151
|
Fouani FZ, Fadaei R, Moradi N, Zandieh Z, Ansaripour S, Yekaninejad MS, Vatannejad A, Mahmoudi M. Circulating levels of Meteorin-like protein in polycystic ovary syndrome: A case-control study. PLoS One 2020; 15:e0231943. [PMID: 32330176 PMCID: PMC7182262 DOI: 10.1371/journal.pone.0231943] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 04/04/2020] [Indexed: 02/06/2023] Open
Abstract
Patients diagnosed with polycystic ovary syndrome (PCOS) are at high risk of developing a myriad of endocrinologic and metabolic derailments. Moreover, PCOS is a leading cause of habitual abortion, also known as recurrent pregnancy loss (RPL). Meteorin-like protein (Metrnl) is a newly discovered adipokine with the potential to counteract the metaflammation. This study aimed at determining the associations of serum Metrnl levels with homocysteine, hs-CRP, and some components of metabolic syndrome in PCOS-RPL and infertile PCOS patients.This case-control study was conducted in 120 PCOS patients (60 PCOS-RPL and 60 infertile) and 60 control. Serum hs-CRP and homocysteine were assessed using commercial kits, while adiponectin, Metrnl, FSH, LH, free testosterone and insulin levels were analyzed using ELISA technique. Serum Metrnl levels were found to be lower in PCOS patients when compared to controls (67.98 ± 26.66 vs. 96.47 ± 28.72 pg/mL, P <0.001)). Furthermore, serum adiponectin levels were lower, while free testosterone, fasting insulin, HOMA-IR, homocysteine, and hs-CRP were significantly higher in PCOS group compared to controls. Moreover, serum Metrnl correlated with BMI, adiponectin, and homocysteine in controls, and inversely correlated with FBG, fasting insulin, and HOMA-IR in PCOS group and subgroups. Besides, it inversely correlated with hs-CRP in control, and PCOS group and subgroups. These findings revealed a possible role of Metrnl in the pathogenesis of PCOS and RPL. Nevertheless, there is a necessity for future studies to prove this concept.
Collapse
Affiliation(s)
- Fatima Zahraa Fouani
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Fadaei
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nariman Moradi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Zandieh
- Cellular and Molecular Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Shahid Akbar Abadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran
| | - Soheila Ansaripour
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mir Saeed Yekaninejad
- Department of Statistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Vatannejad
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Student’s Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mahmoudi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
152
|
Laothaweerungsawat N, Neimkhum W, Anuchapreeda S, Sirithunyalug J, Chaiyana W. Transdermal delivery enhancement of carvacrol from Origanum vulgare L. essential oil by microemulsion. Int J Pharm 2020; 579:119052. [DOI: 10.1016/j.ijpharm.2020.119052] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 12/23/2019] [Accepted: 01/15/2020] [Indexed: 12/15/2022]
|
153
|
Dubois-Deruy E, Rémy G, Alard J, Kervoaze G, Chwastyniak M, Baron M, Beury D, Siegwald L, Caboche S, Hot D, Gosset P, Grangette C, Pinet F, Wolowczuk I, Pichavant M. Modelling the Impact of Chronic Cigarette Smoke Exposure in Obese Mice: Metabolic, Pulmonary, Intestinal, and Cardiac Issues. Nutrients 2020; 12:nu12030827. [PMID: 32244932 PMCID: PMC7175208 DOI: 10.3390/nu12030827] [Citation(s) in RCA: 4] [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: 02/19/2020] [Revised: 03/11/2020] [Accepted: 03/18/2020] [Indexed: 12/15/2022] Open
Abstract
Unhealthy lifestyle choices, such as bad eating behaviors and cigarette smoking, have major detrimental impacts on health. However, the inter-relations between obesity and smoking are still not fully understood. We thus developed an experimental model of high-fat diet-fed obese C57BL/6 male mice chronically exposed to cigarette smoke. Our study evaluated for the first time the resulting effects of the combined exposure to unhealthy diet and cigarette smoke on several metabolic, pulmonary, intestinal, and cardiac parameters. We showed that the chronic exposure to cigarette smoke modified the pattern of body fat distribution in favor of the visceral depots in obese mice, impaired the respiratory function, triggered pulmonary inflammation and emphysema, and was associated with gut microbiota dysbiosis, cardiac hypertrophy and myocardial fibrosis.
Collapse
Affiliation(s)
- Emilie Dubois-Deruy
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000 Lille, France; (E.D.-D.); (M.C.); (F.P.)
| | - Gaëlle Rémy
- University of Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France; (G.R.); (J.A.); (G.K.); (M.B.); (D.B.); (L.S.); (S.C.); (D.H.); (P.G.); (C.G.); (I.W.)
| | - Jeanne Alard
- University of Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France; (G.R.); (J.A.); (G.K.); (M.B.); (D.B.); (L.S.); (S.C.); (D.H.); (P.G.); (C.G.); (I.W.)
| | - Gwenola Kervoaze
- University of Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France; (G.R.); (J.A.); (G.K.); (M.B.); (D.B.); (L.S.); (S.C.); (D.H.); (P.G.); (C.G.); (I.W.)
| | - Maggy Chwastyniak
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000 Lille, France; (E.D.-D.); (M.C.); (F.P.)
| | - Morgane Baron
- University of Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France; (G.R.); (J.A.); (G.K.); (M.B.); (D.B.); (L.S.); (S.C.); (D.H.); (P.G.); (C.G.); (I.W.)
| | - Delphine Beury
- University of Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France; (G.R.); (J.A.); (G.K.); (M.B.); (D.B.); (L.S.); (S.C.); (D.H.); (P.G.); (C.G.); (I.W.)
| | - Léa Siegwald
- University of Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France; (G.R.); (J.A.); (G.K.); (M.B.); (D.B.); (L.S.); (S.C.); (D.H.); (P.G.); (C.G.); (I.W.)
| | - Ségolène Caboche
- University of Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France; (G.R.); (J.A.); (G.K.); (M.B.); (D.B.); (L.S.); (S.C.); (D.H.); (P.G.); (C.G.); (I.W.)
| | - David Hot
- University of Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France; (G.R.); (J.A.); (G.K.); (M.B.); (D.B.); (L.S.); (S.C.); (D.H.); (P.G.); (C.G.); (I.W.)
| | - Philippe Gosset
- University of Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France; (G.R.); (J.A.); (G.K.); (M.B.); (D.B.); (L.S.); (S.C.); (D.H.); (P.G.); (C.G.); (I.W.)
| | - Corinne Grangette
- University of Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France; (G.R.); (J.A.); (G.K.); (M.B.); (D.B.); (L.S.); (S.C.); (D.H.); (P.G.); (C.G.); (I.W.)
| | - Florence Pinet
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000 Lille, France; (E.D.-D.); (M.C.); (F.P.)
| | - Isabelle Wolowczuk
- University of Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France; (G.R.); (J.A.); (G.K.); (M.B.); (D.B.); (L.S.); (S.C.); (D.H.); (P.G.); (C.G.); (I.W.)
| | - Muriel Pichavant
- University of Lille, CNRS UMR9017, Inserm U1019, CHRU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France; (G.R.); (J.A.); (G.K.); (M.B.); (D.B.); (L.S.); (S.C.); (D.H.); (P.G.); (C.G.); (I.W.)
- Correspondence: ; Tel.: +33-320-877-965
| |
Collapse
|
154
|
Faheem SA, Saeed NM, El-Naga RN, Ayoub IM, Azab SS. Hepatoprotective Effect of Cranberry Nutraceutical Extract in Non-alcoholic Fatty Liver Model in Rats: Impact on Insulin Resistance and Nrf-2 Expression. Front Pharmacol 2020; 11:218. [PMID: 32256346 PMCID: PMC7093716 DOI: 10.3389/fphar.2020.00218] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/14/2020] [Indexed: 12/15/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a pathological accumulation of triglycerides (TGs) in the hepatocyte in the absence of alcohol intake. Untreated NAFLD is expected to progress into liver fibrosis. Cranberry is rich in polyphenols with antioxidant and anti-inflammatory activities. Hypothesis The present study was performed to evaluate our hypothesis of the possible anti-fibrotic effect of cranberry nutraceuticals in a high fat cholesterol diet induced (HFCD)-NAFLD in rats, focusing on improving insulin sensitivity and modulating the expression of nuclear factor erythroid-2-related factor-2 (Nrf2) (a transcription factor responsible for regulating cellular redox balance). Method Male albino wistar rats (12 weeks) received HFCD and/or cranberry (50 and 100 mg/kg/day, three times/week) orally for 8 consecutive weeks. Results In comparison to the HFCD group, cranberry treated groups (50 and 100 mg/kg) showed marked hepatoprotection, where it significantly decreased liver enzymes (alanine transaminases by 49 and 64% and aspartate transaminases by 45 and 64%; respectively), TGs, and ameliorated the histopathological alterations (such as inflammatory cells infiltration and ballooning degeneration) induced by HFCD. Cranberry also alleviated oxidative stress (malondialdehyde, glutathione, catalase and superoxide dismutase) and inflammation (tumor necrosis factor- alpha, interleukine-6 and nuclear factor kappa-b) and significantly reduced the HOMA-IR and TyG index. On the other hand, cranberry treated groups (50 and 100 mg/kg) showed a marked increase in the expression of adiponectin, by 8 and 13-fold, insulin receptor substrate-2 by 21 and 79%, and Nrf2 by 13 and 61%, respectively. Notably, cranberry significantly reduced the fibrotic markers, TGF–β and α-SMA expression and collagen deposition. Conclusion The present study showed that cranberry significantly attenuated NAFLD, in a dose dependent manner, which could be partially recognized by its antioxidant, anti-inflammatory activities, and its ability to improve insulin sensitivity. Notably, our study proves for the first time that the anti-fibrotic activity of cranberry is promising.
Collapse
Affiliation(s)
- Safaa A Faheem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Noha M Saeed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Reem N El-Naga
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Iriny M Ayoub
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Samar S Azab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
155
|
Akram F, Gragnoli C, Raheja UK, Snitker S, Lowry CA, Sterns-Yoder KA, Hoisington AJ, Brenner LA, Saunders E, Stiller JW, Ryan KA, Rohan KJ, Mitchell BD, Postolache TT. Seasonal affective disorder and seasonal changes in weight and sleep duration are inversely associated with plasma adiponectin levels. J Psychiatr Res 2020; 122:97-104. [PMID: 31981963 PMCID: PMC7024547 DOI: 10.1016/j.jpsychires.2019.12.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 12/25/2019] [Accepted: 12/30/2019] [Indexed: 12/15/2022]
Abstract
Overlapping pathways between mood and metabolic regulation have increasingly been reported. Although impaired regulation of adiponectin, a major metabolism-regulating hormone, has been implicated in major depressive disorder, its role in seasonal changes in mood and seasonal affective disorder-winter type (SAD), a disorder characterized by onset of mood impairment and metabolic dysregulation (e.g., carbohydrate craving and weight gain) in fall/winter and spontaneous alleviation in spring/summer, has not been previously studied. We studied a convenience sample of 636 Old Order Amish (mean (± SD), 53.6 (±14.8) years; 50.1% males), a population with self-imposed restriction on network electric light at home, and low prevalence of total SAD (t-SAD = syndromal + subsyndromal). We calculated the global seasonality score (GSS), estimated SAD and subsyndromal-SAD after obtaining Seasonal Pattern Assessment Questionnaires (SPAQs), and measured overnight fasting plasma adiponectin levels. We then tested associations between plasma adiponectin levels and GSS, t-SAD, winter-summer difference in self-reported sleep duration, and self-reported seasonal weight change, by using analysis of co-variance (ANCOVA) and linear regression analysis after adjusting for age, gender, and BMI. Participants with t-SAD (N = 14; 2.2%) had significantly lower plasma adiponectin levels (mean ± SEM, 8.76 ± 1.56 μg/mL) than those without t-SAD (mean ± SEM, 11.93 ± 0.22 μg/mL) (p = 0.035). In addition, there was significant negative association between adiponectin levels and winter-summer difference in self-reported sleep duration (p = 0.025) and between adiponectin levels and self-reported seasonal change in weight (p = 0.006). There was no significant association between GSS and adiponectin levels (p = 0.88). To our knowledge, this is the first study testing the association of SAD with adiponectin levels. Replication and extension of our findings longitudinally and, then, interventionally, may implicate low adiponectin as a novel target for therapeutic intervention in SAD.
Collapse
Affiliation(s)
- Faisal Akram
- Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, MD, USA,Saint Elizabeths Hospital, DC Department of Behavioral Health, Washington, DC, USA
| | - Claudia Gragnoli
- Division of Endocrinology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA,Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA,Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, Rome, Italy
| | - Uttam K. Raheja
- Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, MD, USA,Saint Elizabeths Hospital, DC Department of Behavioral Health, Washington, DC, USA
| | - Soren Snitker
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA,Amish Research Clinic of the University of Maryland, Lancaster, PA, USA
| | - Christopher A. Lowry
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO, USA,Department of Physical Medicine & Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA,Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Kelly A. Sterns-Yoder
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO, USA,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA,Department of Physical Medicine & Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew J. Hoisington
- Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA,Department of Systems Engineering, Air Force Institute of Technology, Wright-Patterson AFB, OH, USA
| | - Lisa A. Brenner
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO, USA,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA,Department of Physical Medicine & Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA,Departments of Psychiatry & Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Erika Saunders
- Department of Psychiatry, Penn State University, Hershey, PA, USA
| | - John W. Stiller
- Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, MD, USA,Saint Elizabeths Hospital, DC Department of Behavioral Health, Washington, DC, USA
| | - Kathleen A. Ryan
- Program for Personalized and Genomic Medicine, Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland, School of Medicine, Baltimore, MD, USA,Geriatrics Research and Education Clinical Center, Baltimore, MD, USA,Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Kelly J. Rohan
- Department of Psychological Science, University of Vermont, Burlington, VT, USA
| | - Braxton D. Mitchell
- Program for Personalized and Genomic Medicine, Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland, School of Medicine, Baltimore, MD, USA,Geriatrics Research and Education Clinical Center, Baltimore, MD, USA,Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Teodor T. Postolache
- Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, MD, USA,Saint Elizabeths Hospital, DC Department of Behavioral Health, Washington, DC, USA,Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO, USA,Department of Physical Medicine & Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
156
|
Park JE, Oh SH, Cha YS. Lactobacillus Brevis OPK-3 from Kimchi Prevents Obesity and Modulates the Expression of Adipogenic and Pro-Inflammatory Genes in Adipose Tissue of Diet-Induced Obese Mice. Nutrients 2020; 12:nu12030604. [PMID: 32110872 PMCID: PMC7146405 DOI: 10.3390/nu12030604] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/20/2020] [Accepted: 02/24/2020] [Indexed: 12/15/2022] Open
Abstract
Our previous study reported that lactic acid bacteria (L. brevis OPK-3) isolated from kimchi ameliorated intracellular lipid accumulation in 3T3-L1 adipocyte. The current study explored potential roles of L. brevis OPK-3 (KLAB) on preventing body weight gain and its effect on the inflammatory response of adipose tissue. Male C57BL/6 mice (n = 10) were divided into four groups: normal diet with distilled water (NDC), high-fat diet with distilled water (HDC), high-fat diet with L-ornithine (OTC) or high-fat diet with KLAB. The KLAB supplement resulted in significantly lower body weight, lower epididymal fat tissue mass, and lower serum and hepatic TG levels than the HDC. KLAB supplementation improved serum cytokines, and real-time polymerase chain reaction (PCR) analysis showed significantly lower inflammatory cytokine mRNA levels in epididymal adipose tissue. These results suggest that the administration of KLAB inhibits the induction of inflammation in adipose tissue along with the inhibition of weight gain. Therefore, this study demonstrates the therapeutic and beneficial value of this strain produced during the fermentation of kimchi.
Collapse
Affiliation(s)
- Jung Eun Park
- Department of Food Science and Human Nutrition, Jeonbuk National University and Obesity Research Center, Jeonju, 54896 Jeonbuk, Korea;
| | - Suk-Heung Oh
- Department of Food and Biotechnology, Woosuk University, Samnye-eup, Wanju-gun 55338, Jeonbuk, Korea;
| | - Youn-Soo Cha
- Department of Food Science and Human Nutrition, Jeonbuk National University and Obesity Research Center, Jeonju, 54896 Jeonbuk, Korea;
- Correspondence: ; Tel.: +82-63-270-4258
| |
Collapse
|
157
|
Choi HM, Doss HM, Kim KS. Multifaceted Physiological Roles of Adiponectin in Inflammation and Diseases. Int J Mol Sci 2020; 21:ijms21041219. [PMID: 32059381 PMCID: PMC7072842 DOI: 10.3390/ijms21041219] [Citation(s) in RCA: 262] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 12/28/2022] Open
Abstract
Adiponectin is the richest adipokine in human plasma, and it is mainly secreted from white adipose tissue. Adiponectin circulates in blood as high-molecular, middle-molecular, and low-molecular weight isoforms. Numerous studies have demonstrated its insulin-sensitizing, anti-atherogenic, and anti-inflammatory effects. Additionally, decreased serum levels of adiponectin is associated with chronic inflammation of metabolic disorders including Type 2 diabetes, obesity, and atherosclerosis. However, recent studies showed that adiponectin could have pro-inflammatory roles in patients with autoimmune diseases. In particular, its high serum level was positively associated with inflammation severity and pathological progression in rheumatoid arthritis, chronic kidney disease, and inflammatory bowel disease. Thus, adiponectin seems to have both pro-inflammatory and anti-inflammatory effects. This indirectly indicates that adiponectin has different physiological roles according to an isoform and effector tissue. Knowledge on the specific functions of isoforms would help develop potential anti-inflammatory therapeutics to target specific adiponectin isoforms against metabolic disorders and autoimmune diseases. This review summarizes the current roles of adiponectin in metabolic disorders and autoimmune diseases.
Collapse
Affiliation(s)
- Hyung Muk Choi
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University School of Medicine, Seoul 02447, Korea; (H.M.C.); (H.M.D.)
| | - Hari Madhuri Doss
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University School of Medicine, Seoul 02447, Korea; (H.M.C.); (H.M.D.)
- East-West Bone & Joint Disease Research Institute, Kyung Hee University Hospital at Gangdong, Gandong-gu, Seoul 02447, Korea
| | - Kyoung Soo Kim
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University School of Medicine, Seoul 02447, Korea; (H.M.C.); (H.M.D.)
- East-West Bone & Joint Disease Research Institute, Kyung Hee University Hospital at Gangdong, Gandong-gu, Seoul 02447, Korea
- Correspondence: ; Tel.: +82-2-961-9619
| |
Collapse
|
158
|
Antioxidant and Anti-inflammatory Diagnostic Biomarkers in Multiple Sclerosis: A Machine Learning Study. Mol Neurobiol 2020; 57:2167-2178. [PMID: 31970657 DOI: 10.1007/s12035-019-01856-7] [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: 07/29/2019] [Accepted: 12/12/2019] [Indexed: 12/11/2022]
Abstract
An imbalance of inflammatory/anti-inflammatory and oxidant/antioxidant molecules has been implicated in the demyelination and axonal damage in multiple sclerosis (MS). The current study aimed to evaluate the plasma levels of tumor necrosis factor (TNF)-α, soluble TNF receptor (sTNFR)1, sTNFR2, adiponectin, hydroperoxides, advanced oxidation protein products (AOPP), nitric oxide metabolites, total plasma antioxidant capacity using the total radical-trapping antioxidant parameter (TRAP), sulfhydryl (SH) groups, as well as serum levels of zinc in 174 MS patients and 182 controls. The results show that MS is characterized by lowered levels of zinc, adiponectin, TRAP, and SH groups and increased levels of AOPP. MS was best predicted by a combination of lowered levels of zinc, adiponectin, TRAP, and SH groups yielding an area under the receiver operating characteristic (AUC/ROC) curve of 0.986 (±0.005). The combination of these four antioxidants with sTNFR2 showed an AUC/ROC of 0.997 and TRAP, adiponectin, and zinc are the most important biomarkers for MS diagnosis followed at a distance by sTNFR2. Support vector machine with tenfold validation performed on the four antioxidants showed a training accuracy of 92.9% and a validation accuracy of 90.6%. The results indicate that lowered levels of those four antioxidants are associated with MS and that these antioxidants are more important biomarkers of MS than TNF-α signaling and nitro-oxidative biomarkers. Adiponectin, TRAP, SH groups, zinc, and sTNFR2 play a role in the pathophysiology of MS, and a combination of these biomarkers is useful for predicting MS with high sensitivity, specificity, and accuracy. Drugs that increase the antioxidant capacity may offer novel therapeutic opportunities for MS.
Collapse
|
159
|
Shepard CR. TLR9 in MAFLD and NASH: At the Intersection of Inflammation and Metabolism. Front Endocrinol (Lausanne) 2020; 11:613639. [PMID: 33584545 PMCID: PMC7880160 DOI: 10.3389/fendo.2020.613639] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/10/2020] [Indexed: 12/15/2022] Open
Abstract
Toll-Like Receptor 9 (TLR9) is an ancient receptor integral to the primordial functions of inflammation and metabolism. TLR9 functions to regulate homeostasis in a healthy system under acute stress. The literature supports that overactivation of TLR9 under the chronic stress of obesity is a critical driver of the pathogenesis of NASH and NASH-associated fibrosis. Research has focused on the core contributions of the parenchymal and non-parenchymal cells in the liver, adipose, and gut compartments. TLR9 is activated by endogenous circulating mitochondrial DNA (mtDNA). Chronically elevated circulating levels of mtDNA, caused by the stress of overnutrition, are observed in obesity, metabolic dysfunction-associated fatty liver disease (MAFLD), and NASH. Clinical evidence is supportive of TLR9 overactivation as a driver of disease. The role of TLR9 in metabolism and energy regulation may have an underappreciated contribution in the pathogenesis of NASH. Antagonism of TLR9 in NASH and NASH-associated fibrosis could be an effective therapeutic strategy to target both the inflammatory and metabolic components of such a complex disease.
Collapse
|
160
|
Abstract
Neuroimmunology and immunometabolism are burgeoning topics of study, but the intersection of these two fields is scarcely considered. This interplay is particularly prevalent within adipose tissue, where immune cells and the sympathetic nervous system (SNS) have an important role in metabolic homeostasis and pathology, namely in obesity. In the present Review, we first outline the established reciprocal adipose-SNS relationship comprising the neuroendocrine loop facilitated primarily by adipose tissue-derived leptin and SNS-derived noradrenaline. Next, we review the extensive crosstalk between adipocytes and resident innate immune cells as well as the changes that occur in these secretory and signalling pathways in obesity. Finally, we discuss the effect of SNS adrenergic signalling in immune cells and conclude with exciting new research demonstrating an immutable role for SNS-resident macrophages in modulating SNS-adipose crosstalk. We posit that the latter point constitutes the existence of a new field - neuroimmunometabolism.
Collapse
Affiliation(s)
- Chelsea M Larabee
- Department of Physiology, Anatomy & Genetics, Oxford University, Oxford, UK
| | - Oliver C Neely
- Department of Physiology, Anatomy & Genetics, Oxford University, Oxford, UK
| | - Ana I Domingos
- Department of Physiology, Anatomy & Genetics, Oxford University, Oxford, UK.
- The Howard Hughes Medical Institute (HHMI), New York, NY, USA.
| |
Collapse
|
161
|
Li Y, Jin R, Li L, Hsu HH, You IC, Yoon HJ, Yoon KC. Therapeutic Effect of Topical Adiponectin-Derived Short Peptides Compared with Globular Adiponectin in Experimental Dry Eye and Alkali Burn. J Ocul Pharmacol Ther 2019; 36:88-96. [PMID: 31661350 DOI: 10.1089/jop.2018.0131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose: To evaluate the efficacy of adiponectin (APN)-derived short peptides (ADPs) 355 compared with globular APN in a mouse model of experimental dry eye (EDE) and corneal alkali burn. Methods: EDE and chemical burn were induced in C57BL/6 mice by desiccating stress and application of NaOH, respectively. Eye drops consisting of 0.01% globular APN, 0.01% ADPs, 0.1% ADPs, or balanced salt solution (BSS) were applied. Tear volume, tear film break-up time, and corneal staining scores were measured. Concentrations of interleukin (IL)-1β, interferon (IFN)-γ, IL-6, CXCL-9, and CXCL-10 using multiplex immunobead assay were evaluated, and flow cytometry were performed. Corneal epithelial defects and haze degree were analyzed, and enzyme-linked immunosorbent assay for IL-1β and transforming growth factor (TGF)-β levels were observed. Results: All treatment groups showed an improvement in clinical parameters and CD4+CCR5+ T cell and CD11b+ cell infiltrations in the conjunctiva (all P < 0.05). Both ADPs groups had significantly decreased concentrations of IL-1β, IFN-γ, IL-6, CXCL-9, and CXCL-10 in the conjunctiva than the EDE or BSS group. Significantly improved parameters of epithelial defect, degree of haze, and concentrations of IL-1β and TGF-β were observed in all treatment groups. However, no significant differences were noted in clinical or experimental parameters among treatment groups. Conclusion: Topical ADPs could effectively improve clinical signs and inflammation of ocular surface in the EDE or alkali burn, and its efficacy and potency were similar to those of globular APN.
Collapse
Affiliation(s)
- Ying Li
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| | - Rujun Jin
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| | - Lan Li
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| | - Henry H Hsu
- Allysta Pharmaceuticals, Belmont, California
| | - In Cheon You
- Department of Ophthalmology, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Hyeon Jeong Yoon
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| | - Kyung Chul Yoon
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| |
Collapse
|
162
|
Coutinho CAAC, Marson FAL, Ribeiro JD, Bertuzzo CS. A negative screening of rare genetic variants in the ADIPOQ and STATH genes in cystic fibrosis. Pulmonology 2019; 26:138-144. [PMID: 31606405 DOI: 10.1016/j.pulmoe.2019.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/20/2019] [Accepted: 09/02/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The phenotypic variability in cystic fibrosis (CF) is widely recognized and modulated by environmental and genetic factors, including CFTR pathogenic variants and modifier genes genetic variants. In this context, determining the presence of variants in genes involved in immune response may allow a better understanding of CF variability, mainly in lung disease. Thus, ADIPOQ and STATH genes were selected and the analysis of exons and exon/intron junctions was performed for the determination of variations in its sequence, to determine the possible genetic modulation. METHODS A total of 49 patients with CF, diagnosed for showing abnormal [chloride] levels in the sweat test, and identification of two pathogenic variants in CFTR categorized as class I and II were included. Genetic sequencing was performed for the identification of variants in the modifier genes. RESULTS In our analysis, there was absence of rare genetic variants in STATH and ADIPOQ genes associated with the clinical variability. Thus, we are not able to establish an association between the disease severity and rare genetic variants in STATH and ADIPOQ genes, considering exons and exon/intron junctions. CONCLUSIONS Considering the negative screening for rare genetic variants in ADIPOQ and STATH genes, it may be concluded that these genes are not associated with phenotypic modulation of CF in our population. To understand the modifier genes and its action at CF variability it is essential to promote a better overview of the disease. Also, negative reports can help to direct new studies without the use of unnecessary financial support.
Collapse
Affiliation(s)
- C A A C Coutinho
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas, 13081-970, P.O. Box: 6111, Campinas, SP, Brazil.
| | - F A L Marson
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas, 13081-970, P.O. Box: 6111, Campinas, SP, Brazil; Department of Pediatrics, School of Medical Sciences, University of Campinas, 13081-970, P.O. Box: 6111, Campinas, SP, Brazil; Post Graduate Program in Health Science, São Francisco University, Avenida São Francisco de Assis, 218, Jardim São José, Bragança Paulista, São Paulo 12916-900, Brazil.
| | - J D Ribeiro
- Department of Pediatrics, School of Medical Sciences, University of Campinas, 13081-970, P.O. Box: 6111, Campinas, SP, Brazil.
| | - C S Bertuzzo
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas, 13081-970, P.O. Box: 6111, Campinas, SP, Brazil.
| |
Collapse
|
163
|
Kuo IC, Wu PH, Lin HYH, Niu SW, Huang JC, Hung CC, Chiu YW, Chen HC. The association of adiponectin with metabolic syndrome and clinical outcome in patients with non-diabetic chronic kidney disease. PLoS One 2019; 14:e0220158. [PMID: 31323071 PMCID: PMC6641197 DOI: 10.1371/journal.pone.0220158] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 07/09/2019] [Indexed: 02/07/2023] Open
Abstract
Adiponectin is the most abundant circulating adipokine, and it has insulin-sensitizing and anti-inflammatory properties. Although it has been speculated that kidney function decline associated with elevated adiponectin is attributable to decreased renal clearance and compensatory responses to adiponectin resistance, it is unclear how elevated adiponectin affects clinical outcomes in chronic kidney disease (CKD) patients and whether the effects are the same as those in the general population. Therefore, the aim of this study is to examine whether the association between serum adiponectin levels and clinical outcomes in non-diabetic CKD patients is independent of adiposity and metabolic syndrome. We enrolled 196 non-diabetic CKD patients with eGFR ranging between 10 and 60 mL/min/1.73 m2, these patients were divided into two groups based on the presence of metabolic syndrome. The primary endpoint was all-cause mortality or renal events (renal failure requiring renal replacement therapy [RRT] or 50% reduction in eGFR). During the mean follow-up period of 5 years, 48 (24.5%) incident cases of end-stage renal disease (ESRD) were observed, and 33 (16.8%) deaths occurred. The mean eGFR was 29.8 ± 12.8 mL/min/1.73m2. The baseline median adiponectin concentration in the cohort was 29.4(interquartile range, 13.3-108.7) μg/ml. Adiponectin levels were inversely related to body mass index (BMI) (r = -0.29; P < 0.001) and waist circumference (r = -0.35; P < 0.001). In the fully adjusted Cox regression model, the hazard ratios (HRs) were 2.08 (95% confidence interval [CI], 1.08-4.02; P = 0.03) for RRT and 1.66 (95% CI, 1.03-2.65; P = 0.04) for composite renal outcome. The risks remained consistent within different subgroups. However, no association was observed with mortality risk. In conclusion, higher adiponectin levels are associated with a higher risk of ESRD independent of conventional risk factors, BMI, and metabolic syndrome components.
Collapse
Affiliation(s)
- I-Ching Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ping-Hsun Wu
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hugo You-Hsien Lin
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Sheng-Wen Niu
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jiun-Chi Huang
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chi-Chih Hung
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- * E-mail: (CCH); (YWC)
| | - Yi-Wen Chiu
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- * E-mail: (CCH); (YWC)
| | - Hung-Chun Chen
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
164
|
Żelechowska P, Brzezińska-Błaszczyk E, Wiktorska M, Różalska S, Wawrocki S, Kozłowska E, Agier J. Adipocytokines leptin and adiponectin function as mast cell activity modulators. Immunology 2019; 158:3-18. [PMID: 31220342 DOI: 10.1111/imm.13090] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/10/2019] [Accepted: 05/23/2019] [Indexed: 12/25/2022] Open
Abstract
A growing body of data indicates that adipocytokines, including leptin and adiponectin, are critical components not only of metabolic regulation but also of the immune system, mainly by influencing the activity of cells participating in immunological and inflammatory processes. As mast cells (MCs) are the key players in the course of those mechanisms, this study aimed to evaluate the impact of leptin and adiponectin on some aspects of MC activity. We documented that in vivo differentiated mature tissue MCs from the rat peritoneal cavity express a receptor for leptin (OB-R), as well as receptors for adiponectin (AdipoR1 and AdipoR2). We established that leptin, but not adiponectin, stimulates MCs to release of histamine as well as to generation of cysteinyl leukotrienes (cysLTs) and chemokine CCL2. We also found that both adipocytokines affect mRNA expression of various cytokines/chemokines. Leptin and adiponectin also activate MCs to produce reactive oxygen species. Moreover, we documented that leptin significantly augments the surface expression of receptors for cysLTs, i.e. CYSLTR1, CYSLTR2, and GPR17 on MCs, while adiponectin increases only GPR17 expression, and decreases CYSLTR2. Finally, we showed that both adipocytokines serve as potent chemoattractants for MCs. In intracellular signaling in MCs activated by leptin Janus-activated kinase 2, phospholipase C, phosphatidylinositol 3-kinase (PI3K), extracellular signal-regulated kinase (ERK1/2), and p38 molecules play a part whereas the adiponectin-induced activity of MCs is mediated through PI3K, p38, and ERK1/2 pathways. Our observations that leptin and adiponectin regulate MC activity might indicate that adipocytokines modulate the different processes in which MCs are involved.
Collapse
Affiliation(s)
- Paulina Żelechowska
- Department of Experimental Immunology, Faculty of Health Sciences, Medical University of Lodz, Lodz, Poland
| | - Ewa Brzezińska-Błaszczyk
- Department of Experimental Immunology, Faculty of Health Sciences, Medical University of Lodz, Lodz, Poland
| | - Magdalena Wiktorska
- Department of Molecular Cell Mechanisms, Faculty of Health Sciences, Medical University of Lodz, Lodz, Poland
| | - Sylwia Różalska
- Department of Industrial Microbiology and Biotechnology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Sebastian Wawrocki
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Elżbieta Kozłowska
- Department of Experimental Immunology, Faculty of Health Sciences, Medical University of Lodz, Lodz, Poland
| | - Justyna Agier
- Department of Experimental Immunology, Faculty of Health Sciences, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
165
|
Namgoong S, Lee H, Lee JS, Jeong SH, Han SK, Dhong ES. Comparative Biological Effects of Human Amnion and Chorion Membrane Extracts on Human Adipose-Derived Stromal Cells. J Craniofac Surg 2019; 30:947-954. [PMID: 30817541 DOI: 10.1097/scs.0000000000005393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Although therapies with human amnion/chorion are used to ameliorate acute and chronic wounds, it is unclear which component of the amnion/chorion tissue promotes wound healing. To characterize the comparative effects of amnion and chorion in wound healing, we used human adipose-derived stromal cells to assess cell viability, migration, and gel contraction after treatment with amnion membrane extract (AME) or chorion membrane extract (CME). We then correlated the possible effectors via AME and CME protein profiling, and compared them by enzyme-linked immunosorbent assay (ELISA), western blotting, and immunocytochemistry. Cell viability was significantly increased with 50 and 100 μg/mL AME treatment, but with CME treatment, a significant increase was only observed with 100 μg/mL. With CME treatment, cell migration was 2.22-fold greater than the control, and collagen gels showed 20% greater contraction. Compared to control, the expression levels of α-smooth muscle actin (SMA) and smooth muscle protein 22-alpha (SM22α) increased both with AME and CME treatments, whereas calponin expression decreased. Protein profiling revealed significantly higher tissue inhibitor of metalloproteinase-1 (TIMP-1), interleukin-8, exotoxin, and adiponectin levels in CME than in AME, and ELISA revealed 8-fold higher adiponectin levels in cells treated with CME than those treated with AME. Immunocytochemistry revealed that α-SMA, SM22α, and calponin were significantly higher in CME- than AME-treated cells; however, adiponectin treatment did not enhance α-SMA, SM22α, or calponin expression. In conclusion, amnion and chorion membrane extracts exerted differential effects on proliferation and contraction of human adipose-derived stromal cells. Amnion extract was superior at inducing cell proliferation and migration, whereas CME was superior at inducing cell contraction.
Collapse
Affiliation(s)
- Sik Namgoong
- Department of Plastic Surgery, Korea University Guro Hospital, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
166
|
Transcriptomic profile of anterior pituitary cells of pigs is affected by adiponectin. Anim Reprod Sci 2019; 206:17-26. [DOI: 10.1016/j.anireprosci.2019.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 03/14/2019] [Accepted: 05/03/2019] [Indexed: 12/15/2022]
|
167
|
Cox AR, Chernis N, Masschelin PM, Hartig SM. Immune Cells Gate White Adipose Tissue Expansion. Endocrinology 2019; 160:1645-1658. [PMID: 31107528 PMCID: PMC6591013 DOI: 10.1210/en.2019-00266] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/14/2019] [Indexed: 12/15/2022]
Abstract
The immune system plays a critical role in white adipose tissue (WAT) energy homeostasis and, by extension, whole-body metabolism. Substantial evidence from mouse and human studies firmly establishes that insulin sensitivity deteriorates as a result of subclinical inflammation in the adipose tissue of individuals with diabetes. However, the relationship between adipose tissue expandability and immune cell infiltration remains a complex problem important for understanding the pathogenesis of obesity. Notably, a large body of work challenges the idea that all immune responses are deleterious to WAT function. This review highlights recent advances that describe how immune cells and adipocytes coordinately enable WAT expansion and regulation of energy homeostasis.
Collapse
Affiliation(s)
- Aaron R Cox
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Natasha Chernis
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Peter M Masschelin
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Sean M Hartig
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Correspondence: Sean M. Hartig, PhD, Baylor College of Medicine, One Baylor Plaza, BCM185, Houston, Texas 77030. E-mail:
| |
Collapse
|
168
|
Metabolic Obesity Phenotypes and Risk of Cellulitis: A Cohort Study. J Clin Med 2019; 8:jcm8070953. [PMID: 31262086 PMCID: PMC6679047 DOI: 10.3390/jcm8070953] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/15/2019] [Accepted: 06/24/2019] [Indexed: 12/15/2022] Open
Abstract
No cohort studies have evaluated the effect of obesity on the risk of cellulitis according to metabolic health status. We investigated an association of BMI and metabolic health status with the development of cellulitis. We conducted a cohort study of 171,322 Korean adults who underwent a health checkup examination and were followed from 2011 to 2016 for cellulitis and hospital admission related to cellulitis, which were ascertained through the linkage to the Health Insurance and Review Agency database. Being metabolically healthy was defined as not having any metabolic syndrome component and having a homeostasis model assessment of insulin resistance <2.5. During 638,240.4 person-years of follow-up, 14,672 cases of incident cellulitis were identified with 225 cases of cellulitis-related admission. After adjustment for possible confounders, the multivariable-adjusted hazard ratios (95% CI) for incident cellulitis comparing BMIs 23–24.9, 25–29.9, and ≥30 with a BMI of 18.5–22.9 kg/m2 as the reference were 1.07 (1.02–1.11), 1.09 (1.04-1.13), and 1.19 (1.08–1.31), respectively, whereas the corresponding multivariable-adjusted hazard ratios (95% CI) for cellulitis-related admission were 1.55 (1.05–2.3), 2.47 (1.73–3.53), and 4.8 (2.86–8.05), respectively. These associations were consistently observed in both metabolically healthy and unhealthy individuals with no significant interaction. In a large cohort of apparently healthy adults, increased BMI was associated with an increased risk of cellulitis and hospitalization for cellulitis in both metabolically healthy and unhealthy individuals. Obesity appears to be an independent risk factor for cellulitis regardless of metabolic phenotype.
Collapse
|
169
|
Abstract
PURPOSE OF REVIEW Obesity is a state of chronic inflammation. This review aims to summarize recent data supporting the role of the intestinal mucosal barrier and the microbiome in causing adipose tissue inflammation as well as metabolic factors that can affect the intestinal barrier. RECENT FINDINGS Obesity and its metabolic consequences, such as diabetes mellitus, are associated with disruption of the intestinal barrier function. Intestinal microbiota and diet play a key role in the maintenance of a healthy intestinal epithelium. Intestinal barrier dysfunction can lead to heightened inflammation, which in turn can further damage the intestinal barrier through the disruption of tight junction proteins. Intestinal barrier breakdown is associated with adipose tissue inflammation in different disease states, such as obesity, diabetes mellitus, HIV, and inflammatory bowel disease. Future therapeutic strategies to ameliorate intestinal barrier function may help reduce inflammation in obesity and other chronic conditions of increased intestinal permeability.
Collapse
Affiliation(s)
- Lediya Cheru
- Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, LON-207, Boston, MA, 02114, USA
| | - Charles F Saylor
- Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, LON-207, Boston, MA, 02114, USA
| | - Janet Lo
- Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, LON-207, Boston, MA, 02114, USA.
| |
Collapse
|
170
|
Agra-Bermejo RM, Gonzalez-Ferreiro R, Lopez-Canoa JN, Varela-Roman A, Gomez-Otero I, Eiras S, González-Juanatey JR. Long-Term Weight Gain Associated With High Omentin Levels at Hospital Discharge Improves Prognosis of Patients Following Acute Heart Failure. J Cardiovasc Transl Res 2019; 12:231-239. [PMID: 30353296 DOI: 10.1007/s12265-018-9840-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 10/10/2018] [Indexed: 12/13/2022]
Abstract
A obesity paradox has been described following heart failure (HF). The aim of this study was to analyze the association between food intake-involved adipokines and long-term weight changes. Leptin, adiponectin, and omentin were analyzed in 92 acute HF (AHF) patients at discharge, classified on the basis of weight gains or losses > 6%. The mean follow-up was 256 ± 143 days. Leptin and adiponectin levels were similar among weight groups. However, omentin levels were higher in those patients who had gained weight (16 ± 5 ng/mL) than in those who had lost weight (12 ± 4 ng/mL) or showed no weight change (11 ± 5 ng/mL; p < 0.002). Omentin levels were the best independent predictors for patients with weight gain, who had less mortality and hospital readmission during the follow-up. The association between omentin levels and weight gain might explain part of the obesity paradox in HF.
Collapse
Affiliation(s)
- Rosa M Agra-Bermejo
- Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
- CIBERCV: Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | - Rocio Gonzalez-Ferreiro
- Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
- CIBERCV: Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | - J Nicolos Lopez-Canoa
- Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
- CIBERCV: Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | - Alfonso Varela-Roman
- Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
- CIBERCV: Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | - Ines Gomez-Otero
- Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
- CIBERCV: Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | - Sonia Eiras
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain.
- CIBERCV: Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain.
- Complejo Hospitalario Universitario de Santiago de Compostela, Laboratorio 6. IDIS. Planta-2, C/Choupana s/n, 15706, Santiago de Compostela, Spain.
| | - José R González-Juanatey
- Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Cardiology Group, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
- CIBERCV: Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| |
Collapse
|
171
|
Chan SMH, Selemidis S, Bozinovski S, Vlahos R. Pathobiological mechanisms underlying metabolic syndrome (MetS) in chronic obstructive pulmonary disease (COPD): clinical significance and therapeutic strategies. Pharmacol Ther 2019; 198:160-188. [PMID: 30822464 PMCID: PMC7112632 DOI: 10.1016/j.pharmthera.2019.02.013] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a major incurable global health burden and is currently the 4th largest cause of death in the world. Importantly, much of the disease burden and health care utilisation in COPD is associated with the management of its comorbidities (e.g. skeletal muscle wasting, ischemic heart disease, cognitive dysfunction) and infective viral and bacterial acute exacerbations (AECOPD). Current pharmacological treatments for COPD are relatively ineffective and the development of effective therapies has been severely hampered by the lack of understanding of the mechanisms and mediators underlying COPD. Since comorbidities have a tremendous impact on the prognosis and severity of COPD, the 2015 American Thoracic Society/European Respiratory Society (ATS/ERS) Research Statement on COPD urgently called for studies to elucidate the pathobiological mechanisms linking COPD to its comorbidities. It is now emerging that up to 50% of COPD patients have metabolic syndrome (MetS) as a comorbidity. It is currently not clear whether metabolic syndrome is an independent co-existing condition or a direct consequence of the progressive lung pathology in COPD patients. As MetS has important clinical implications on COPD outcomes, identification of disease mechanisms linking COPD to MetS is the key to effective therapy. In this comprehensive review, we discuss the potential mechanisms linking MetS to COPD and hence plausible therapeutic strategies to treat this debilitating comorbidity of COPD.
Collapse
Affiliation(s)
- Stanley M H Chan
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Ross Vlahos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia.
| |
Collapse
|
172
|
Adiponectin, Obesity, and Cancer: Clash of the Bigwigs in Health and Disease. Int J Mol Sci 2019; 20:ijms20102519. [PMID: 31121868 PMCID: PMC6566909 DOI: 10.3390/ijms20102519] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 02/07/2023] Open
Abstract
Adiponectin is one of the most important adipocytokines secreted by adipocytes and is called a “guardian angel adipocytokine” owing to its unique biological functions. Adiponectin inversely correlates with body fat mass and visceral adiposity. Identified independently by four different research groups, adiponectin has multiple names; Acrp30, apM1, GBP28, and AdipoQ. Adiponectin mediates its biological functions via three known receptors, AdipoR1, AdipoR2, and T-cadherin, which are distributed throughout the body. Biological functions of adiponectin are multifold ranging from anti-diabetic, anti-atherogenic, anti-inflammatory to anti-cancer. Lower adiponectin levels have been associated with metabolic syndrome, type 2 diabetes, insulin resistance, cardiovascular diseases, and hypertension. A plethora of experimental evidence supports the role of obesity and increased adiposity in multiple cancers including breast, liver, pancreatic, prostrate, ovarian, and colorectal cancers. Obesity mediates its effect on cancer progression via dysregulation of adipocytokines including increased production of oncogenic adipokine leptin along with decreased production of adiponectin. Multiple studies have shown the protective role of adiponectin in obesity-associated diseases and cancer. Adiponectin modulates multiple signaling pathways to exert its physiological and protective functions. Many studies over the years have shown the beneficial effect of adiponectin in cancer regression and put forth various innovative ways to increase adiponectin levels.
Collapse
|
173
|
Dermal White Adipose Tissue: A Newly Recognized Layer of Skin Innate Defense. J Invest Dermatol 2019; 139:1002-1009. [DOI: 10.1016/j.jid.2018.12.031] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/27/2018] [Accepted: 12/03/2018] [Indexed: 12/14/2022]
|
174
|
Abstract
The prevalence of food allergy is raising in industrialized countries, but the mechanisms behind this increased incidence are not fully understood. Environmental factors are believed to play a role in allergic diseases, including lifestyle influences, such as diet. There is a close relationship between allergens and lipids, with many allergenic proteins having the ability to bind lipids. Dietary lipids exert pro-inflammatory or anti-inflammatory functions on cells of the innate immunity and influence antigen presentation to cells of the adaptive immunity. In addition to modifying the immunostimulating properties of proteins, lipids also alter their digestibility and intestinal absorption, changing allergen bioavailability. This study provides an overview of the role of dietary lipids in food allergy, taking into account epidemiological information, as well as results of mechanistic investigations using in vivo, ex vivo and in vitro models. The emerging link among high-fat diets, obesity, and allergy is also discussed.
Collapse
Affiliation(s)
- Rosina López-Fandiño
- Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Madrid, Spain
| |
Collapse
|
175
|
Silva AR, Gonçalves-de-Albuquerque CF, Pérez AR, Carvalho VDF. Immune-endocrine interactions related to a high risk of infections in chronic metabolic diseases: The role of PPAR gamma. Eur J Pharmacol 2019; 854:272-281. [PMID: 30974105 DOI: 10.1016/j.ejphar.2019.04.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 03/11/2019] [Accepted: 04/02/2019] [Indexed: 02/06/2023]
Abstract
Diverse disturbances in immune-endocrine circuitries are involved in the development and aggravation of several chronic metabolic diseases (CMDs), including obesity, diabetes, and metabolic syndrome. The chronic inflammatory syndrome observed in CMDs culminates in dysregulated immune responses with low microbial killing efficiency, by means low host innate immune response, and loss of ability to eliminate the pathogens, which results in a high prevalence of infectious diseases, including pneumonia, tuberculosis, and sepsis. Herein, we review evidence pointing out PPARγ as a putative player in immune-endocrine disturbances related to increased risk of infections in CMDs. Cumulated evidence indicates that PPARγ activation modulates host cells to control inflammation during CMDs because of PPARγ agonists have anti-inflammatory and pro-resolutive properties, increasing host ability to eliminate pathogen, modulating hormone production, and restoring glucose and lipid homeostasis. As such, we propose PPARγ as a putative therapeutic adjuvant for patients with CMDs to favor a better infection control.
Collapse
Affiliation(s)
- Adriana Ribeiro Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil, 4365, Rio de Janeiro, RJ, Brazil.
| | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil, 4365, Rio de Janeiro, RJ, Brazil; Laboratório de Imunofarmacologia, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Unirio, Brazil.
| | - Ana Rosa Pérez
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER-CONICET UNR), 2000, Rosario, Argentina.
| | - Vinicius de Frias Carvalho
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil, 4365, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
176
|
Beneficial Effects of Adiponectin on Glucose and Lipid Metabolism and Atherosclerotic Progression: Mechanisms and Perspectives. Int J Mol Sci 2019; 20:ijms20051190. [PMID: 30857216 PMCID: PMC6429491 DOI: 10.3390/ijms20051190] [Citation(s) in RCA: 294] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/01/2019] [Accepted: 03/05/2019] [Indexed: 12/14/2022] Open
Abstract
Circulating adiponectin concentrations are reduced in obese individuals, and this reduction has been proposed to have a crucial role in the pathogenesis of atherosclerosis and cardiovascular diseases associated with obesity and the metabolic syndrome. We focus on the effects of adiponectin on glucose and lipid metabolism and on the molecular anti-atherosclerotic properties of adiponectin and also discuss the factors that increase the circulating levels of adiponectin. Adiponectin reduces inflammatory cytokines and oxidative stress, which leads to an improvement of insulin resistance. Adiponectin-induced improvement of insulin resistance and adiponectin itself reduce hepatic glucose production and increase the utilization of glucose and fatty acids by skeletal muscles, lowering blood glucose levels. Adiponectin has also β cell protective effects and may prevent the development of diabetes. Adiponectin concentration has been found to be correlated with lipoprotein metabolism; especially, it is associated with the metabolism of high-density lipoprotein (HDL) and triglyceride (TG). Adiponectin appears to increase HDL and decrease TG. Adiponectin increases ATP-binding cassette transporter A1 and lipoprotein lipase (LPL) and decreases hepatic lipase, which may elevate HDL. Increased LPL mass/activity and very low density lipoprotein (VLDL) receptor and reduced apo-CIII may increase VLDL catabolism and result in the reduction of serum TG. Further, adiponectin has various molecular anti-atherosclerotic properties, such as reduction of scavenger receptors in macrophages and increase of cholesterol efflux. These findings suggest that high levels of circulating adiponectin can protect against atherosclerosis. Weight loss, exercise, nutritional factors, anti-diabetic drugs, lipid-lowering drugs, and anti-hypertensive drugs have been associated with an increase of serum adiponectin level.
Collapse
|
177
|
Tabari FS, Karimian A, Parsian H, Rameshknia V, Mahmoodpour A, Majidinia M, Maniati M, Yousefi B. The roles of FGF21 in atherosclerosis pathogenesis. Rev Endocr Metab Disord 2019; 20:103-114. [PMID: 30879171 DOI: 10.1007/s11154-019-09488-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
FGF21 is a peptide hormone that regulates homeostasis of lipid and glucose as well as energy metabolism. It is mainly expressed and secreted in liver and adipose tissues, and it is expressed in lower amounts in the aorta. Recent clinical and preclinical studies indicate increased serum FGF21 levels in atherosclerosis patients. Also, FGF21 therapy has been reported to reduce the initiation and progression of atherosclerosis in animal models and in vitro studies. Moreover, growing evidence indicates that administration of exogenous FGF21 induces anti-atherosclerotic effects, because of its ability to reduce lipid profile, alleviation of oxidative stress, inflammation, and apoptosis. Therefore, FGF21 can not only be considered as a biomarker for predicting atherosclerosis, but also induce protective effects against atherosclerosis. Besides, serum levels of FGF21 increase in various diseases including in diabetes mellitus, hypertension, and obesity, which may be related to initiating and exacerbating atherosclerosis. On the other hand, FGF21 therapy significantly improves lipid profiles, and reduces vascular inflammation and oxidative stress in atherosclerosis related diseases. Therefore, further prospective studies are needed to clarify whether FGF21 can be used as a prognostic biomarker to identify individuals at future risk of atherosclerosis in these atherosclerosis-associated diseases. In this review, we will discuss the possible mechanism by which FGF21 protects against atherosclerosis.
Collapse
Affiliation(s)
- Farzane Shanebandpour Tabari
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Ansar Karimian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Hadi Parsian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Vahid Rameshknia
- Faculty of Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
- Department of Biochemistry, Baku State University, Baku, Azerbaijan
| | - Ata Mahmoodpour
- Anesthesiology Research Team, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Mahmood Maniati
- Faculty of Medicine, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bahman Yousefi
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran.
| |
Collapse
|
178
|
Rizzetti DA, Corrales P, Piagette JT, Uranga-Ocio JA, Medina-Gomez G, Peçanha FM, Vassallo DV, Miguel M, Wiggers GA. Chronic mercury at low doses impairs white adipose tissue plasticity. Toxicology 2019; 418:41-50. [PMID: 30807803 DOI: 10.1016/j.tox.2019.02.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/31/2019] [Accepted: 02/23/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The toxic effects of mercury (Hg) are involved in homeostasis of energy systems such as lipid and glucose metabolism, and white adipose tissue dysfunction is considered as a central mechanism leading to metabolic disorders. OBJECTIVE The aim of this study was to determine the effects of chronic inorganic Hg exposure at low doses on the lipid and glycemic metabolism. METHODS Male Wistar rats were divided into two groups and treated for 60 days with: saline solution, i.m. (Untreated) and mercury chloride, i.m. - 1st dose 4.6 μg/kg, subsequent doses 0.07 μg/kg/day - (Mercury). Histological analyses, Hg levels measurement and GRP78, CHOP, PPARα, PPARγ, leptin, adiponectin and CD11 mRNA expressions were performed in epididymal white adipose tissue (eWAT). Glucose, triglycerides, total cholesterol and insulin plasma levels were also measured. RESULTS Hg exposure reduced the absolute and relative eWAT weights, adipocyte size, plasma insulin levels, glucose tolerance, antioxidant defenses and increased plasma glucose and triglyceride levels. In addition, CHOP, GRP78, PPARα, PPARγ, leptin and adiponectin mRNA expressions were increased in Hg-treated animals. No differences in Hg concentration were found in eWAT between the untreated and Hg groups. These results suggest that the reduction in adipocyte size is related to the impaired antioxidant defenses, endoplasmic reticulum (ER) stress, the disrupted PPARs and adipokines mRNA expression induced by the metal in eWAT. These disturbances possibly induced a decrease in circulating insulin levels, an imbalance between lipolysis and lipogenesis mechanisms in eWAT, with an increase in fatty acids mobilization, a reduction in glucose uptake and an activation of pro-apoptotic pathways, leading to hyperglycemia and hyperlipidemia. CONCLUSIONS Hg is a powerful environmental WAT disruptor that influences signaling events and impairs metabolic activity and hormonal balance of adipocytes.
Collapse
Affiliation(s)
- Danize Aparecida Rizzetti
- Cardiovascular Physiology Laboratory, Universidade Federal do Pampa, BR 472, Km 592, Uruguaiana, Rio Grande do Sul, Brazil; Polytechnic School, Federal University of Santa Maria, Av. Roraima, n° 1000, Santa Maria, Rio Grande do Sul, Brazil.
| | - Patricia Corrales
- Department of Basic Health Sciences, Universidad Rey Juan Carlos, Antenas s/n, Alcorcón, Spain.
| | - Janaina Trindade Piagette
- Cardiovascular Physiology Laboratory, Universidade Federal do Pampa, BR 472, Km 592, Uruguaiana, Rio Grande do Sul, Brazil.
| | | | - Gema Medina-Gomez
- Department of Basic Health Sciences, Universidad Rey Juan Carlos, Antenas s/n, Alcorcón, Spain.
| | - Franck Maciel Peçanha
- Cardiovascular Physiology Laboratory, Universidade Federal do Pampa, BR 472, Km 592, Uruguaiana, Rio Grande do Sul, Brazil.
| | - Dalton Valentim Vassallo
- Cardiac Electromechanical and Vascular Reactivity Laboratory, Universidade Federal do Espírito Santo, Marechal Campos, 1468, Vitória, Espírito Santo, Brazil.
| | - Marta Miguel
- Bioactivity and Food Analysis Laboratory, Instituto de Investigación en Ciencias de la Alimentación, Nicolás Cabrera, 9, Campus Universitario de Cantoblanco, Madrid, Spain.
| | - Giulia Alessandra Wiggers
- Cardiovascular Physiology Laboratory, Universidade Federal do Pampa, BR 472, Km 592, Uruguaiana, Rio Grande do Sul, Brazil.
| |
Collapse
|
179
|
Pulmonary Hypertension and Obesity: Focus on Adiponectin. Int J Mol Sci 2019; 20:ijms20040912. [PMID: 30791536 PMCID: PMC6412189 DOI: 10.3390/ijms20040912] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 02/15/2019] [Accepted: 02/16/2019] [Indexed: 02/07/2023] Open
Abstract
Pulmonary hypertension is an umbrella term including many different disorders causing an increase of the mean pulmonary arterial pressure (mPAP) ≥ 25 mmHg. Recent data revealed a strong association between obesity and pulmonary hypertension. Adiponectin is a protein synthetized by the adipose tissue with pleiotropic effects on inflammation and cell proliferation, with a potential protective role on the pulmonary vasculature. Both in vivo and in vitro studies documented that adiponectin is an endogenous modulator of NO production and interferes with AMP-activated protein kinase (AMPK) activation, mammalian target of rapamycin (mTOR), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κβ) signaling preventing endothelial dysfunction and proliferation. Furthermore, adiponectin ameliorates insulin resistance by mediating the biological effects of peroxisome proliferator-activated receptor-gamma (PPARγ). Therefore, adiponectin modulation emerged as a theoretical target for the treatment of pulmonary hypertension, currently under investigation. Recently, consistent data showed that hypoglycemic agents targeting PPARγ as well as renin–angiotensin system inhibitors and mineralocorticoid receptor blockers may influence pulmonary hemodynamics in different models of pulmonary hypertension.
Collapse
|
180
|
Leptin and adiponectin supplementation modifies mesenteric lymph node lymphocyte composition and functionality in suckling rats. Br J Nutr 2019; 119:486-495. [PMID: 29508690 DOI: 10.1017/s0007114517003786] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
At birth, when immune responses are insufficient, there begins the development of the defence capability against pathogens. Leptin and adiponectin, adipokines that are present in breast milk, have been shown to play a role in the regulation of immune responses. We report here, for the first time, the influence of in vivo adipokine supplementation on the intestinal immune system in early life. Suckling Wistar rats were daily supplemented with leptin (0·7 μg/kg per d, n 36) or adiponectin (35 μg/kg per d, n 36) during the suckling period. The lymphocyte composition, proliferation and cytokine secretion from mesenteric lymph node lymphocytes (on days 14 and 21), as well as intestinal IgA and IgM concentration (day 21), were evaluated. At day 14, leptin supplementation significantly increased the TCRαβ + cell proportion in mesenteric lymph nodes, in particular owing to an increase in the TCRαβ + CD8+ cell population. Moreover, the leptin or adiponectin supplementation promoted the early development CD8+ cells, with adiponectin being the only adipokine capable of enhancing the lymphoproliferative ability at the end of the suckling period. Although leptin decreased intestinal IgA concentration, it had a trophic effect on the intestine in early life. Supplementation of both adipokines modulated the cytokine profile during (day 14) and at the end (day 21) of the suckling period. These results suggest that leptin and adiponectin during suckling play a role in the development of mucosal immunity in early life.
Collapse
|
181
|
Enhancement of immune maturation in suckling rats by leptin and adiponectin supplementation. Sci Rep 2019; 9:1786. [PMID: 30742004 PMCID: PMC6370875 DOI: 10.1038/s41598-018-38418-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 12/12/2018] [Indexed: 12/15/2022] Open
Abstract
Leptin and adiponectin, adipokines present in breast milk, have shown immunomodulatory properties. The current study aimed to ascertain whether a nutritional supplementation with leptin or adiponectin in neonatal rats was able to influence the maturation of the systemic immune response in early life. To achieve this, suckling Wistar rats were supplemented with either leptin (0.7 μg/kg/day) or adiponectin (35 μg/kg/day) during the whole suckling period. Plasmatic immunoglobulins were quantified, and spleen lymphocyte composition and their ability to proliferate and release cytokines were evaluated during (day 14) and at the end (day 21) of the suckling period. Rats fed with either adipokine showed higher plasma IgM and IgG1 concentrations and adiponectin supplementation also increased IgG2a at both studied days (P < 0.05). With regard to the lymphocyte composition, both adipokine supplementations increased T cell proportion and both CD4+ and CD8+ T cell subsets after two weeks of supplementation (P < 0.05). Moreover, only leptin administration increased NK and NKT cell proportions at the end of the suckling period. Finally, both adipokines influenced the cytokine secretion pattern by splenocytes. In conclusion, these results suggest that leptin and adiponectin play a role in the maturation of the systemic immune response during the suckling period.
Collapse
|
182
|
Flynn MG, Markofski MM, Carrillo AE. Elevated Inflammatory Status and Increased Risk of Chronic Disease in Chronological Aging: Inflamm-aging or Inflamm-inactivity? Aging Dis 2019; 10:147-156. [PMID: 30705775 PMCID: PMC6345337 DOI: 10.14336/ad.2018.0326] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 03/26/2018] [Indexed: 12/12/2022] Open
Abstract
Age-associated hyper-inflammation or "inflamm-aging" has been linked to the development of chronic diseases and characterized as an unavoidable aspect of aging. However, the inflamm-aging model does not adequately address the potential anti-inflammatory effects of exercise training and the potential for exercise to ameliorate several age-related diseases. In this brief review, we introduce a new paradigm-inflamm-inactivity-that describes a potent counter-measure to age-associated inflammatory illness.
Collapse
Affiliation(s)
- Michael G. Flynn
- HCA South Atlantic Division, Charleston, SC 29492, USA
- College of Charleston, Charleston, SC 29424, USA
| | - Melissa M. Markofski
- University of Houston, Department of Health and Human Performance, Houston, TX 77204, USA
| | - Andres E. Carrillo
- Department of Exercise Science, Chatham University, Pittsburgh, PA 15232, USA
- FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece
| |
Collapse
|
183
|
Jasinski-Bergner S, Kielstein H. Adipokines Regulate the Expression of Tumor-Relevant MicroRNAs. Obes Facts 2019; 12:211-225. [PMID: 30999294 PMCID: PMC6547259 DOI: 10.1159/000496625] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 01/05/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Increasing prevalence of obesity requires the investigation of respective comorbidities, including tumor diseases like colorectal, renal, post-menopausal breast, prostate cancer, and leukemia. To date, molecular mechanisms of the malignant transformation of these peripheral tissues induced by obesity remain unclear. Adipose tissue secretes factors with hormone-like functions, the adipokines, and is therefore categorized as an endocrine organ. Current research demonstrates the ability of adipose tissue to alter DNA methylation and gene expression in peripheral tissues, probably affecting microRNA (miR) expression. METHODS Literature was analyzed for adipokine-regulated miRs. Many of these adipokine upregulated or downregulated miRs exert either oncogenic or anti-tumoral potential. RESULTS The three selected and analyzed adipokines, adiponectin, leptin, and resistin, induce more strongly oncogenic miRs and simultaneously reduce anti-tumoral miRs than vice versa. This effect is not only true for the pure number of regulated miRs, it is also the case by consideration of the abundance of the respective miR expression based on actual data sets derived from next-generation sequencing. CONCLUSION The link of obesity and cancer is analyzed under the aspect of adipokine-regulated miRs. At the same time the impact of miR abundance is considered as a regulatory variable. This context offers new strategies for tumor therapy and diagnostics.
Collapse
Affiliation(s)
- Simon Jasinski-Bergner
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany,
| | - Heike Kielstein
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
184
|
Abstract
OBJECTIVE Little is known about the impact of the home environment on biomarkers of obesity, such as adipokines, in children. In this study, we examined the relationship of maternal depressive symptoms and potentially protective social factors, including maternal support and the home learning environment, with body mass index and adipokines. METHODS Data were obtained from 326 Mexican American participants from the Center for the Health Assessment of Mothers and Children of Salinas cohort. Plasma adipokine levels were assessed in 326 children by enzyme-linked immunoassay at birth or ages 5, 9, or 14 years. Maternal depressive symptoms were evaluated using the Center for Epidemiological Studies Depression Scale when children were 1, 3.5, 7, and 9 years old; social support was assessed by the Duke-University of North Carolina Questionnaire at ages 1 and 5 years; and home learning environment by the Home Observation for the Measurement of the Environment (HOME) at ages of 6 months and 1, 2, 3.5, 7, 9, and 10.5 years. RESULTS Age was significantly associated with adiponectin (B = -5.0, SE = 0.2) and leptin (B = 0.01, SE = 0.003) levels. Individual time point analyses identified significant positive associations of HOME scores in childhood with adiponectin at ages 9 years (HOME score; age 3.5 years: B = 0.9, p = .04) and 14 years (HOME score; age 7 years: B = 0.6, p = .02, age 9 years: B = 0.6, p = .05, age 10.5 years: B = 0.5, p = .04). We observed significant relationships of maternal depressive symptoms at age 9 years with adiponectin and body mass index z-score at age 14 years (B = -0.2, p = .003 and B = 0.02, p = .002, resp.), which were confirmed in longitudinal models. CONCLUSIONS This study adds new evidence that adverse and protective aspects of the home environment could lead to altered obesity status in children.
Collapse
|
185
|
Zhu L, Chen X, Chong L, Kong L, Wen S, Zhang H, Zhang W, Li C. Adiponectin alleviates exacerbation of airway inflammation and oxidative stress in obesity-related asthma mice partly through AMPK signaling pathway. Int Immunopharmacol 2018; 67:396-407. [PMID: 30584969 DOI: 10.1016/j.intimp.2018.12.030] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 12/07/2018] [Accepted: 12/13/2018] [Indexed: 12/15/2022]
Abstract
Adiponectin plays a role in asthma and obesity, but its effects and mechanism in obesity-related asthma remain elusive. This study aimed to evaluate the effects of adiponectin on airway inflammation and oxidative stress and to determine its mechanism in obesity-related asthma. Male C57BL6/J mice fed with a high-fat diet to induce obesity were sensitized and challenged with ovalbumin to induce asthma, and treated with adiponectin (1 mg/kg) and AMP-activated protein kinase (AMPK) inhibitor compound C (20 mg/kg) twice before the first ovalbumin challenge. We found exogenous adiponectin significantly reduced airway resistance, inflammatory infiltration in lung tissue, and cell counts in bronchoalveolar lavage fluid. Adiponectin inhibited great levels of eotaxin, myeloperoxidase, tumor necrosis factor-α, 8‑hydroxy‑2'‑deoxyguanosine, and nitric oxide in obesity-related asthma mice. Moreover, we found increased nuclear factor kappa B p65, inducible nitric oxide synthase and B-cell lymphoma 2 protein expression were down-regulated with adiponectin administration. Additionally, adiponectin elevated the lower levels of pAMPK and AMPK activity in lung tissue. These protective effects of adiponectin were reversed after treatment with the AMPK inhibitor compound C. Thus, we conclude that adiponectin alleviates exacerbation of airway inflammation and oxidative stress in a murine model of obesity-related asthma partly through AMPK signaling pathway.
Collapse
Affiliation(s)
- Lili Zhu
- Department of Pediatric Pulmonology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No.109 Xueyuanxi Road, Lucheng District, Wenzhou 325027, Zhejiang Province, China
| | - Xiuzhen Chen
- Department of Pediatric Pulmonology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No.109 Xueyuanxi Road, Lucheng District, Wenzhou 325027, Zhejiang Province, China; Department of Pediatrics, Hubei Maternal and Child Health Hospital, No.745 Wuluo Road, Hongshan District, Wuhan 430070, Hubei Province, China
| | - Lei Chong
- Department of Pediatric Pulmonology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No.109 Xueyuanxi Road, Lucheng District, Wenzhou 325027, Zhejiang Province, China
| | - Ludan Kong
- Department of Pediatric Pulmonology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No.109 Xueyuanxi Road, Lucheng District, Wenzhou 325027, Zhejiang Province, China
| | - Shunhang Wen
- Department of Pediatric Pulmonology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No.109 Xueyuanxi Road, Lucheng District, Wenzhou 325027, Zhejiang Province, China
| | - Hailin Zhang
- Department of Pediatric Pulmonology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No.109 Xueyuanxi Road, Lucheng District, Wenzhou 325027, Zhejiang Province, China
| | - Weixi Zhang
- Department of Pediatric Pulmonology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No.109 Xueyuanxi Road, Lucheng District, Wenzhou 325027, Zhejiang Province, China
| | - Changchong Li
- Department of Pediatric Pulmonology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No.109 Xueyuanxi Road, Lucheng District, Wenzhou 325027, Zhejiang Province, China.
| |
Collapse
|
186
|
Burhans MS, Hagman DK, Kuzma JN, Schmidt KA, Kratz M. Contribution of Adipose Tissue Inflammation to the Development of Type 2 Diabetes Mellitus. Compr Physiol 2018; 9:1-58. [PMID: 30549014 DOI: 10.1002/cphy.c170040] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The objective of this comprehensive review is to summarize and discuss the available evidence of how adipose tissue inflammation affects insulin sensitivity and glucose tolerance. Low-grade, chronic adipose tissue inflammation is characterized by infiltration of macrophages and other immune cell populations into adipose tissue, and a shift toward more proinflammatory subtypes of leukocytes. The infiltration of proinflammatory cells in adipose tissue is associated with an increased production of key chemokines such as C-C motif chemokine ligand 2, proinflammatory cytokines including tumor necrosis factor α and interleukins 1β and 6 as well as reduced expression of the key insulin-sensitizing adipokine, adiponectin. In both rodent models and humans, adipose tissue inflammation is consistently associated with excess fat mass and insulin resistance. In humans, associations with insulin resistance are stronger and more consistent for inflammation in visceral as opposed to subcutaneous fat. Further, genetic alterations in mouse models of obesity that reduce adipose tissue inflammation are-almost without exception-associated with improved insulin sensitivity. However, a dissociation between adipose tissue inflammation and insulin resistance can be observed in very few rodent models of obesity as well as in humans following bariatric surgery- or low-calorie-diet-induced weight loss, illustrating that the etiology of insulin resistance is multifactorial. Taken together, adipose tissue inflammation is a key factor in the development of insulin resistance and type 2 diabetes in obesity, along with other factors that likely include inflammation and fat accumulation in other metabolically active tissues. © 2019 American Physiological Society. Compr Physiol 9:1-58, 2019.
Collapse
Affiliation(s)
- Maggie S Burhans
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Derek K Hagman
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jessica N Kuzma
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Kelsey A Schmidt
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Mario Kratz
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
187
|
Żelechowska P, Kozłowska E, Pastwińska J, Agier J, Brzezińska-Błaszczyk E. Adipocytokine Involvement in Innate Immune Mechanisms. J Interferon Cytokine Res 2018; 38:527-538. [PMID: 30431386 DOI: 10.1089/jir.2018.0102] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The innate immune response is defined as an immensely complex and sophisticated process aimed at defending the organism against any disturbance in the body homeostasis, including invading pathogens. It requires a close cooperation of a vast amount of different cell types, recognized as inflammatory migrating cells, as well as stationary cells that form tissues. Moreover, innate immune mechanisms require an efficient functioning of various humoral components that exert a significant impact on physiological and pathological processes. Apart from commonly mentioned humoral factors, this group also includes a family of proteins known as adipocytokines that may act as pro- or anti-inflammatory agents or act both ways. Leptin, predominantly characterized as a proinflammatory adipokine, plays a crucial role in endothelium remodeling and regulation, as well as in cell survival and production of numerous cytokines. Adiponectin, similar to leptin, acts on the endothelial cells and the phagocytic properties of immune cells; however, it exerts an anti-inflammatory impact. Resistin has a documented role in the control of angiogenesis and stimulation of proinflammatory mediator generation and release. Furthermore, there are adipokines, ie, visfatin and chemerin, whose participation in the inflammatory processes is ambiguous. This review focuses on the current knowledge on the extensive role of selected adipokines in innate immune response.
Collapse
Affiliation(s)
- Paulina Żelechowska
- Department of Experimental Immunology, Medical University of Lodz, Lodz, Poland
| | - Elżbieta Kozłowska
- Department of Experimental Immunology, Medical University of Lodz, Lodz, Poland
| | - Joanna Pastwińska
- Department of Experimental Immunology, Medical University of Lodz, Lodz, Poland
- Laboratory of Cellular Immunology, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Justyna Agier
- Department of Experimental Immunology, Medical University of Lodz, Lodz, Poland
| | | |
Collapse
|
188
|
Yang J, Du G, Wang J, Chen J, Yang C, Li J, Zhang Y. Reduced Serum Adiponectin Level and Risk of Poststroke Depression in Patients with Ischemic Stroke. J Stroke Cerebrovasc Dis 2018; 28:305-310. [PMID: 30391327 DOI: 10.1016/j.jstrokecerebrovasdis.2018.09.057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 09/08/2018] [Accepted: 09/30/2018] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND AND AIMS Decreased adiponectin (APN) level has been indicated to be associated with depression. In the present study, we aimed to investigate whether serum APN could predict poststroke depression (PSD) at 3 months in patients with acute ischemic stroke. METHOD Patients with first-ever ischemic stroke and hospitalized within 24 hours of symptoms onset were enrolled prospectively during March 2017 to September 2017. Serum APN level was measured at admission by enzyme-linked immunosorbent assay. Neuropsychological evaluations were performed at the 3-month follow-up. PSD was diagnosed using the Chinese version of the Structured Clinical Interview for DSM-IV. The association between APN level and predict PSD was analyzed by binary logistic regression analysis. RESULTS Of the 255 acute ischemic stroke patients included, the median (interquartile range) APN level was 5.4 (3.0-7.5) μg/mL. PSD was observed in 69 patients, which accounted for 27.1% (95% confidence interval, 24.3%-29.9%) of the cohort. Patients with PSD showed lower level of APN (3.5 [2.5-6.3] μg/mL versus 6.2 [3.5-8.0] μg/mL, P = .001) at admission. Univariate logistic regression analysis indicated that patients with APN level in the first tertile compared with the third tertile were more likely to have PSD (odds ratio, 3.550; 95% confidence interval, 1.732-7.276; P = .008). The association remained significant even after multivariable adjustment for potential confounders. CONCLUSIONS This study demonstrated that decreased APN level at admission might be associated with PSD in patients after acute ischemic stroke.
Collapse
Affiliation(s)
- Junhua Yang
- Center of Psychosomatic Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.
| | - Guanghui Du
- Center of Psychosomatic Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.
| | - Jinyu Wang
- Center of Psychosomatic Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.
| | - Jia Chen
- Center of Psychosomatic Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.
| | - Chenghui Yang
- University of Electronic Science and Technology of China, Chengdu, China.
| | - Jia Li
- Center of Psychosomatic Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.
| | - Yun Zhang
- Department of Neurology, Mianyang Central Hospital, Mianyang, China.
| |
Collapse
|
189
|
Liu H, Lu XJ, Chen J. Full-length and a smaller globular fragment of adiponectin have opposite roles in regulating monocyte/macrophage functions in ayu, Plecoglossus altivelis. FISH & SHELLFISH IMMUNOLOGY 2018; 82:319-329. [PMID: 30130657 DOI: 10.1016/j.fsi.2018.08.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 08/17/2018] [Accepted: 08/18/2018] [Indexed: 06/08/2023]
Abstract
Adiponectin (ADP), a regulator of the innate immune system, plays a role in the progression of inflammation and metabolic disorders in mammals. However, the role of ADP in fish is poorly understood. Here, we cloned the cDNA sequence of a ADP homolog (PaADP) gene from ayu. Multiple sequence alignment revealed that PaADP presented typical characteristics of ADPs. Phylogenetic tree analysis showed that PaADP was most closely related to that of rainbow trout. In healthy ayu, the transcripts of PaADP were detected in most of the tested tissues and cells, with the highest level in the adipose tissue. Upon V. anguillarum infection, the mRNA expression of PaADP was significantly up-regulated in the tissues and cells except adipose tissue. Subsequently, the full-length mature PaADP (fPaADP) and the globular domain fragment (gPaADP) were prokaryotically expressed in bacteria and purified, and anti-PaADP antibodies were produced. Western blot analysis revealed that three fragments including fPaADP and gPaADP were existed in ayu serum. The recombinant fPaADP (rfPaADP) had an anti-inflammatory effect on ayu MO/MФ by upregulating anti-inflammatory cytokine expressions, downregulating pro-inflammatory cytokine expressions, inhibiting the phagocytosis and subsequent bacterial killing. In contrast, the recombinant gPaADP (rgPaADP) presented a pro-inflammatory effect on ayu MO/MФ by upregulating pro-inflammatory cytokine expression, downregulating anti-inflammatory cytokine expressions, enhancing the phagocytosis and subsequent bacterial killing. These results suggested that fPaADP and gPaADP have opposite roles in the regulation of MO/MФ functions in ayu.
Collapse
Affiliation(s)
- He Liu
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Xin-Jiang Lu
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Jiong Chen
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
190
|
Udomsinprasert W, Honsawek S, Poovorawan Y. Adiponectin as a novel biomarker for liver fibrosis. World J Hepatol 2018; 10:708-718. [PMID: 30386464 PMCID: PMC6206156 DOI: 10.4254/wjh.v10.i10.708] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/02/2018] [Accepted: 08/06/2018] [Indexed: 02/06/2023] Open
Abstract
Adiponectin is known to play primary roles in the regulation of systemic glucose homeostasis and lipid metabolism. Interestingly, emerging evidence indicates beneficial effects of adiponectin on liver fibrosis; however, the exact mechanisms of this action remain unclear. Herein, we aimed to summarize the recent findings regarding the role of adiponectin in liver fibrogenesis and update the current comprehensive knowledge regarding usefulness of adiponectin-based treatments in liver fibrosis. Adiponectin has been demonstrated to have an anti-fibrotic action in the liver by blocking the activation of hepatic stellate cell-mediated adenosine monophosphate-activated protein kinase and peroxisome proliferator-activated receptor-alpha pathways, which in turn diminish the expression of pro-fibrotic genes. In addition, hyperadiponectinemia was noted in patients with various chronic liver diseases (CLDs)-related liver fibrosis. An increase in circulating adiponectin levels was also found to be associated with the development of liver fibrosis, indicating a role of adiponectin as a non-invasive biomarker for predicting the progression of liver fibrosis. It is therefore reasonable to speculate that adiponectin may be developed as a new therapeutic candidate for the treatment of liver fibrosis. Nonetheless, future observations are still necessary to fully elucidate the extent of the effects of adiponectin on liver fibrotic outcomes, in order to modify adiponectin as an anti-fibrotic therapy that would speed up fibrosis reversal in patients with CLD.
Collapse
Affiliation(s)
- Wanvisa Udomsinprasert
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Sittisak Honsawek
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand.
| |
Collapse
|
191
|
Lee KH, Jeong J, Woo J, Lee CH, Yoo CG. Globular Adiponectin Exerts a Pro-Inflammatory Effect via IκB/NF-κB Pathway Activation and Anti-Inflammatory Effect by IRAK-1 Downregulation. Mol Cells 2018; 41:762-770. [PMID: 30078230 PMCID: PMC6125418 DOI: 10.14348/molcells.2018.0005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 06/08/2018] [Accepted: 06/20/2018] [Indexed: 12/15/2022] Open
Abstract
Adiponectin, a hormone produced by adipose tissue, is very abundant in plasma, and its anti- and pro-inflammatory effects are reported. However, the mechanisms of these pro- and anti-inflammatory effects are not fully defined. Herein, we evaluated the dual inflammatory response mechanism of adiponectin in macrophages. Short-term globular adiponectin (gAd) treatment induced IκBα degradation, NF-κB nuclear translocation, and TNF-α production in RAW 264.7 cells. Polymyxin B pretreatment did not block gAd-induced IκBα degradation, and heated gAd was unable to degrade IκBα, suggesting that the effects of gAd were not due to endotoxin contamination. gAd activated IKK and Akt, and inhibition of either IKK or Akt by dominant-negative IKKβ (DN-IKKβ) or DN-Akt overexpression blocked gAd-induced IκBα degradation, suggesting that short-term incubation with gAd mediates inflammatory responses by activating the IκB/NF-κB and PI3K/Akt pathways. Contrastingly, long-term stimulation with gAd induced, upon subsequent stimulation, tolerance to gAd, lipopolysaccharide, and CpG-oligodeoxynucleotide, which is associated with gAd-induced downregulation of IL-receptor-associated kinase-1 (IRAK-1) due to IRAK-1 transcriptional repression. Conclusively, our findings demonstrate that the pro- and anti-inflammatory responses to gAd in innate immune cells are time-dependent, and mediated by the activation of the IκB/NF-κB pathway, and IRAK-1 downregulation, respectively.
Collapse
Affiliation(s)
- Kyoung-Hee Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080,
Korea
| | - Jiyeong Jeong
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080,
Korea
| | - Jisu Woo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080,
Korea
| | - Chang-Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080,
Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080,
Korea
| | - Chul-Gyu Yoo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080,
Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080,
Korea
| |
Collapse
|
192
|
Miller EM. Ecological immunity of human milk: Life history perspectives from the United States and Kenya. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2018; 167:389-399. [DOI: 10.1002/ajpa.23639] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 06/04/2018] [Accepted: 06/06/2018] [Indexed: 12/15/2022]
|
193
|
Jiang L, Zhu X, Rong J, Xing B, Wang S, Liu A, Chu M, Huang G. Obesity, osteoarthritis and genetic risk: The rs182052 polymorphism in the ADIPOQ gene is potentially associated with risk of knee osteoarthritis. Bone Joint Res 2018; 7:494-500. [PMID: 30123499 PMCID: PMC6076358 DOI: 10.1302/2046-3758.77.bjr-2017-0274.r1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objectives Given the function of adiponectin (ADIPOQ) on the inflammatory condition of obesity and osteoarthritis (OA), we hypothesized that the ADIPOQ gene might be a candidate gene for a marker of susceptibility to OA. Methods We systematically screened three tagging polymorphisms (rs182052, rs2082940 and rs6773957) in the ADIPOQ gene, and evaluated the association between the genetic variants and OA risk in a case-controlled study that included 196 OA patients and 442 controls in a northern Chinese population. Genotyping was performed using the Sequenom MassARRAY iPLEX platform. Results The single nucleotide polymorphism (SNP) rs182052 was found to be potentially associated with knee OA risk (additive model: odds ratio = 1.38; 95% confidence interval 1.07 to 1.76; p = 0.012). Furthermore, a non-significant association was observed for rs182052 and body mass index with regard to OA risk in interaction analyses (p = 0.063). Similarly, no significant interaction was detected for rs182052 and age with regard to OA risk (p = 0.614). Conclusion These findings suggest that the SNP rs182052 in the ADIPOQ gene may potentially modify individual susceptibility to knee OA in the Chinese population. Further studies are warranted to investigate our findings in more depth. Cite this article: L. Jiang, X. Zhu, J. Rong, B. Xing, S. Wang, A. Liu, M. Chu, G. Huang. Obesity, osteoarthritis and genetic risk: The rs182052 polymorphism in the ADIPOQ gene is potentially associated with risk of knee osteoarthritis. Bone Joint Res 2018;7:494–500. DOI: 10.1302/2046-3758.77.BJR-2017-0274.R1.
Collapse
Affiliation(s)
- L Jiang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - X Zhu
- Baoshan Center for Disease Control and Prevention,Shanghai, China
| | - J Rong
- Second Department of Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - B Xing
- Hongqi Community Health Service Center, Xiangfang District, Harbin, Heilongjiang Province, China
| | - S Wang
- Second Department of Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - A Liu
- Department of Nutrition, China National Center for Food Safety Risk Assessment, Beijing, China
| | - M Chu
- Department of Epidemiology, Public Health College, Nantong University, Nantong, Jiangsu Province, China
| | - G Huang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
194
|
Impact of obesity on autoimmune arthritis and its cardiovascular complications. Autoimmun Rev 2018; 17:821-835. [PMID: 29885537 PMCID: PMC9996646 DOI: 10.1016/j.autrev.2018.02.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 02/25/2018] [Indexed: 02/06/2023]
Abstract
Obesity can instigate and sustain a systemic low-grade inflammatory environment that can amplify autoimmune disorders and their associated comorbidities. Metabolic changes and inflammatory factors produced by the adipose tissue have been reported to aggravate autoimmunity and predispose the patient to cardiovascular disease (CVD) and metabolic comorbidities. Rheumatoid arthritis (RA) and psoriatic arthritis (PsA) are autoimmune arthritic diseases, often linked with altered body mass index (BMI). Severe joint inflammation and bone destruction have a debilitating impact on the patient's life; there is also a staggering risk of cardiovascular morbidity and mortality. Furthermore, these patients are at risk of developing metabolic symptoms, including insulin resistance resulting in type 2 diabetes mellitus (T2DM). In addition, arthritis severity, progression and response to therapy can be markedly affected by the patient's BMI. Hence, a complex integrative pathogenesis interconnects autoimmunity with metabolic and cardiovascular disorders. This review aims to shed light on the network that connects obesity with RA, PsA, systemic lupus erythematosus and Sjӧgren's syndrome. We have focused on clarifying the mechanism by which obesity affects different cell types, inflammatory factors and traditional therapies in these autoimmune disorders. We conclude that to further optimize arthritis therapy and to prevent CVD, it is imperative to uncover the intricate relation between obesity and arthritis pathology.
Collapse
|
195
|
Šimják P, Cinkajzlová A, Anderlová K, Pařízek A, Mráz M, Kršek M, Haluzík M. The role of obesity and adipose tissue dysfunction in gestational diabetes mellitus. J Endocrinol 2018; 238:R63-R77. [PMID: 29743342 DOI: 10.1530/joe-18-0032] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 05/08/2018] [Indexed: 12/15/2022]
Abstract
Gestational diabetes mellitus is defined as diabetes diagnosed in the second or third trimester of pregnancy in patients with no history of diabetes prior to gestation. It is the most common complication of pregnancy. The underlying pathophysiology shares some common features with type 2 diabetes mellitus (T2DM) combining relatively insufficient insulin secretion with increased peripheral insulin resistance. While a certain degree of insulin resistance is the physiological characteristics of the second half of pregnancy, it is significantly more pronounced in patients with gestational diabetes. Adipose tissue dysfunction and subclinical inflammation in obesity are well-described causes of increased insulin resistance in non-pregnant subjects and are often observed in individuals with T2DM. Emerging evidence of altered adipokine expression and local inflammation in adipose tissue in patients with gestational diabetes suggests an important involvement of adipose tissue in its etiopathogenesis. This review aims to summarize current knowledge of adipose tissue dysfunction and its role in the development of gestational diabetes. We specifically focus on the significance of alterations of adipokines and immunocompetent cells number and phenotype in fat. Detailed understanding of the role of adipose tissue in gestational diabetes may provide new insights into its pathophysiology and open new possibilities of its prevention and treatment.
Collapse
Affiliation(s)
- Patrik Šimják
- Department of Gynaecology and Obstetrics, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Anna Cinkajzlová
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Kateřina Anderlová
- Department of Gynaecology and Obstetrics, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- 3rd Department of Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Antonín Pařízek
- Department of Gynaecology and Obstetrics, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Miloš Mráz
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Michal Kršek
- 3rd Department of Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- 2nd Internal Department, 3rd Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic
| | - Martin Haluzík
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
196
|
|
197
|
Guzik TJ, Skiba DS, Touyz RM, Harrison DG. The role of infiltrating immune cells in dysfunctional adipose tissue. Cardiovasc Res 2018; 113:1009-1023. [PMID: 28838042 PMCID: PMC5852626 DOI: 10.1093/cvr/cvx108] [Citation(s) in RCA: 298] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 07/05/2017] [Indexed: 12/15/2022] Open
Abstract
Adipose tissue (AT) dysfunction, characterized by loss of its homeostatic functions, is a hallmark of non-communicable diseases. It is characterized by chronic low-grade inflammation and is observed in obesity, metabolic disorders such as insulin resistance and diabetes. While classically it has been identified by increased cytokine or chemokine expression, such as increased MCP-1, RANTES, IL-6, interferon (IFN) gamma or TNFα, mechanistically, immune cell infiltration is a prominent feature of the dysfunctional AT. These immune cells include M1 and M2 macrophages, effector and memory T cells, IL-10 producing FoxP3+ T regulatory cells, natural killer and NKT cells and granulocytes. Immune composition varies, depending on the stage and the type of pathology. Infiltrating immune cells not only produce cytokines but also metalloproteinases, reactive oxygen species, and chemokines that participate in tissue remodelling, cell signalling, and regulation of immunity. The presence of inflammatory cells in AT affects adjacent tissues and organs. In blood vessels, perivascular AT inflammation leads to vascular remodelling, superoxide production, endothelial dysfunction with loss of nitric oxide (NO) bioavailability, contributing to vascular disease, atherosclerosis, and plaque instability. Dysfunctional AT also releases adipokines such as leptin, resistin, and visfatin that promote metabolic dysfunction, alter systemic homeostasis, sympathetic outflow, glucose handling, and insulin sensitivity. Anti-inflammatory and protective adiponectin is reduced. AT may also serve as an important reservoir and possible site of activation in autoimmune-mediated and inflammatory diseases. Thus, reciprocal regulation between immune cell infiltration and AT dysfunction is a promising future therapeutic target.
Collapse
Affiliation(s)
- Tomasz J Guzik
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,Translational Medicine Laboratory, Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland
| | - Dominik S Skiba
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,Translational Medicine Laboratory, Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland
| | - Rhian M Touyz
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - David G Harrison
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,Department of Clinical Pharmacology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
198
|
Abstract
OBJECTIVES The inflammatory process in Crohn disease (CD) involves the visceral fat, characterized by adipocyte hyperplasia and altered adipose tissue and serum concentrations of tumor necrosis factor (TNF), leptin, adiponectin and resistin. We investigated the effect of anti-TNF therapy with infliximab (IFX) on serum adipokine levels in pediatric CD. METHODS Serum concentrations of resistin (ng/mL), leptin (ng/mL), and total adiponectin (μg/mL) were assessed by enzyme-linked immunosorbent assays (ELISA) in 18 pediatric CD patients (mean age 15.0 ± 1.5 years) before first, second, and fourth IFX infusion (weeks 0, 2, and 14) and compared with baseline values from sex- and BMI-matched healthy controls (HC, mean age 13.4 ± 1.6 years). RESULTS At baseline, CD patients (mean age 15.0 ± 1.5 years, 10 of 18 boys) compared with HC (13.4 ± 1.6 years, 7 of 15 boys) had higher resistin levels (median 14.7 ng/mL, range 5.1-50.5 vs 7.3 ng/mL, 0.5-14.5); P = 0.0002). At weeks 2 and 14, resistin decreased to 6.9 ng/mL (2.9-16.8) (P < 0.0001) and 9.2 ng/mL (4.1-20.6; P = 0.0011), respectively. Leptin and adiponectin were comparable between patients and HC at baseline. Leptin increased in girls from 9.5 ng/mL (4.0-30.1) to 16.0 ng/mL (7.9-35.2; P = 0.0156) and 17.2 ng/mL (10.8- 26.8; P = 0.1953) at weeks 0, 2, and 14 respectively; with a trend in boys from 2 (0.6-12.9) to 2.8 (1.7-8.6; P = 0.0840) and 3.3 (1.3-4.6; P = 0.1309). Adiponectin peaked initially from 7.8 μg/mL (4.6-11.9) at week 0 to 9.2 μg/mL (4.1-20.7; P = 0.0005) at week 2 and thereafter fell to 6.5 μg/mL (3.0-12.7; P = 0.0182) at week 14. CONCLUSIONS TNF blockade is associated with changes in circulating adipokines. The marked early increase of the potent anti-inflammatory adiponectin may contribute to the rapid response to IFX in CD.
Collapse
|
199
|
Alwarawrah Y, Kiernan K, MacIver NJ. Changes in Nutritional Status Impact Immune Cell Metabolism and Function. Front Immunol 2018; 9:1055. [PMID: 29868016 PMCID: PMC5968375 DOI: 10.3389/fimmu.2018.01055] [Citation(s) in RCA: 326] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 04/27/2018] [Indexed: 12/15/2022] Open
Abstract
Immune cell function and metabolism are closely linked. Many studies have now clearly demonstrated that alterations in cellular metabolism influence immune cell function and that, conversely, immune cell function determines the cellular metabolic state. Less well understood, however, are the effects of systemic metabolism or whole organism nutritional status on immune cell function and metabolism. Several studies have demonstrated that undernutrition is associated with immunosuppression, which leads to both increased susceptibility to infection and protection against several types of autoimmune disease, whereas overnutrition is associated with low-grade, chronic inflammation that increases the risk of metabolic and cardiovascular disease, promotes autoreactivity, and disrupts protective immunity. Here, we review the effects of nutritional status on immunity and highlight the effects of nutrition on circulating cytokines and immune cell populations in both human studies and mouse models. As T cells are critical members of the immune system, which direct overall immune response, we will focus this review on the influence of systemic nutritional status on T cell metabolism and function. Several cytokines and hormones have been identified which mediate the effects of nutrition on T cell metabolism and function through the expression and action of key regulatory signaling proteins. Understanding how T cells are sensitive to both inadequate and overabundant nutrients may enhance our ability to target immune cell metabolism and alter immunity in both malnutrition and obesity.
Collapse
Affiliation(s)
- Yazan Alwarawrah
- Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Kaitlin Kiernan
- Department of Immunology, Duke University Medical Center, Durham, NC, United States
| | - Nancie J MacIver
- Department of Pediatrics, Duke University Medical Center, Durham, NC, United States.,Department of Immunology, Duke University Medical Center, Durham, NC, United States.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
200
|
Gamberi T, Magherini F, Modesti A, Fiaschi T. Adiponectin Signaling Pathways in Liver Diseases. Biomedicines 2018; 6:biomedicines6020052. [PMID: 29735928 PMCID: PMC6027295 DOI: 10.3390/biomedicines6020052] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/26/2018] [Accepted: 05/02/2018] [Indexed: 02/07/2023] Open
Abstract
In the liver, adiponectin regulates both glucose and lipid metabolism and exerts an insulin-sensitizing effect. The binding of adiponectin with its specific receptors induces the activation of a proper signaling cascade that becomes altered in liver pathologies. This review describes the different signaling pathways in healthy and diseased hepatocytes, also highlighting the beneficial role of adiponectin in autophagy activation and hepatic regeneration.
Collapse
Affiliation(s)
- Tania Gamberi
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Viale Morgagni 50, 50134 Firenze, Italy.
| | - Francesca Magherini
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Viale Morgagni 50, 50134 Firenze, Italy.
| | - Alessandra Modesti
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Viale Morgagni 50, 50134 Firenze, Italy.
| | - Tania Fiaschi
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Viale Morgagni 50, 50134 Firenze, Italy.
| |
Collapse
|