various intracellular and metabolic pathways. The modulation of these pathways at the level of APCs or T-cells offers great potential to enhance the induction of robust effector cells and the generation of long-lived memory cells. On the one hand, signaling through pathogen recognition receptors (PRRs) expressed by APCs can greatly influence T-cell priming, and the potential of several PRR ligands as adjuvants are being studied. On the other hand, the engagement of several metabolic processes, at play in APCs and T-cells upon stimulation, implies that modulating cellular metabolism can impact on priming efficacy. Here, we review recent efforts to understand the interplay between PRR mediated signaling and metabolic pathway modulation in this context, through three examples: interplay between TLR4 and fatty acid metabolism, between TLR9 and IDO, and between STING and autophagy. These initial works highlight the potential for harnessing the induction of antiviral CD8+ T-cell responses using synergistic modulation of metabolic and PRR pathways.
Collapse
Affiliation(s)
- Francesco Nicoli
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Stéphane Paul
- GIMAP/EA3064, Université de Lyon, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Victor Appay
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses, Paris, France.,International Research Center of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
133
|
α-Mangostin Alleviated Lipopolysaccharide Induced Acute Lung Injury in Rats by Suppressing NAMPT/NAD Controlled Inflammatory Reactions. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:5470187. [PMID: 30405740 PMCID: PMC6199890 DOI: 10.1155/2018/5470187] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/08/2018] [Accepted: 09/23/2018] [Indexed: 12/18/2022]
Abstract
α-Mangostin (MAN) is a bioactive xanthone isolated from mangosteen. This study was designed to investigate its therapeutic effects on acute lung injury (ALI) and explore the underlying mechanisms of action. Rats from treatment groups were subject to oral administration of MAN for 3 consecutive days beforehand, and then ALI was induced in all the rats except for normal controls via an intraperitoneal injection with lipopolysaccharide. The severity of disease was evaluated by histological examination and hematological analysis. Protein expressions in tissues and cells were examined with immunohistochemical and immunoblotting methods, respectively. The levels of cytokines and nicotinamide adenine dinucleotide (NAD) were determined using ELISA and colorimetric kits, respectively. It was found that MAN treatment significantly improved histological conditions, reduced leucocytes counts, relieved oxidative stress, and declined TNF-α levels in ALI rats. Meanwhile, MAN treatment decreased expressions of nicotinamide phosphoribosyltransferase (NAMPT) and Sirt1 both in vivo and in vitro, which was accompanied with a synchronized decline of NAD and TNF-α. Immunoblotting assay further showed that MAN downregulated HMGB1, TLR4, and p-p65 in RAW 264.7 cells. MAN induced declines of both HMGB1/TLR4/p-p65 and TNF-α were substantially reversed by cotreatment with nicotinamide mononucleotide or NAD. These results suggest that downregulation of NAMPT/NAD by MAN treatments contributes to the alleviation of TLR4/NF-κB-mediated inflammations in macrophage, which is essential for amelioration of ALI in rats.
Collapse
|
134
|
Chevre R, Silvestre-Roig C, Soehnlein O. Nutritional Modulation of Innate Immunity: The Fat-Bile-Gut Connection. Trends Endocrinol Metab 2018; 29:686-698. [PMID: 30197155 DOI: 10.1016/j.tem.2018.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/31/2018] [Accepted: 08/02/2018] [Indexed: 02/08/2023]
Abstract
Altered nutritional behavior in Western societies has unleashed numerous metabolic disorders, intimately linked to profound disruptions of the immune system. Here we summarize how nutrition modulates innate immunity. We outline recent findings regarding nutrient signaling and we particularly focus on the collateral impact of nutrition on the microbiome and on the bile acid (BA) pool. We discuss how the integration of postprandial signals by the gut microbiota, along with the absorption routes of metabolites, differentially affects immune niches to orchestrate immune responses. Finally, we discuss the potential consequences of these signals in the light of trained immunity. A better understanding of nutrition signaling will permit the optimization of therapeutic and dietary strategies against the arising immune disorders.
Collapse
Affiliation(s)
- Raphael Chevre
- Institute for Cardiovascular Prevention, LMU Munich, Munich, Germany.
| | | | - Oliver Soehnlein
- Institute for Cardiovascular Prevention, LMU Munich, Munich, Germany; Department of Physiology and Pharmacology (FyFa) and Department of Medicine, Karolinska Institutet, Stockholm, Sweden; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
135
|
García-Cazorla Y, Getino M, Sanabria-Ríos DJ, Carballeira NM, de la Cruz F, Arechaga I, Cabezón E. Conjugation inhibitors compete with palmitic acid for binding to the conjugative traffic ATPase TrwD, providing a mechanism to inhibit bacterial conjugation. J Biol Chem 2018; 293:16923-16930. [PMID: 30201608 DOI: 10.1074/jbc.ra118.004716] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/04/2018] [Indexed: 01/14/2023] Open
Abstract
Bacterial conjugation is a key mechanism by which bacteria acquire antibiotic resistance. Therefore, conjugation inhibitors (COINs) are promising compounds in the fight against the spread of antibiotic resistance genes among bacteria. Unsaturated fatty acids (uFAs) and alkynoic fatty acid derivatives, such as 2-hexadecanoic acid (2-HDA), have been reported previously as being effective COINs. The traffic ATPase TrwD, a VirB11 homolog in plasmid R388, is the molecular target of these compounds, which likely affect binding of TrwD to bacterial membranes. In this work, we demonstrate that COINs are abundantly incorporated into Escherichia coli membranes, replacing palmitic acid as the major component of the membrane. We also show that TrwD binds palmitic acid, thus facilitating its interaction with the membrane. Our findings also suggest that COINs bind TrwD at a site that is otherwise occupied by palmitic acid. Accordingly, molecular docking predictions with palmitic acid indicated that it shares the same binding site as uFAs and 2-HDA, although it differs in the contacts involved in this interaction. We also identified 2-bromopalmitic acid, a palmitate analog that inhibits many membrane-associated enzymes, as a compound that effectively reduces TrwD ATPase activity and bacterial conjugation. Moreover, we demonstrate that 2-bromopalmitic and palmitic acids both compete for the same binding site in TrwD. Altogether, these detailed findings open up a new avenue in the search for effective synthetic inhibitors of bacterial conjugation, which may be pivotal for combating multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Yolanda García-Cazorla
- From the Departamento de Biología Molecular and Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-Consejo Superior de Investigaciones Científicas, 39011 Santander, Spain
| | - María Getino
- From the Departamento de Biología Molecular and Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-Consejo Superior de Investigaciones Científicas, 39011 Santander, Spain
| | - David J Sanabria-Ríos
- the Inter-American University of Puerto Rico, Metropolitan Campus, Faculty of Science and Technology, San Juan, Puerto Rico 00919, and
| | - Néstor M Carballeira
- the Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico 00925
| | - Fernando de la Cruz
- From the Departamento de Biología Molecular and Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-Consejo Superior de Investigaciones Científicas, 39011 Santander, Spain
| | - Ignacio Arechaga
- From the Departamento de Biología Molecular and Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-Consejo Superior de Investigaciones Científicas, 39011 Santander, Spain,
| | - Elena Cabezón
- From the Departamento de Biología Molecular and Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-Consejo Superior de Investigaciones Científicas, 39011 Santander, Spain,
| |
Collapse
|
136
|
Haygert P, Roversi K, Milanesi LH, Maurer LH, Camponogara C, Duarte T, Barcelos RCS, Emanuelli T, Oliveira SM, Duarte MMMF, Trevizol F, Burger ME. Can the dietary fat type facilitate memory impairments in adulthood? A comparative study between Mediterranean and Western-based diet in rats. J Nutr Biochem 2018; 59:104-113. [DOI: 10.1016/j.jnutbio.2018.05.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/18/2018] [Accepted: 05/10/2018] [Indexed: 12/13/2022]
|
137
|
Messedi M, Naifar M, Grayaa S, Frikha F, Messoued M, Sethom MM, Feki M, Kaabach N, Bahloul Z, Jamoussi K, Ayedi F. Plasma Saturated and Monounsaturated Fatty Acids in Behçet's Disease. Open Rheumatol J 2018; 12:139-151. [PMID: 30258503 PMCID: PMC6128021 DOI: 10.2174/1874312901812010139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/09/2018] [Accepted: 07/05/2018] [Indexed: 12/11/2022] Open
Abstract
Background: Fatty Acid (FA) composition of serum has been associated with many markers of inflammation. In this study, we tried to examine plasma Saturated Fatty Acid (SFA) and Monounsaturated Fatty Acid (MUFA) composition in Behçet's Disease (BD) patients. The associations between the circulating FA levels and some markers of inflammation have also been investigated. Methods: This study is a cross-sectional one. In fact, a total of 101 BD patients and healthy controls group of 99 subjects are enrolled. Gas Chromatograph equipped with a Capillary Split/Splitless Injector and flame ionization detector was used to analyze the plasma SFA and MUFA compositions. The high sensitivity C-Reactive Protein (hsCRP) and fibrinogen levels were measured using standard techniques. Results: BD patients had significantly higher proportions of Mystiric Acid (MA), Palmitic Acid (PAM), Palmitoleic Acid (POA) and Stearoyl-CoA Desaturase (SCD)-16, compared to controls. The results revealed that patients with severe involvements had high levels of POA and total MUFA associated with higher SCD-16 activity compared to those with minor ones. The receiver operator characteristic curve analysis revealed that POA could well discriminate BD patients with severe clinical manifestations. In the bivariate analysis, hsCRP was found to be positively correlated with total SAFA and POA elongase activity index but negatively correlated with SCD-18 activity index. The STA, POA, elongase and SCD-16 activity index are correlated with fibrinogen. On the other hand, the multivariate analysis showed that POA remained associated with higher levels of hsCRP. Conclusion: Unfavourable plasma SFA and MUFA profile were reported in BD patients. POA, which is associated with higher plasma hsCRP level, may play a role in the pathogenesis of BD.
Collapse
Affiliation(s)
- Meriam Messedi
- Unit of Research Molecular Bases of Human Diseases, 12ES17, Faculty of Medicine of Sfax, University of Sfax, 3029 Sfax, Sfax, Tunisia
| | - Manel Naifar
- Biochemistry laboratory, Habib Bourguiba University Hospital, 3029 Sfax, Sfax, Tunisia
| | - Sahar Grayaa
- Unit of Research Molecular Bases of Human Diseases, 12ES17, Faculty of Medicine of Sfax, University of Sfax, 3029 Sfax, Sfax, Tunisia
| | - Faten Frikha
- Internal Medicine Department, Hedi Chaker Hospital, 3029 Sfax, Sfax, Tunisia
| | - Mariem Messoued
- Unit of Research Molecular Bases of Human Diseases, 12ES17, Faculty of Medicine of Sfax, University of Sfax, 3029 Sfax, Sfax, Tunisia
| | - Mohamed Marouene Sethom
- Faculty of Medicine of Tunis, Biochemistry laboratory, La Rabta Hospital and UR05/08-08, Tunis 1007, Tunisia
| | - Moncef Feki
- Faculty of Medicine of Tunis, Biochemistry laboratory, La Rabta Hospital and UR05/08-08, Tunis 1007, Tunisia
| | - Naziha Kaabach
- Faculty of Medicine of Tunis, Biochemistry laboratory, La Rabta Hospital and UR05/08-08, Tunis 1007, Tunisia
| | - Zouheir Bahloul
- Internal Medicine Department, Hedi Chaker Hospital, 3029 Sfax, Sfax, Tunisia
| | - Kamel Jamoussi
- Unit of Research Molecular Bases of Human Diseases, 12ES17, Faculty of Medicine of Sfax, University of Sfax, 3029 Sfax, Sfax, Tunisia
| | - Fatma Ayedi
- Unit of Research Molecular Bases of Human Diseases, 12ES17, Faculty of Medicine of Sfax, University of Sfax, 3029 Sfax, Sfax, Tunisia.,Biochemistry laboratory, Habib Bourguiba University Hospital, 3029 Sfax, Sfax, Tunisia
| |
Collapse
|
138
|
Wiciński M, Wódkiewicz E, Słupski M, Walczak M, Socha M, Malinowski B, Pawlak-Osińska K. Neuroprotective Activity of Sitagliptin via Reduction of Neuroinflammation beyond the Incretin Effect: Focus on Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6091014. [PMID: 30186862 PMCID: PMC6116461 DOI: 10.1155/2018/6091014] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/22/2018] [Accepted: 07/26/2018] [Indexed: 12/25/2022]
Abstract
Sitagliptin is a member of a class of drugs that inhibit dipeptidyl peptidase (DPP-4). It increases the levels of the active form of incretins such as GLP-1 (glucagon-like peptide-1) or GIP (gastric inhibitory polypeptide) and by their means positively affects glucose metabolism. It is successfully applied in the treatment of diabetes mellitus type 2. The most recent scientific reports suggest beneficial effect of sitagliptin on diseases in which neuron damage occurs. Result of experimental studies may indicate a reducing influence of sitagliptin on inflammatory response within encephalon area. Sitagliptin decreased the levels of proinflammatory factors: TNF-α (tumor necrosis factor-α), IL-6 (interleukin-6), IL-17 (interleukin-17), and CD-163 (cluster of differentiation 163), and contributed to an increase in levels of anti-inflammatory factors: IL-10 (interleukin-10) and TGF-β (transforming growth factor β). Moreover, sitagliptin demonstrated antioxidative and antiapoptotic properties by modifying glutamate and glutathione levels within the region of hippocampus in mice. It has been observed that sitagliptin decreases accumulation of β-amyloid within encephalon structures in experimental models of Alzheimer's dementia. This effect may be connected with SDF-1α (stromal cell-derived factor 1α) concentration. Administration of sitagliptin caused a significant improvement in MMSE (Mini-Mental State Examination) tests used for assessment of dementias. The paper presents potential mechanisms of sitagliptin activity in conditions connected with neuroinflammation with special emphasis on Alzheimer's disease.
Collapse
Affiliation(s)
- Michał Wiciński
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland
| | - Eryk Wódkiewicz
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland
| | - Maciej Słupski
- Department of Hepatobiliary and General Surgery, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland
| | - Maciej Walczak
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland
| | - Maciej Socha
- Department of Obstetrics, Gynecology and Gynecological Oncology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Ujejskiego 75, 85-168 Bydgoszcz, Poland
| | - Bartosz Malinowski
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland
| | - Katarzyna Pawlak-Osińska
- Department of Pathophysiology of Hearing and Balance System, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland
| |
Collapse
|
139
|
Caputi V, Giron MC. Microbiome-Gut-Brain Axis and Toll-Like Receptors in Parkinson's Disease. Int J Mol Sci 2018; 19:ijms19061689. [PMID: 29882798 PMCID: PMC6032048 DOI: 10.3390/ijms19061689] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 05/31/2018] [Accepted: 06/03/2018] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s disease (PD) is a progressively debilitating neurodegenerative disease characterized by α-synucleinopathy, which involves all districts of the brain-gut axis, including the central, autonomic and enteric nervous systems. The highly bidirectional communication between the brain and the gut is markedly influenced by the microbiome through integrated immunological, neuroendocrine and neurological processes. The gut microbiota and its relevant metabolites interact with the host via a series of biochemical and functional inputs, thereby affecting host homeostasis and health. Indeed, a dysregulated microbiota-gut-brain axis in PD might lie at the basis of gastrointestinal dysfunctions which predominantly emerge many years prior to the diagnosis, corroborating the theory that the pathological process is spread from the gut to the brain. Toll-like receptors (TLRs) play a crucial role in innate immunity by recognizing conserved motifs primarily found in microorganisms and a dysregulation in their signaling may be implicated in α-synucleinopathy, such as PD. An overstimulation of the innate immune system due to gut dysbiosis and/or small intestinal bacterial overgrowth, together with higher intestinal barrier permeability, may provoke local and systemic inflammation as well as enteric neuroglial activation, ultimately triggering the development of alpha-synuclein pathology. In this review, we provide the current knowledge regarding the relationship between the microbiota-gut⁻brain axis and TLRs in PD. A better understanding of the dialogue sustained by the microbiota-gut-brain axis and innate immunity via TLR signaling should bring interesting insights in the pathophysiology of PD and provide novel dietary and/or therapeutic measures aimed at shaping the gut microbiota composition, improving the intestinal epithelial barrier function and balancing the innate immune response in PD patients, in order to influence the early phases of the following neurodegenerative cascade.
Collapse
Affiliation(s)
- Valentina Caputi
- Pharmacology Building, Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy.
- APC Microbiome Ireland, University College Cork, T12YT20 Cork, Ireland.
| | - Maria Cecilia Giron
- Pharmacology Building, Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
140
|
Sharma N, Akkoyunlu M, Rabin RL. Macrophages-common culprit in obesity and asthma. Allergy 2018; 73:1196-1205. [PMID: 29178573 DOI: 10.1111/all.13369] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2017] [Indexed: 12/29/2022]
Abstract
Macrophages are essential innate immune cells that also regulate local metabolism. Endogenous or exogenous stimuli may polarize macrophages toward phenotypes that serve distinct innate immunological metabolic functions. IFN-γ or lipopolysaccharide (LPS) polarizes macrophages toward the M1, or "classically activated" phenotype that participates in defense against intracellular pathogens. IL-4, IL-13, or chitin polarizes macrophages toward the M2, or "alternatively activated" phenotype, which defends against multicellular nematodes and fungi. As macrophages polarize in local environments, M1 and M2 macrophages may coexist in different organs and may differentially affect asthma and obesity, two comorbid diseases where polarized macrophages contribute to their pathogenesis. While M1 macrophages are considered beneficial in asthma and contribute to the pathology of obesity, M2 macrophages contribute to the pathology of asthma, but limit metabolic syndrome associated with obesity. Here, we discuss the roles for M1 and M2 macrophages in asthma and obesity, and propose a model by which M1-mediated inflammation in adipose tissue enhances M2-mediated inflammation in the asthmatic lung.
Collapse
Affiliation(s)
- N. Sharma
- Division of Bacterial, Parasitic and Allergenic Products Center for Biologics Evaluation and Research Office of Vaccines Research and Review U.S. Food and Drug Administration Silver Spring MD USA
| | - M. Akkoyunlu
- Division of Bacterial, Parasitic and Allergenic Products Center for Biologics Evaluation and Research Office of Vaccines Research and Review U.S. Food and Drug Administration Silver Spring MD USA
| | - R. L. Rabin
- Division of Bacterial, Parasitic and Allergenic Products Center for Biologics Evaluation and Research Office of Vaccines Research and Review U.S. Food and Drug Administration Silver Spring MD USA
| |
Collapse
|
141
|
Obesity, Inflammation, Toll-Like Receptor 4 and Fatty Acids. Nutrients 2018; 10:nu10040432. [PMID: 29601492 PMCID: PMC5946217 DOI: 10.3390/nu10040432] [Citation(s) in RCA: 423] [Impact Index Per Article: 70.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/24/2018] [Accepted: 03/28/2018] [Indexed: 02/06/2023] Open
Abstract
Obesity leads to an inflammatory condition that is directly involved in the etiology of cardiovascular diseases, type 2 diabetes mellitus, and certain types of cancer. The classic inflammatory response is an acute reaction to infections or to tissue injuries, and it tends to move towards resolution and homeostasis. However, the inflammatory process that was observed in individuals affected by obesity and metabolic syndrome differs from the classical inflammatory response in certain respects. This inflammatory process manifests itself systemically and it is characterized by a chronic low-intensity reaction. The toll-like receptor 4 (TLR4) signaling pathway is acknowledged as one of the main triggers of the obesity-induced inflammatory response. The aim of the present review is to describe the role that is played by the TLR4 signaling pathway in the inflammatory response and its modulation by saturated and omega-3 polyunsaturated fatty acids. Studies indicate that saturated fatty acids can induce inflammation by activating the TLR4 signaling pathway. Conversely, omega-3 polyunsaturated fatty acids, such as eicosapentaenoic acid and docosahexaenoic acid, exert anti-inflammatory actions through the attenuation of the activation of the TLR4 signaling pathway by either lipopolysaccharides or saturated fatty acids.
Collapse
|
142
|
Agustí A, García-Pardo MP, López-Almela I, Campillo I, Maes M, Romaní-Pérez M, Sanz Y. Interplay Between the Gut-Brain Axis, Obesity and Cognitive Function. Front Neurosci 2018; 12:155. [PMID: 29615850 PMCID: PMC5864897 DOI: 10.3389/fnins.2018.00155] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 02/26/2018] [Indexed: 12/12/2022] Open
Abstract
Obesity continues to be one of the major public health problems due to its high prevalence and co-morbidities. Common co-morbidities not only include cardiometabolic disorders but also mood and cognitive disorders. Obese subjects often show deficits in memory, learning and executive functions compared to normal weight subjects. Epidemiological studies also indicate that obesity is associated with a higher risk of developing depression and anxiety, and vice versa. These associations between pathologies that presumably have different etiologies suggest shared pathological mechanisms. Gut microbiota is a mediating factor between the environmental pressures (e.g., diet, lifestyle) and host physiology, and its alteration could partly explain the cross-link between those pathologies. Westernized dietary patterns are known to be a major cause of the obesity epidemic, which also promotes a dysbiotic drift in the gut microbiota; this, in turn, seems to contribute to obesity-related complications. Experimental studies in animal models and, to a lesser extent, in humans suggest that the obesity-associated microbiota may contribute to the endocrine, neurochemical and inflammatory alterations underlying obesity and its comorbidities. These include dysregulation of the HPA-axis with overproduction of glucocorticoids, alterations in levels of neuroactive metabolites (e.g., neurotransmitters, short-chain fatty acids) and activation of a pro-inflammatory milieu that can cause neuro-inflammation. This review updates current knowledge about the role and mode of action of the gut microbiota in the cross-link between energy metabolism, mood and cognitive function.
Collapse
Affiliation(s)
- Ana Agustí
- Microbial Ecology and Nutrition Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Maria P García-Pardo
- Microbial Ecology and Nutrition Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Inmaculada López-Almela
- Microbial Ecology and Nutrition Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Isabel Campillo
- Microbial Ecology and Nutrition Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Marina Romaní-Pérez
- Microbial Ecology and Nutrition Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Yolanda Sanz
- Microbial Ecology and Nutrition Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| |
Collapse
|
143
|
Qiu B, Wang Q, Liu W, Xu TC, Liu LN, Zong AZ, Jia M, Li J, Du FL. Biological effects of trans fatty acids and their possible roles in the lipid rafts in apoptosis regulation. Cell Biol Int 2018; 42:904-912. [PMID: 29500886 DOI: 10.1002/cbin.10958] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 02/24/2018] [Indexed: 12/25/2022]
Abstract
A large number of recent studies are focused on evaluating the mechanism of action of trans fatty acids (TFAs) on the progression of apoptosis. A strong positive association has been reported between TFA and coronary heart disease (CHD), obesity and nonalcoholic steatohepatitis and so on. The present study reviewed the biological effects of trans fatty acids (TFA) and their possible roles in lipid rafts in regulating apoptosis. The following aspects of TFA were included: the research about TFA and diseases affecting serum lipid levels, inducing system inflammation and immune response, and the correlation between TFA and apoptosis. The primary purpose of the review article was to comprehensively evaluate the potential correlation between lipid rafts and apoptosis induced by different structures of TFA and provide some new research progress and future directions about it.
Collapse
Affiliation(s)
- Bin Qiu
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Agro-Products Processing Technology of Shandong Province, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture, 202 Gongye North Road, Jinan, 250100, PR China
| | - Qing Wang
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Agro-Products Processing Technology of Shandong Province, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture, 202 Gongye North Road, Jinan, 250100, PR China
| | - Wei Liu
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Agro-Products Processing Technology of Shandong Province, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture, 202 Gongye North Road, Jinan, 250100, PR China
| | - Tong-Cheng Xu
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Agro-Products Processing Technology of Shandong Province, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture, 202 Gongye North Road, Jinan, 250100, PR China
| | - Li-Na Liu
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Agro-Products Processing Technology of Shandong Province, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture, 202 Gongye North Road, Jinan, 250100, PR China
| | - Ai-Zhen Zong
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Agro-Products Processing Technology of Shandong Province, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture, 202 Gongye North Road, Jinan, 250100, PR China
| | - Min Jia
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Agro-Products Processing Technology of Shandong Province, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture, 202 Gongye North Road, Jinan, 250100, PR China
| | - Jing Li
- State Key Lab of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, 330047, PR China
| | - Fang-Ling Du
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Agro-Products Processing Technology of Shandong Province, 202, Gongye North Road, Jinan, 250100, PR China.,Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture, 202 Gongye North Road, Jinan, 250100, PR China
| |
Collapse
|
144
|
Gianfrancesco MA, Paquot N, Piette J, Legrand-Poels S. Lipid bilayer stress in obesity-linked inflammatory and metabolic disorders. Biochem Pharmacol 2018; 153:168-183. [PMID: 29462590 DOI: 10.1016/j.bcp.2018.02.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/15/2018] [Indexed: 12/13/2022]
Abstract
The maintenance of the characteristic lipid compositions and physicochemical properties of biological membranes is essential for their proper function. Mechanisms allowing to sense and restore membrane homeostasis have been identified in prokaryotes for a long time and more recently in eukaryotes. A membrane remodeling can result from aberrant metabolism as seen in obesity. In this review, we describe how such lipid bilayer stress can account for the modulation of membrane proteins involved in the pathogenesis of obesity-linked inflammatory and metabolic disorders. We address the case of the Toll-like receptor 4 that is implicated in the obesity-related low grade inflammation and insulin resistance. The lipid raft-mediated TLR4 activation is promoted by an enrichment of the plasma membrane with saturated lipids or cholesterol increasing the lipid phase order. We discuss of the plasma membrane Na, K-ATPase that illustrates a new concept according to which direct interactions between specific residues and particular lipids determine both stability and activity of the pump in parallel with indirect effects of the lipid bilayer. The closely related sarco(endo)-plasmic Ca-ATPase embedded in the more fluid ER membrane seems to be more sensitive to a lipid bilayer stress as demonstrated by its inactivation in cholesterol-loaded macrophages or its inhibition mediated by an increased PtdCho/PtdEtn ratio in obese mice hepatocytes. Finally, we describe the model recently proposed for the activation of the conserved IRE-1 protein through alterations in the ER membrane lipid packing and thickness. Such IRE-1 activation could occur in response to abnormal lipid synthesis and membrane remodeling as observed in hepatocytes exposed to excess nutrients. Since the IRE-1/XBP1 branch also stimulates the lipid synthesis, this pathway could create a vicious cycle "lipogenesis-ER lipid bilayer stress-lipogenesis" amplifying hepatic ER pathology and the obesity-linked systemic metabolic defects.
Collapse
Affiliation(s)
- Marco A Gianfrancesco
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium; Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, University Hospital of Liège, Liège, Belgium
| | - Nicolas Paquot
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium; Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, University Hospital of Liège, Liège, Belgium
| | - Jacques Piette
- Laboratory of Virology and Immunology, GIGA-Molecular Biology of Diseases, University of Liège, Liège, Belgium
| | - Sylvie Legrand-Poels
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium; Laboratory of Virology and Immunology, GIGA-Molecular Biology of Diseases, University of Liège, Liège, Belgium.
| |
Collapse
|
145
|
Matsumoto M, Zhang J, Zhang X, Liu J, Jiang JX, Yamaguchi K, Taruno A, Katsuyama M, Iwata K, Ibi M, Cui W, Matsuno K, Marunaka Y, Itoh Y, Torok NJ, Yabe-Nishimura C. The NOX1 isoform of NADPH oxidase is involved in dysfunction of liver sinusoids in nonalcoholic fatty liver disease. Free Radic Biol Med 2018; 115:412-420. [PMID: 29274380 PMCID: PMC5969997 DOI: 10.1016/j.freeradbiomed.2017.12.019] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/13/2017] [Accepted: 12/15/2017] [Indexed: 01/07/2023]
Abstract
The increased production of reactive oxygen species (ROS) has been postulated to play a key role in the progression of nonalcoholic fatty liver disease (NAFLD). However, the source of ROS and mechanisms underlying the development of NAFLD have yet to be established. We observed a significant up-regulation of a minor isoform of NADPH oxidase, NOX1, in the liver of nonalcoholic steatohepatitis (NASH) patients as well as of mice fed a high-fat and high-cholesterol (HFC) diet for 8 weeks. In mice deficient in Nox1 (Nox1KO), increased levels of serum alanine aminotransferase and hepatic cleaved caspase-3 demonstrated in HFC diet-fed wild-type mice (WT) were significantly attenuated. Concomitantly, increased protein nitrotyrosine adducts, a marker of peroxynitrite-induced injury detected in hepatic sinusoids of WT, were significantly suppressed in Nox1KO. The expression of NOX1 mRNA was much higher in the fractions of enriched liver sinusoidal endothelial cells (LSECs) than in those of hepatocytes. In primary cultured LSECs, palmitic acid (PA) up-regulated the mRNA level of NOX1, but not of NOX2 or NOX4. The production of nitric oxide by LSECs was significantly attenuated by PA-treatment in WT but not in Nox1KO. When the in vitro relaxation of TWNT1, a cell line that originated from hepatic stellate cells, was assessed by the gel contraction assay, the relaxation of stellate cells induced by LSECs was attenuated by PA treatment. In contrast, the relaxation effect of LSECs was preserved in cells isolated from Nox1KO. Taken together, the up-regulation of NOX1 in LSECs may elicit peroxynitrite-mediated cellular injury and impaired hepatic microcirculation through the reduced bioavailability of nitric oxide. ROS derived from NOX1 may therefore constitute a critical component in the progression of NAFLD.
Collapse
Affiliation(s)
- Misaki Matsumoto
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Jia Zhang
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Xueqing Zhang
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Junjie Liu
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Joy X Jiang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, UC Davis Medical Center, Sacramento, CA, USA
| | - Kanji Yamaguchi
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akiyuki Taruno
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masato Katsuyama
- Radioisotope Center, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazumi Iwata
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Masakazu Ibi
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Wenhao Cui
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Kuniharu Matsuno
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yoshinori Marunaka
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Natalie J Torok
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, UC Davis Medical Center, Sacramento, CA, USA
| | - Chihiro Yabe-Nishimura
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| |
Collapse
|
146
|
Sobocińska J, Roszczenko-Jasińska P, Ciesielska A, Kwiatkowska K. Protein Palmitoylation and Its Role in Bacterial and Viral Infections. Front Immunol 2018; 8:2003. [PMID: 29403483 PMCID: PMC5780409 DOI: 10.3389/fimmu.2017.02003] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/26/2017] [Indexed: 12/11/2022] Open
Abstract
S-palmitoylation is a reversible, enzymatic posttranslational modification of proteins in which palmitoyl chain is attached to a cysteine residue via a thioester linkage. S-palmitoylation determines the functioning of proteins by affecting their association with membranes, compartmentalization in membrane domains, trafficking, and stability. In this review, we focus on S-palmitoylation of proteins, which are crucial for the interactions of pathogenic bacteria and viruses with the host. We discuss the role of palmitoylated proteins in the invasion of host cells by bacteria and viruses, and those involved in the host responses to the infection. We highlight recent data on protein S-palmitoylation in pathogens and their hosts obtained owing to the development of methods based on click chemistry and acyl-biotin exchange allowing proteomic analysis of protein lipidation. The role of the palmitoyl moiety present in bacterial lipopolysaccharide and lipoproteins, contributing to infectivity and affecting recognition of bacteria by innate immune receptors, is also discussed.
Collapse
Affiliation(s)
- Justyna Sobocińska
- Laboratory of Molecular Membrane Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Paula Roszczenko-Jasińska
- Laboratory of Molecular Membrane Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Anna Ciesielska
- Laboratory of Molecular Membrane Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Kwiatkowska
- Laboratory of Molecular Membrane Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
147
|
Abstract
Central to the understanding of the relationships between diet, gut microbiota, and vitamins D and A in multiple sclerosis is low-grade inflammation, which is involved in all chronic inflammatory diseases and is influenced by each of the above effectors. We show that food components have either proinflammatory or anti-inflammatory effects and influence both the human metabolism (the "metabolome") and the composition of gut microbiota. Hypercaloric, high-animal-fat Western diets favor anabolism and change gut microbiota composition towards dysbiosis. Subsequent intestinal inflammation leads to leakage of the gut barrier, disruption of the blood-brain barrier, and neuroinflammation. Conversely, a vegetarian diet, rich in fiber, is coherent with gut eubiosis and a healthy condition. Vitamin D levels, mainly insufficient in a persistent low-grade inflammatory status, can be restored to optimal values only by administration of high amounts of cholecalciferol. At its optimal values (>30 ng/ml), vitamin D requires vitamin A for the binding to the vitamin D receptor and exert its anti-inflammatory action. Both vitamins must be supplied to the subjects lacking vitamin D. We conclude that nutrients, including the nondigestible dietary fibers, have a leading role in tackling the low-grade inflammation associated with chronic inflammatory diseases. Their action is mediated by gut microbiota and any microbial change induced by diet modifies host-microbe interactions in a consequent way, to improve the disease or worsen it.
Collapse
Affiliation(s)
- Paolo Riccio
- Department of Sciences, University of Basilicata, Viale dell'Ateneo Lucano, 10, 85100, Potenza, Italy.
| | - Rocco Rossano
- Department of Sciences, University of Basilicata, Viale dell'Ateneo Lucano, 10, 85100, Potenza, Italy
| |
Collapse
|
148
|
Wollina U, Wetzker R, Abdel-Naser MB, Kruglikov IL. Role of adipose tissue in facial aging. Clin Interv Aging 2017; 12:2069-2076. [PMID: 29255352 PMCID: PMC5723114 DOI: 10.2147/cia.s151599] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Age-dependent modification of the facial subcutaneous white adipose tissue (sWAT) connected with reduction of its volume, modification of collagen content and adhesion between dermal and adipose layers can significantly influence mechanical stability of the skin and cause the development of aging symptoms such as wrinkles. Typical aging appearance in facial skin is at least partly connected with special phenotypical features of facial preadipocytes and mature adipocytes. In this paper, we have discussed the possible roles of local inflammation, compartmental structure of facial sWAT and trans-differentiation processes such as beiging of white adipocytes and adipocyte-myofibroblast transition in facial skin aging.
Collapse
Affiliation(s)
- Uwe Wollina
- Department of Dermatology and Allergology, Academic Teaching Hospital Dresden-Friedrichstadt, Dresden
| | - Reinhard Wetzker
- Department of Anesthesiology and Intensive Care Medicine, and Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | | | | |
Collapse
|
149
|
Hashemzadeh AA, Nasoohi N, Raygan F, Aghadavod E, Akbari E, Taghizadeh M, Memarzadeh MR, Asemi Z. Flaxseed Oil Supplementation Improve Gene Expression Levels of PPAR-γ, LP(a), IL-1 and TNF-α in Type 2 Diabetic Patients with Coronary Heart Disease. Lipids 2017; 52:907-915. [PMID: 28916975 DOI: 10.1007/s11745-017-4295-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 09/01/2017] [Indexed: 12/16/2022]
Abstract
This study was carried out to determine the effects of flaxseed oil administration on gene expression levels related to insulin, lipid and inflammation in overweight diabetic patients with coronary heart disease (CHD). This randomized double-blind, placebo-controlled trial was conducted among 60 diabetic patients with CHD. Subjects were randomly allocated into two groups to intake either 1000 mg n-3 fatty acid from flaxseed oil containing 400 mg α-Linolenic acid [ALA (18:3n-3)] (n = 30) or placebo (n = 30) twice a day for 12 weeks. Gene expression related to insulin, lipid and inflammation were quantified in peripheral blood mononuclear cells (PBMC) of diabetic patients with CHD with RT-PCR method. Results of RT-PCR demonstrated that after the 12-week intervention, compared with the placebo, flaxseed oil supplementation could up-regulate gene expression of peroxisome proliferator-activated receptor gamma (PPAR-γ) (P = 0.02) in PBMC of diabetic patients with CHD. In addition, compared with the placebo, taking flaxseed oil supplements down-regulated gene expression levels of lipoprotein(a) [LP(a)] (P = 0.001), interleukin-1 (IL-1) (P = 0.001) and tumor necrosis factor alpha (TNF-α) (P = 0.02) in PBMC of diabetic patients with CHD. We did not observe any significant effect of flaxseed oil supplementation on gene expression levels of low-density lipoprotein receptor (LDLR), IL-8 and transforming growth factor beta (TGF-β) in PBMC of diabetic patients with CHD. Overall, flaxseed oil supplementation for 12 weeks in diabetic patients with CHD significantly improved gene expression levels of PPAR-γ, LP(a), IL-1 and TNF-α, but did not influence LDLR, IL-8 and TGF-β.
Collapse
Affiliation(s)
- Ali Akbar Hashemzadeh
- Department of Molecular and Cellular Sciences, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Nikoo Nasoohi
- Department of Molecular and Cellular Sciences, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Fariba Raygan
- Department of Cardiology, School of Medicine, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Esmat Aghadavod
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Elmira Akbari
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | | | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| |
Collapse
|
150
|
Agusti A, Moya-Pérez A, Campillo I, Montserrat-de la Paz S, Cerrudo V, Perez-Villalba A, Sanz Y. Bifidobacterium pseudocatenulatum CECT 7765 Ameliorates Neuroendocrine Alterations Associated with an Exaggerated Stress Response and Anhedonia in Obese Mice. Mol Neurobiol 2017; 55:5337-5352. [PMID: 28921462 DOI: 10.1007/s12035-017-0768-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 09/06/2017] [Indexed: 02/06/2023]
Abstract
Obesity, besides being a problem of metabolic dysfunction, constitutes a risk factor for psychological disorders. Experimental models of diet-induced obesity have revealed that obese animals are prone to anxious and depressive-like behaviors. The present study aimed to evaluate whether Bifidobacterium pseudocatenulatum CECT 7765 could reverse the neurobehavioral consequences of obesity in a high-fat diet (HFD) fed mouse model via regulation of the gut-brain axis. Adult male wild-type C57BL-6 mice were fed a standard diet or HFD, supplemented with either placebo or the bifidobacterial strain for 13 weeks. Behavioral tests were performed, and immune and neuroendocrine parameters were analyzed including leptin and corticosterone and their receptors, Toll-like receptor 2 (TLR2) and neurotransmitters. We found that obese mice showed anhedonia (p < 0.050) indicative of a depressive-like behavior and an exaggerated hypothalamic-pituitary axis (HPA)-mediated stress response to acute physical (p < 0.001) and social stress (p < 0.050), but these alterations were ameliorated by B. pseudocatenulatum CECT 7765 (p < 0.050). These behavioral effects were parallel to reductions of the obesity-associated hyperleptinemia (p < 0.001) and restoration of leptin signaling (p < 0.050), along with fat mass loss (p < 0.010). B. pseudocatenulatum CECT 7765 administration also led to restoration of the obesity-induced reductions in adrenaline in the hypothalamus (p < 0.010), involved in the hypothalamic control of energy balance. Furthermore, the bifidobacterial strain reduced the obesity-induced upregulation of TLR2 protein or gene expression in the intestine (p < 0.010) and the hippocampus (p < 0.050) and restored the alterations of 5-HT levels in the hippocampus (p < 0.050), which could contribute to attenuating the obesity-associated depressive-like behavior (p < 0.050). In summary, the results indicate that B. pseudocatenulatum CECT 7765 could play a role in depressive behavior comorbid with obesity via regulation of endocrine and immune mediators of the gut-brain axis.
Collapse
Affiliation(s)
- Ana Agusti
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), C/ Catedratico Agustin Escardino 7, 46980, Paterna-Valencia, Spain. .,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Cell Biology Department, University of Valencia, Valencia, Spain.
| | - A Moya-Pérez
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), C/ Catedratico Agustin Escardino 7, 46980, Paterna-Valencia, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Cell Biology Department, University of Valencia, Valencia, Spain
| | - I Campillo
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), C/ Catedratico Agustin Escardino 7, 46980, Paterna-Valencia, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Cell Biology Department, University of Valencia, Valencia, Spain
| | - S Montserrat-de la Paz
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), C/ Catedratico Agustin Escardino 7, 46980, Paterna-Valencia, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Cell Biology Department, University of Valencia, Valencia, Spain
| | - V Cerrudo
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), C/ Catedratico Agustin Escardino 7, 46980, Paterna-Valencia, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Cell Biology Department, University of Valencia, Valencia, Spain
| | - A Perez-Villalba
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), C/ Catedratico Agustin Escardino 7, 46980, Paterna-Valencia, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Cell Biology Department, University of Valencia, Valencia, Spain
| | - Yolanda Sanz
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), C/ Catedratico Agustin Escardino 7, 46980, Paterna-Valencia, Spain. .,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Cell Biology Department, University of Valencia, Valencia, Spain.
| |
Collapse
|