1
|
Mounika N, Mungase SB, Verma S, Kaur S, Deka UJ, Ghosh TS, Adela R. Inflammatory Protein Signatures as Predictive Disease-Specific Markers for Non-Alcoholic Steatohepatitis (NASH). Inflammation 2025; 48:25-41. [PMID: 38676759 DOI: 10.1007/s10753-024-02035-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic disease worldwide, consisting of a broad spectrum of diseases such as simple steatosis (NAFL), non-alcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and hepatocellular carcinoma. Hepatic inflammation plays a key role in the pathophysiology of NAFLD. Inflammatory mediators such as cytokines and chemokines are considered as contributing factors to NAFLD development and progression. In the present study, we aimed to investigate the inflammatory protein signatures as predictive disease-specific markers for non-alcoholic fatty liver disease (NAFLD). This cross-sectional study included healthy control (n = 64), NAFL (n = 109), and NASH (n = 60) human subjects. Serum concentrations of various cytokines and chemokines were evaluated using sensitive multiplex assays. We used principal component analysis (PCoA) to reveal distinct differences in the levels of cytokines and chemokines between each of the study groups. Further, a random forest classification model was developed to identify the panel of markers that could predict diseases. The protein-protein network analysis was performed to determine the various signaling pathways associated with the disease-specific panel of markers. Serum concentrations of TNF-α, IL-1β, IL-1ra, G-CSF, PDGF-BB, MCP-1, MIP-1a, MIP-1b, RANTES, eotaxin, IL-8 and IP-10 were significantly increased in NASH group as compared to control group. Furthermore, serum concentrations of IL-9 and IL-13 were significantly lower in the NASH group, whereas IL-2 levels were significantly decreased in the NAFL group when compared to the control group. PCoA results demonstrated statistically significant differences in cytokines and chemokines between each of the study groups (PERMANOVA p = 0.001; R2 = 0.102). RANTES, IL-1ra, MIP-1b, IL-2, and G-CSF could differentiate the NAFL group from the controls; G-CSF, IL-1ra, TNF-α, RANTES, and IL-9 could differentiate the NASH group from the controls; and G-CSF, IL-9, IL-13, eotaxin, and TNF- α could differentiate the NASH group from the NAFL group. Our protein-protein network revealed that these markers are involved in cytokine-cytokine receptor interaction, Th1 and Th2 cell differentiation, TNF, chemokine, JAK/STAT, P13K/Akt, TLR, NOD-like receptor, NF-kB, and adipocytokine signaling pathways which might be responsible for disease pathogenesis. Our study findings revealed a set of distinct cytokine and chemokine markers and they might be considered as biomarkers in distinguishing NASH from NAFL. Future multicentre studies with larger sample size are recommended to determine the potential utility of these panels of markers.
Collapse
Affiliation(s)
- Nadella Mounika
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur (Halugurisuk), Changsari, Kamrup, Assam-781101, India
| | - Suraj Bhausaheb Mungase
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur (Halugurisuk), Changsari, Kamrup, Assam-781101, India
| | - Shivangi Verma
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi (IIIT-Delhi), Okhla Phase III, New Delhi, 110020, India
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver & Biliary Science (ILBS), New Delhi-110 070, Vasant Kunj, India
| | - Utpal Jyoti Deka
- Department of Gastroenterology, Downtown Hospital, GS Road, Bormotoria, Guwahati, Assam-781006, India
| | - Tarini Shankar Ghosh
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi (IIIT-Delhi), Okhla Phase III, New Delhi, 110020, India
| | - Ramu Adela
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur (Halugurisuk), Changsari, Kamrup, Assam-781101, India.
| |
Collapse
|
2
|
Tayefi M, Svedbom A, Ivert L, Lundqvist M, Ruas J, Bradley M, Johansson E. Risk Factors Associated with Weight Gain during Treatment with Dupilumab among Patients with Moderate to Severe Atopic Dermatitis. Acta Derm Venereol 2024; 104:adv40796. [PMID: 39545373 PMCID: PMC11586677 DOI: 10.2340/actadv.v104.40796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
This cohort study used prospectively collected data from the Swedish national quality registry, SwedAD, to investigate weight gain as a possible side effect of dupilumab treatment for atopic dermatitis. Patients on dupilumab were compared with patients on other systemic medications, e.g., methotrexate, cyclosporine, or Janus kinase inhibitors, and possible risk factors for weight change during treatment with dupilumab were analysed. All patients aged 18 years or above, included in SwedAD between March 2018 and April 2023, who initiated systemic treatment at or after inclusion and had data on weight at baseline and at least 1 follow-up weight measurement were included (n = 157). After 2 years on dupilumab, patients had a mean weight gain of 1.6 kg (p = 0.007, 95% confidence interval [CI] 0.4-2.7). In the multivariable analysis, controlling for age at start, sex, asthma, and body mass index at start, dupilumab was associated with higher weight gain than other systemic treatments (3.3 kg, p = 0.005 [95% CI 1.0-5.6]). Asthma was associated with weight loss; male sex tended to be associated with weight gain.
Collapse
Affiliation(s)
- Mahsa Tayefi
- Division of Dermatology and Venereology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Department of Dermatology and Venereology, Karolinska University Hospital, Stockholm, Sweden.
| | - Axel Svedbom
- Division of Dermatology and Venereology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Lina Ivert
- Division of Dermatology and Venereology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Department of Dermatology and Venereology, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Lundqvist
- Department of Dermatology and Venereology, Karolinska University Hospital, Stockholm, Sweden
| | - Jorge Ruas
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, Stockholm, Sweden; Department of Pharmacology and Stanley and Judith Frankel Institute for Heart & Brain Health, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Maria Bradley
- Division of Dermatology and Venereology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Department of Dermatology and Venereology, Karolinska University Hospital, Stockholm, Sweden
| | - Emma Johansson
- Division of Dermatology and Venereology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Department of Dermatology and Venereology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
3
|
Reale M, Licci G, Orlando P, Matucci A, Trabalzini F, Maggiore G, Gallo O. Efficacy and safety of dupilumab in the treatment of CRSwNP in the real-life setting: a review of the literature. Eur Arch Otorhinolaryngol 2024; 281:5023-5031. [PMID: 38762844 DOI: 10.1007/s00405-024-08725-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 05/04/2024] [Indexed: 05/20/2024]
Abstract
INTRODUCTION The recent approval of Dupilumab has profoundly revolutionized the management of patients affected by severe and recalcitrant Chronic Rhinosinusitis with Nasal Polyps (CRSwNP). However, a review that summarizes the results of real-life studies and compares them to phase 3 studies SINUS-24 and 52 is still lacking. MATERIALS AND METHODS A search of all real-life studies published from 2019 to 2023 was performed. Patients characteristics at baseline and 6 and 12 months after starting Dupilumab were extracted and compared to those from phase 3 trials: age, sex, smoking habits, comorbid asthma and aspirin-exacerbated respiratory disease (AERD), previous endoscopic sinus surgery (ESS), hematic eosinophils and total IgE, NasalAQ2 Polyps Score (NPS), smell, SNOT-22, adverse events (AEs), and response to treatment. RESULTS 15 papers were included with an overall number of 1658 patients. A higher rate of comorbidities and previous ESS was found in patients from real-life studies. In addition, they had worse smell and SNOT-22 at baseline compared to patients from SINUS-24 and 52. Comorbid and post-ESS patients tended to have a faster NPS and SNOT-22 improvement, although the absolute values were not clinically relevant. A more extensive surgery and a number of ESS ≥ 2 were related to worse olfactory outcomes, probably due to iatrogenic damage. No correlation was found between hematic eosinophils and outcomes. AEs were reported by 12.4% of patients and 2.2% had to discontinue dupilumab. Weight gain was an emergent AE (0.8%), probably related to the restored sense of smell and taste. Non-responders were 3.5% and they were switched to systemic steroid, ESS, or another biologic. CONCLUSION Despite some differences in prescription criteria between countries, dupilumab was demonstrated to be effective even in the real-life scenario. However, emerging AEs and possible unknown long-term AEs of a likely lifelong therapy should be considered.
Collapse
Affiliation(s)
- Marella Reale
- Department of Otorhinolaryngology, Careggi University Hospital, Largo Brambilla, 3 - 50134, Florence, Italy
- Department of Otorhinolaryngology, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Giuseppe Licci
- Department of Otorhinolaryngology, Careggi University Hospital, Largo Brambilla, 3 - 50134, Florence, Italy
| | - Pietro Orlando
- Department of Otorhinolaryngology, Careggi University Hospital, Largo Brambilla, 3 - 50134, Florence, Italy.
| | - Andrea Matucci
- Department of Immunology, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Franco Trabalzini
- Department of Otorhinolaryngology, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giandomenico Maggiore
- Department of Otorhinolaryngology, Careggi University Hospital, Largo Brambilla, 3 - 50134, Florence, Italy
| | - Oreste Gallo
- Department of Otorhinolaryngology, Careggi University Hospital, Largo Brambilla, 3 - 50134, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
4
|
Nishimura Y. [Regulation of adipose progenitors and fibro-adipogenic progenitors through primary cilia]. Nihon Yakurigaku Zasshi 2024; 159:188-191. [PMID: 38684399 DOI: 10.1254/fpj.23108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
The primary cilium, an antenna-like structure of cell membrane, detects various signals and regulates cellular functions such as proliferation and differentiation. The impairment of primary cilium is associated with the etiologies of diseases including cancer, obesity, and congenital anomalies. In this review, novel functions of trichoplein, a suppressor of ciliogenesis, on the regulation of adipose progenitors and fibro-adipogenic progenitors are focused. Trichoplein-knockout mice show resistance to high-fat diet-induced obesity and accelerated regeneration after skeletal muscle injury. The primary cilia of adipose progenitors from trichoplein-knockout mice are elongated, leading to the inhibitions of the accumulation of lipid raft to the base of primary cilia and the phosphorylation of AKT. The primary cilia of fibro-adipogenic progenitors from trichoplein-knockout mice are also elongated, causing the increased expression of IL-13 through IL-33 receptor signaling. These mechanisms are involved in the resistance to diet-induced obesity and improved regeneration. These findings suggest that targeting the primary cilia of specific cells may be a novel therapeutic approach through modulating cellular functions.
Collapse
Affiliation(s)
- Yuhei Nishimura
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine
- Research Center for Cilia and Diseases, Mie University Graduate School of Medicine
| |
Collapse
|
5
|
Garneau L, Mulvihill EE, Smith SR, Sparks LM, Aguer C. Myokine Secretion following an Aerobic Exercise Intervention in Individuals with Type 2 Diabetes with or without Exercise Resistance. Int J Mol Sci 2024; 25:4889. [PMID: 38732106 PMCID: PMC11084395 DOI: 10.3390/ijms25094889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/19/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Type 2 diabetes (T2D) is characterized by muscle metabolic dysfunction that exercise can minimize, but some patients do not respond to an exercise intervention. Myokine secretion is intrinsically altered in patients with T2D, but the role of myokines in exercise resistance in this patient population has never been studied. We sought to determine if changes in myokine secretion were linked to the response to an exercise intervention in patients with T2D. The participants followed a 10-week aerobic exercise training intervention, and patients with T2D were grouped based on muscle mitochondrial function improvement (responders versus non-responders). We measured myokines in serum and cell-culture medium of myotubes derived from participants pre- and post-intervention and in response to an in vitro model of muscle contraction. We also quantified the expression of genes related to inflammation in the myotubes pre- and post-intervention. No significant differences were detected depending on T2D status or response to exercise in the biological markers measured, with the exception of modest differences in expression patterns for certain myokines (IL-1β, IL-8, IL-10, and IL-15). Further investigation into the molecular mechanisms involving myokines may explain exercise resistance with T2D; however, the role in metabolic adaptations to exercise in T2D requires further investigation.
Collapse
Affiliation(s)
- Léa Garneau
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (L.G.); (E.E.M.)
- Institut du Savoir Montfort, Ottawa, ON K1K 0T2, Canada
| | - Erin E. Mulvihill
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (L.G.); (E.E.M.)
- University of Ottawa Heart Institute, Ottawa, ON K1Y 4W7, Canada
| | - Steven R. Smith
- Translational Research Institute for Metabolism and Diabetes, AdventHealth Orlando, Orlando, FL 32804, USA; (S.R.S.); (L.M.S.)
| | - Lauren M. Sparks
- Translational Research Institute for Metabolism and Diabetes, AdventHealth Orlando, Orlando, FL 32804, USA; (S.R.S.); (L.M.S.)
| | - Céline Aguer
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (L.G.); (E.E.M.)
- Institut du Savoir Montfort, Ottawa, ON K1K 0T2, Canada
- Faculty of Medicine and Health Sciences, Department of Physiology, McGill University–Campus Outaouais, Gatineau, QC J8V 3T4, Canada
- Faculty of Health Sciences, School of Human Kinetics, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
6
|
Engin A. Reappraisal of Adipose Tissue Inflammation in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:297-327. [PMID: 39287856 DOI: 10.1007/978-3-031-63657-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Chronic low-grade inflammation is a central component in the pathogenesis of obesity-related expansion of adipose tissue and complications in other metabolic tissues. Five different signaling pathways are defined as dominant determinants of adipose tissue inflammation: These are increased circulating endotoxin due to dysregulation in the microbiota-gut-brain axis, systemic oxidative stress, macrophage accumulation, and adipocyte death. Finally, the nucleotide-binding and oligomerization domain (NOD) leucine-rich repeat family pyrin domain-containing 3 (NLRP3) inflammasome pathway is noted to be a key regulator of metabolic inflammation. The NLRP3 inflammasome and associated metabolic inflammation play an important role in the relationships among fatty acids and obesity. Several highly active molecules, including primarily leptin, resistin, adiponectin, visfatin, and classical cytokines, are abundantly released from adipocytes. The most important cytokines that are released by inflammatory cells infiltrating obese adipose tissue are tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), monocyte chemoattractant protein 1 (MCP-1) (CCL-2), and IL-1. All these molecules mentioned above act on immune cells, causing local and then general inflammation. Three metabolic pathways are noteworthy in the development of adipose tissue inflammation: toll-like receptor 4 (TLR4)/phosphatidylinositol-3'-kinase (PI3K)/Protein kinase B (Akt) signaling pathway, endoplasmic reticulum (ER) stress-derived unfolded protein response (UPR), and inhibitor of nuclear factor kappa-B kinase beta (IKKβ)-nuclear factor kappa B (NF-κB) pathway. In fact, adipose tissue inflammation is an adaptive response that contributes to a visceral depot barrier that effectively filters gut-derived endotoxin. Excessive fatty acid release worsens adipose tissue inflammation and contributes to insulin resistance. However, suppression of adipose inflammation in obesity with anti-inflammatory drugs is not a rational solution and paradoxically promotes insulin resistance, despite beneficial effects on weight gain. Inflammatory pathways in adipocytes are indeed indispensable for maintaining systemic insulin sensitivity. Cannabinoid type 1 receptor (CB1R) is important in obesity-induced pro-inflammatory response; however, blockade of CB1R, contrary to anti-inflammatory drugs, breaks the links between insulin resistance and adipose tissue inflammation. Obesity, however, could be decreased by improving leptin signaling, white adipose tissue browning, gut microbiota interactions, and alleviating inflammation. Furthermore, capsaicin synthesized by chilies is thought to be a new and promising therapeutic option in obesity, as it prevents metabolic endotoxemia and systemic chronic low-grade inflammation caused by high-fat diet.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
7
|
Roeb E. Interleukin-13 (IL-13)-A Pleiotropic Cytokine Involved in Wound Healing and Fibrosis. Int J Mol Sci 2023; 24:12884. [PMID: 37629063 PMCID: PMC10454844 DOI: 10.3390/ijms241612884] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/09/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
The liver, as a central metabolic organ, is systemically linked to metabolic-inflammatory diseases. In the pathogenesis of the metabolic syndrome, inflammatory and metabolic interactions between the intestine, liver, and adipose tissue lead to the progression of hepatic steatosis to metabolic-dysfunction-associated steatohepatitis (MASH) and consecutive MASH-induced fibrosis. Clinical and animal studies revealed that IL-13 might be protective in the development of MASH through both the preservation of metabolic functions and Th2-polarized inflammation in the liver and the adipose tissue. In contrast, IL-13-associated loss of mucosal gut barrier function and IL-13-associated enhanced hepatic fibrosis may contribute to the progression of MASH. However, there are only a few publications on the effect of IL-13 on metabolic diseases and possible therapies to influence them. In this review article, different aspects of IL-13-associated effects on the liver and metabolic liver diseases, which are partly contradictory, are summarized and discussed on the basis of the recent literature.
Collapse
Affiliation(s)
- Elke Roeb
- Department of Gastroenterology, Justus Liebig University Giessen, Klinikstr. 33, 35392 Giessen, Germany
| |
Collapse
|
8
|
Son T, Jeong I, Park J, Jun W, Kim A, Kim OK. Adipose tissue-derived exosomes contribute to obesity-associated liver diseases in long-term high-fat diet-fed mice, but not in short-term. Front Nutr 2023; 10:1162992. [PMID: 37229466 PMCID: PMC10203204 DOI: 10.3389/fnut.2023.1162992] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/17/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction Our study aimed to investigate the changes in hepatic endoplasmic reticulum (ER) stress, inflammation, insulin signaling, and lipid metabolism during the administration of a high-fat diet (HFD) in mice in order to identify correlations between obesity and metabolic disease development in the liver. Methods We used short-, medium-, and long-term HFD periods, corresponding to 4, 8, and 12 weeks, respectively, and isolated exosomes from adipose tissue. We confirmed the effect of adipose tissue-derived exosomes on metabolic disorders in obesity in alpha mouse liver 12 (AML12) hepatocytes. Results Adipose tissue-derived exosomes from HFD mice did not affect the AML12 cells after 4 weeks, but ER stress, inflammatory response, insulin resistance, and lipid synthesis were observed after 8 and 12 weeks. Furthermore, we confirmed that an HFD increases the amount of adipose tissue-derived exosomes in mice. Consequently, we can infer that adipose tissue-derived exosomes from HFD-fed mice significantly increase ER stress, inflammatory response, insulin resistance, and lipid synthesis in AML12 cells. Discussion Our results demonstrate that obesity alters the effects of adipose tissue-derived exosomes in the liver, potentially becoming a risk factor in the development of obesity-induced liver diseases.
Collapse
Affiliation(s)
- Taesang Son
- Division of Food and Nutrition, Chonnam National University, Gwangju, Republic of Korea
| | - Inae Jeong
- Division of Food and Nutrition, Chonnam National University, Gwangju, Republic of Korea
| | - Jeongjin Park
- Division of Food and Nutrition, Chonnam National University, Gwangju, Republic of Korea
- Human Ecology Research Institute, Chonnam National University, Gwangju, Republic of Korea
| | - Woojin Jun
- Division of Food and Nutrition, Chonnam National University, Gwangju, Republic of Korea
- Human Ecology Research Institute, Chonnam National University, Gwangju, Republic of Korea
| | - Andre Kim
- Department of Pharmaceutical Engineering, Silla University, Busan, Republic of Korea
| | - Ok-Kyung Kim
- Division of Food and Nutrition, Chonnam National University, Gwangju, Republic of Korea
- Human Ecology Research Institute, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
9
|
Yang Y, Liu H, Liu D. Preventing high-fat diet-induced obesity and related metabolic disorders by hydrodynamic transfer of Il-27 gene. Int J Obes (Lond) 2023; 47:413-421. [PMID: 36959288 DOI: 10.1038/s41366-023-01293-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND AND OBJECTIVES Interleukin-27 (IL-27) is a multifaceted heterodimer cytokine that exerts both pro-inflammatory and anti-inflammatory effects under different physiological conditions. IL-27 signaling plays a role in promoting energy expenditure through enhanced thermogenesis. The objective of the study is to determine the functional role of IL-27 in regulating weight gain, and glucose and lipid homeostasis in mice fed a high-fat diet (HFD). METHODS C57BL/6 mice were hydrodynamically transferred with pLIVE-IL-27 plasmids to achieve elevated level of IL-27 in blood and then kept on a HFD for 8 weeks. The impacts of Il-27 gene transfer on HFD-induced weight gain, adiposity, hepatic lipid accumulation, insulin resistance, glucose homeostasis and the mRNA levels of genes responsible for lipogenesis, glucose homeostasis and proinflammation were assessed by methods of biochemistry, histology, and molecular biology. RESULTS Hydrodynamic gene transfer of Il-27 gene resulted in a peak level of serum IL-27 in mice at 14.5 ng/ml 24 h after gene transfer followed by a sustained level at 2 ng/ml. The elevated level of IL-27 blocked HFD-induced fat accumulation and weight gain without reducing food intake. It also prevented metabolic abnormities of liver steatosis and insulin resistance. IL-27 overexpression promoted expression of major thermogenic genes in brown adipose tissues; and attenuated chronic inflammation and macrophage infiltration into white adipose tissues. CONCLUSIONS The results demonstrate that regulation of IL-27 level could be an effective strategy for management of obesity and obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Yueze Yang
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Huan Liu
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Dexi Liu
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA.
| |
Collapse
|
10
|
Reyes J, Yap GS. Emerging Roles of Growth Differentiation Factor 15 in Immunoregulation and Pathogenesis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:5-11. [PMID: 36542831 PMCID: PMC9779231 DOI: 10.4049/jimmunol.2200641] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/08/2022] [Indexed: 12/24/2022]
Abstract
Growth differentiation factor 15 (GDF-15) is a cytokine that is widely used as a biomarker for the severity of diverse disease states. It also has been shown to play a protective role after tissue injury and to promote a negative energy balance during obesity and diabetes. In addition to its metabolic effects, GDF-15 also regulates the host's immune responses to infectious and noninfectious diseases. GDF-15 can suppress a type 1 and, in contrast, promote a type 2 inflammatory response. In this brief review, we discuss how GDF-15 affects the effector function and recruitment of immune cells, the pathways that induce its expression, and the diverse mechanisms by which it is regulated during inflammation and infection. We further highlight outstanding questions that should be the focus of future investigations in this emerging field.
Collapse
Affiliation(s)
- Jojo Reyes
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ 07101
| | - George S. Yap
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ 07101
| |
Collapse
|
11
|
Silva-Veiga FM, Miranda CS, Vasques-Monteiro IML, Souza-Tavares H, Martins FF, Daleprane JB, Souza-Mello V. Peroxisome proliferator-activated receptor-alpha activation and dipeptidyl peptidase-4 inhibition target dysbiosis to treat fatty liver in obese mice. World J Gastroenterol 2022; 28:1814-1829. [PMID: 35633911 PMCID: PMC9099201 DOI: 10.3748/wjg.v28.i17.1814] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/04/2022] [Accepted: 03/26/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Obesity and comorbidities onset encompass gut dysbiosis, altered intestinal permeability, and endotoxemia. Treatments that target gut dysbiosis can cope with obesity and nonalcoholic fatty liver disease (NAFLD) management. Peroxisome proliferator-activated receptor (PPAR)-alpha activation and dipeptidyl-peptidase-4 (DPP-4) inhibition alleviate NAFLD, but the mechanism may involve gut microbiota modulation and merits further investigation. AIM To address the effects of PPAR-alpha activation and DPP-4 inhibition (isolated or combined) upon the gut-liver axis, emphasizing inflammatory pathways in NAFLD management in high-fat-fed C57BL/6J mice. METHODS Male C57BL/6J mice were fed a control diet (C, 10% of energy as lipids) or a high-fat diet (HFD, 50% of energy as lipids) for 12 wk, when treatments started, forming the groups: C, HF, HFA (HFD + PPAR-alpha agonist WY14643, 2.5 mg/kg body mass), HFL (HFD + DPP-4 inhibitor linagliptin, 15 mg/kg body mass), and HFC (HFD + the combination of WY14643 and linagliptin). RESULTS The HFD was obesogenic compared to the C diet. All treatments elicited significant body mass loss, and the HFC group showed similar body mass to the C group. All treatments tackled oral glucose intolerance and raised plasma glucagon-like peptide-1 concentrations. These metabolic benefits restored Bacteroidetes/Firmicutes ratio, resulting in increased goblet cells per area of the large intestine and reduced lipopolysaccharides concentrations in treated groups. At the gene level, treated groups showed higher intestinal Mucin 2, Occludin, and Zo-1 expression than the HFD group. The reduced endotoxemia suppressed inflammasome and macrophage gene expression in the liver of treated animals. These observations complied with the mitigation of liver steatosis and reduced hepatic triacylglycerol, reassuring the role of the proposed treatments on NAFLD mitigation. CONCLUSION PPAR alpha activation and DPP-4 inhibition (isolated or combined) tackled NAFLD in diet-induced obese mice by restoration of gut-liver axis. The reestablishment of the intestinal barrier and the rescued phylogenetic gut bacteria distribution mitigated liver steatosis through anti-inflammatory signals. These results can cope with NAFLD management by providing pre-clinical evidence that drugs used to treat obesity comorbidities can help to alleviate this silent and harmful liver disease.
Collapse
Affiliation(s)
- Flavia Maria Silva-Veiga
- Department of Anatomy, Institute of Biology, State University of Rio de Janeiro, Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Rio de Janeiro 20551-030, Brazil
| | - Carolline Santos Miranda
- Department of Anatomy, Institute of Biology, State University of Rio de Janeiro, Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Rio de Janeiro 20551-030, Brazil
| | - Isabela Macedo Lopes Vasques-Monteiro
- Department of Anatomy, Institute of Biology, State University of Rio de Janeiro, Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Rio de Janeiro 20551-030, Brazil
| | - Henrique Souza-Tavares
- Department of Anatomy, Institute of Biology, State University of Rio de Janeiro, Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Rio de Janeiro 20551-030, Brazil
| | - Fabiane Ferreira Martins
- Department of Anatomy, Institute of Biology, State University of Rio de Janeiro, Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Rio de Janeiro 20551-030, Brazil
| | - Julio Beltrame Daleprane
- Department of Clinical and Toxicology Analysis, State University of Rio de Janeiro, Nutrition Institute, State University of Rio de Janeiro, Nutrition Institute, University of Sao Paulo, Rio de Janeiro 20551-030, Brazil
| | - Vanessa Souza-Mello
- Department of Anatomy, Institute of Biology, State University of Rio de Janeiro, Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Rio de Janeiro 20551-030, Brazil
| |
Collapse
|
12
|
Heeran AB, McCready J, Dunne MR, Donlon NE, Nugent TS, Bhardwaj A, Mitchelson KAJ, Buckley AM, Ravi N, Roche HM, Reynolds JV, Lynam-Lennon N, O’Sullivan J. Opposing Immune-Metabolic Signature in Visceral Versus Subcutaneous Adipose Tissue in Patients with Adenocarcinoma of the Oesophagus and the Oesophagogastric Junction. Metabolites 2021; 11:768. [PMID: 34822426 PMCID: PMC8624269 DOI: 10.3390/metabo11110768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 12/24/2022] Open
Abstract
Oesophageal adenocarcinoma (OAC) is an exemplar model of obesity-associated cancer. Previous work in our group has demonstrated that overweight/obese OAC patients have better responses to neoadjuvant therapy, but the underlying mechanisms are unknown. Unravelling the immune-metabolic signatures of adipose tissue may provide insight for this observation. We hypothesised that different metabolic pathways predominate in visceral (VAT) and subcutaneous adipose tissue (SAT) and inflammatory secretions will differ between the fat depots. Real-time ex vivo metabolic profiles of VAT and SAT from 12 OAC patients were analysed. These samples were screened for the secretion of 54 inflammatory mediators, and data were correlated with patient body composition. Oxidative phosphorylation (OXPHOS) was significantly higher in VAT when compared to SAT. OXPHOS was significantly higher in the SAT of patients receiving neoadjuvant treatment. VEGF-A, VEGF-C, P1GF, Flt-1, bFGF, IL-15, IL-16, IL-17A, CRP, SAA, ICAM-1, VCAM-1, IL-2, IL-13, IFN-γ, and MIP-1β secretions were significantly higher from VAT than SAT. Higher levels of bFGF, Eotaxin-3, and TNF-α were secreted from the VAT of obese patients, while higher levels of IL-23 and TARC were secreted from the SAT of obese patients. The angiogenic factors, bFGF and VEGF-C, correlated with visceral fat area. Levels of OXPHOS are higher in VAT than SAT. Angiogenic, vascular injury and inflammatory cytokines are elevated in VAT versus SAT, indicating that VAT may promote inflammation, linked to regulating treatment response.
Collapse
Affiliation(s)
- Aisling B. Heeran
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Jessica McCready
- Department of Biological and Physical Sciences, Assumption University, Worcester, MA 01609, USA;
| | - Margaret R. Dunne
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Noel E. Donlon
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Timothy S. Nugent
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Anshul Bhardwaj
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Kathleen A. J. Mitchelson
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, UCD Conway Institute, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland; (K.A.J.M.); (H.M.R.)
| | - Amy M. Buckley
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Narayanasamy Ravi
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Helen M. Roche
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, UCD Conway Institute, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland; (K.A.J.M.); (H.M.R.)
- Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK
| | - John V. Reynolds
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Niamh Lynam-Lennon
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Jacintha O’Sullivan
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| |
Collapse
|
13
|
Bake S, Pinson MR, Pandey S, Chambers JP, Mota R, Fairchild AE, Miranda RC, Sohrabji F. Prenatal alcohol-induced sex differences in immune, metabolic and neurobehavioral outcomes in adult rats. Brain Behav Immun 2021; 98:86-100. [PMID: 34390803 PMCID: PMC8591773 DOI: 10.1016/j.bbi.2021.08.207] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 07/10/2021] [Accepted: 08/06/2021] [Indexed: 12/15/2022] Open
Abstract
Prenatal alcohol exposure (PAE) can result in neurobehavioral anomalies, that may be exacerbated by co-occurring metabolic and immune system deficits. To test the hypothesis that the peripheral inflammation in adult PAE offspring is linked to poor glucose metabolism and neurocognitive deficits, pregnant Sprague-Dawley rats were exposed to ethanol vapor or ambient air during the latter half of gestation. We assessed, in adult offspring of both sexes, performance on a battery of neurocognitive behaviors, glucose tolerance, circulating and splenic immune cells by flow-cytometry, and circulating and tissue (liver, mesenteric adipose, and spleen) cytokines by multiplexed assays. PAE reduced both the ratio of spleen to body weight and splenic regulatory T-cell (Treg) numbers. PAE males, but not females exhibited an increase in circulating monocytes. Overall, PAE males exhibited a suppression of cytokine levels, while PAE females exhibited elevated cytokines in mesenteric adipose tissue (IL-6 and IL1α) and liver (IFN-γ, IL-1β, IL-13, IL-18, IL-12p70, and MCP-1), along with increased glucose intolerance. Behavioral analysis also showed sex-dependent PAE effects. PAE-males exhibited increased anxiety-like behavior while PAE-females showed decreased social interaction. PAE offspring of both sexes exhibited impaired recognition of novel objects. Multilinear regression modeling to predict the association between peripheral immune status, glucose intolerance and behavioral outcomes, showed that in PAE offspring, higher levels of adipose leptin and liver TNF- α predicted higher circulating glucose levels. Lower liver IL-1 α and higher plasma fractalkine predicted more time spent in the center of an open-field with sex being an additional predictor. Higher circulating and splenic Tregs predicted better social interaction in the PAE-offspring. Collectively, our data show that peripheral immune status is a persistent, sex-dependent predictor of glucose intolerance and neurobehavioral function in adult PAE offspring.
Collapse
Affiliation(s)
- Shameena Bake
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Marisa R Pinson
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Sivani Pandey
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Joanna P Chambers
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Roxanna Mota
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Ashlyn E Fairchild
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Rajesh C Miranda
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA.
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA.
| |
Collapse
|
14
|
Beckerman M, Harel C, Michael I, Klip A, Bilan PJ, Gallagher EJ, LeRoith D, Lewis EC, Karnieli E, Levenberg S. GLUT4-overexpressing engineered muscle constructs as a therapeutic platform to normalize glycemia in diabetic mice. SCIENCE ADVANCES 2021; 7:eabg3947. [PMID: 34644106 PMCID: PMC8514095 DOI: 10.1126/sciadv.abg3947] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 08/23/2021] [Indexed: 05/29/2023]
Abstract
Skeletal muscle insulin resistance is a main defect in type 2 diabetes (T2D), which is associated with impaired function and content of glucose transporter type 4 (GLUT4). GLUT4 overexpression in skeletal muscle tissue can improve glucose homeostasis. Therefore, we created an engineered muscle construct (EMC) composed of GLUT4-overexpressing (OEG4) cells. The ability of the engineered implants to reduce fasting glucose levels was tested in diet-induced obesity mice. Decrease and stabilization of basal glucose levels were apparent up to 4 months after implantation. Analysis of the retrieved constructs showed elevated expression of myokines and proteins related to metabolic processes. In addition, we validated the efficiency of OEG4-EMCs in insulin-resistant mice. Following high glucose load administration, mice showed improved glucose tolerance. Our data indicate that OEG4-EMC implant is an efficient mode for restoring insulin sensitivity and improving glucose homeostasis in diabetic mice. Such procedure is a potential innovative modality for T2D therapy.
Collapse
Affiliation(s)
- Margarita Beckerman
- Faculty of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
- Rina and Avner Schneur Center of Diabetes Research, Technion—Israel Institute of Technology, Haifa, Israel
| | - Chava Harel
- Rina and Avner Schneur Center of Diabetes Research, Technion—Israel Institute of Technology, Haifa, Israel
- Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Inbal Michael
- Faculty of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Amira Klip
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Philip J. Bilan
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Emily J. Gallagher
- Division of Endocrinology, Diabetes and Bone Diseases, Samuel Bronfman Department of Medicine, Ichan School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, Samuel Bronfman Department of Medicine, Ichan School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eli C. Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eddy Karnieli
- Rina and Avner Schneur Center of Diabetes Research, Technion—Israel Institute of Technology, Haifa, Israel
- Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Shulamit Levenberg
- Faculty of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
- Rina and Avner Schneur Center of Diabetes Research, Technion—Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
15
|
Ramadhin J, Silva-Moraes V, Nagy T, Norberg T, Harn D. A Comparison of Two Structurally Related Human Milk Oligosaccharide Conjugates in a Model of Diet-Induced Obesity. Front Immunol 2021; 12:668217. [PMID: 34093565 PMCID: PMC8173488 DOI: 10.3389/fimmu.2021.668217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/30/2021] [Indexed: 12/29/2022] Open
Abstract
Obesity is the largest risk factor for the development of chronic diseases in industrialized countries. Excessive fat accumulation triggers a state of chronic low-grade inflammation to the detriment of numerous organs. To address this problem, our lab has been examining the anti-inflammatory mechanisms of two human milk oligosaccharides (HMOs), lacto-N-fucopentaose III (LNFPIII) and lacto-N-neotetraose (LNnT). LNFPIII and LNnT are HMOs that differ in structure via presence/absence of an α1,3-linked fucose. We utilize LNFPIII and LNnT in conjugate form, where 10-12 molecules of LNFPIII or LNnT are conjugated to a 40 kDa dextran carrier (P3DEX/NTDEX). Previous studies from our lab have shown that LNFPIII conjugates are anti-inflammatory, act on multiple cell types, and are therapeutic in a wide range of murine inflammatory disease models. The α1,3-linked fucose residue on LNFPIII makes it difficult and more expensive to synthesize. Therefore, we asked if LNnT conjugates induced similar therapeutic effects to LNFPIII. Herein, we compare the therapeutic effects of P3DEX and NTDEX in a model of diet-induced obesity (DIO). Male C57BL/6 mice were placed on a high-fat diet for six weeks and then injected twice per week for eight weeks with 25µg of 40 kDa dextran (DEX; vehicle control), P3DEX, or NTDEX. We found that treatment with P3DEX, but not NTDEX, led to reductions in body weight, adipose tissue (AT) weights, and fasting blood glucose levels. Mice treated with P3DEX also demonstrated improvements in glucose homeostasis and insulin tolerance. Treatment with P3DEX or NTDEX also induced different profiles of serum chemokines, cytokines, adipokines, and incretin hormones, with P3DEX notably reducing circulating levels of leptin and resistin. P3DEX also reduced WAT inflammation and hepatic lipid accumulation, whereas NTDEX seemed to worsen these parameters. These results suggest that the small structural difference between P3DEX and NTDEX has significant effects on the conjugates' therapeutic abilities. Future work will focus on identifying the receptors for these conjugates and delineating the mechanisms by which P3DEX and NTDEX exert their effects.
Collapse
Affiliation(s)
- Jessica Ramadhin
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States
| | - Vanessa Silva-Moraes
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States
| | - Tamas Nagy
- Department of Pathology, University of Georgia, Athens, GA, United States
| | - Thomas Norberg
- Department of Chemistry-BMC, Uppsala University, Uppsala, Sweden
| | - Donald Harn
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States
| |
Collapse
|
16
|
Michailidou Z, Gomez-Salazar M, Alexaki VI. Innate Immune Cells in the Adipose Tissue in Health and Metabolic Disease. J Innate Immun 2021; 14:4-30. [PMID: 33849008 DOI: 10.1159/000515117] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/09/2021] [Indexed: 11/19/2022] Open
Abstract
Metabolic disorders, such as obesity, type 2 diabetes mellitus, and nonalcoholic fatty liver disease, are characterized by chronic low-grade tissue and systemic inflammation. During obesity, the adipose tissue undergoes immunometabolic and functional transformation. Adipose tissue inflammation is driven by innate and adaptive immune cells and instigates insulin resistance. Here, we discuss the role of innate immune cells, that is, macrophages, neutrophils, eosinophils, natural killer cells, innate lymphoid type 2 cells, dendritic cells, and mast cells, in the adipose tissue in the healthy (lean) and diseased (obese) state and describe how their function is shaped by the obesogenic microenvironment, and humoral, paracrine, and cellular interactions. Moreover, we particularly outline the role of hypoxia as a central regulator in adipose tissue inflammation. Finally, we discuss the long-lasting effects of adipose tissue inflammation and its potential reversibility through drugs, caloric restriction, or exercise training.
Collapse
Affiliation(s)
- Zoi Michailidou
- Centre for Cardiovascular Sciences, Edinburgh University, Edinburgh, United Kingdom
| | - Mario Gomez-Salazar
- Centre for Cardiovascular Sciences, Edinburgh University, Edinburgh, United Kingdom
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
17
|
Simoes E, Correia-Lima J, Sardas L, Storti F, Otani TZDS, Vasques DAC, Otani VHO, Bertolazzi P, Kochi C, Seelaender M, Uchida RR. Sex dimorphism in inflammatory response to obesity in childhood. Int J Obes (Lond) 2021; 45:879-887. [PMID: 33526854 PMCID: PMC8005372 DOI: 10.1038/s41366-021-00753-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 11/23/2020] [Accepted: 01/12/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Childhood overweight and obesity are a global concern, with prevalence rising dramatically over the last decades. The condition is caused by an increase in energy intake and reduction of physical activity, leading to excessive fat accumulation, followed by systemic chronic inflammation and altered function of immune cell responses. This study aimed at providing new insights regarding sex-specificity on the inflammatory response to obesity in the young patient. DESIGN Forty-three Brazilian obese adolescents (Female = 22 and Male=21, BMI (body mass index) Z-score average = 2.78 ± 0.51) and forty-nine eutrophic adolescents (Female = 24 and Male = 25, BMI Z-score average = -0.35 ± 0.88) were enrolled in the study. Anthropometrical analyses and blood cell counts were carried out. Using Luminex®xMAP™ technology, circulating serum cytokines, chemokines, and inflammatory biomarkers were analyzed. Two-way ANOVA test, Tukey's test, and Spearman's correlation coefficient were employed, with a significance threshold set at p < 0.05. RESULTS We identified increased levels of serum amyloid A (SAA), platelets, and leukocytes solely in male obese patients. We found a noteworthy sex-dependent pattern in regard to inflammatory response: obese boys showed higher TNFβ, IL15, and IL2 and lower IL10 and IL13, while obese girls showed increased TNFα, CCL3, CCL4, and IP10 content in the circulation. BMI Z-score was significantly linearly correlated with neutrophils, leukocytes, platelets, SAA, TNFα, CCL3, CCL4, IP10, and IL13 levels within the entire cohort (non-sex-dependent). CONCLUSIONS Our data support a complex relationship between adiposity, blood cell count, and circulating inflammatory cytokine content. High SAA levels suggest that this factor may play a critical role in local and systemic inflammation. In the eutrophic group, females presented a lower status of inflammation, as compared to males. Both obese boys and girls showed an increased inflammatory response in relation to eutrophic counterparts. Taken together, results point out to clear sex dimorphism in the inflammatory profile of obese adolescents.
Collapse
Affiliation(s)
- Estefania Simoes
- Cancer Metabolism Research Group, University of São Paulo, São Paulo, Brazil.
| | - Joanna Correia-Lima
- Cancer Metabolism Research Group, University of São Paulo, São Paulo, Brazil
| | - Leonardo Sardas
- Mental Health Department, Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| | - Felipe Storti
- Mental Health Department, Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| | | | | | | | - Pamela Bertolazzi
- Mental Health Department, Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| | - Cristiane Kochi
- Physiology Department, Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| | - Marilia Seelaender
- Cancer Metabolism Research Group, University of São Paulo, São Paulo, Brazil
- Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
- LIM 26, Hospital das Clínicas of the University of São Paulo, São Paulo, Brazil
| | - Ricardo Riyoiti Uchida
- Mental Health Department, Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| |
Collapse
|
18
|
Wong SW, Ting YW, Yong YK, Tan HY, Barathan M, Riazalhosseini B, Bee CJ, Tee KK, Larsson M, Velu V, Shankar EM, Mohamed R. Chronic inflammation involves CCL11 and IL-13 to facilitate the development of liver cirrhosis and fibrosis in chronic hepatitis B virus infection. Scandinavian Journal of Clinical and Laboratory Investigation 2021; 81:147-159. [PMID: 33528280 DOI: 10.1080/00365513.2021.1876245] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The pathogenesis involving non-alcoholic fatty liver disease (NAFLD) in the context of chronic HBV (CHB) virus infection requires to be understood for developing improved modalities of diagnosis and treatment. We retrospectively investigated the association between NAFLD and CHB virus infection in the context of liver fibrosis. Among the 522 consecutive CHB patients who underwent transient elastography between years 2013 and 2016, we studied 455 subjects in the current investigation. Controlled attenuation parameter (CAP) and liver stiffness measurement (LSM) scores were generally higher in patients with steatosis and fibrosis or cirrhosis. Antiviral treatment had significantly reduced the hepatitis B virus (HBV) viral load. Other liver function markers showed a significant positive correlation with both CAP and LSM scores. Plasma IL-13 was independently associated with increased CAP score where every increase of 1 unit of IL-13 was associated with an increase in CAP score by 0.98 unit. CCL11 was independently associated with LSM with every increase of CCL11 by a unit that, in turn, was associated with an increase of LSM score. We found that there was a high concurrence of NAFLD among patients with CHB virus infection. The presence of metabolic syndrome and chronic inflammation in CHB virus-infected patients were two independent factors that led to the progression of liver cirrhosis, with IL-13 playing the key role in linking the metabolic with the inflammatory components.
Collapse
Affiliation(s)
- Sui-Weng Wong
- Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yi-Wen Ting
- Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yean-Kong Yong
- Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Laboratory Centre, Xiamen University Malaysia, Sepang, Malaysia
| | - Hong-Yien Tan
- Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Traditional Chinese Medicine, Xiamen University Malaysia, Sepang, Malaysia
| | - Muttiah Barathan
- Faculty of Medicine, Department of Medical Microbiology, University of Malaya, Kuala Lumpur, Malaysia
| | - Behnaz Riazalhosseini
- Faculty of Medicine, Department of Pharmacology, University of Malaya, Kuala Lumpur, Malaysia.,Department of Microbiology, College of Basic Science, Shahr-e-Qods Branch, Islamic Azad University, Tehran, Iran
| | - Chook Jack Bee
- School of Healthcare and Medical Sciences, Sunway University, Selangor, Malaysia
| | - Kok-Keng Tee
- Faculty of Medicine, Department of Medical Microbiology, University of Malaya, Kuala Lumpur, Malaysia
| | - Marie Larsson
- Division of Molecular Medicine and Virology, Department of Biomedicine and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Vijayakumar Velu
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Esaki M Shankar
- Infection Biology, Department of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India
| | - Rosmawati Mohamed
- Gastroenterology Unit, Department of Medicine, University of Malaya Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
19
|
Liao KY, Chen CJ, Hsieh SK, Pan PH, Chen WY. Interleukin-13 ameliorates postischemic hepatic gluconeogenesis and hyperglycemia in rat model of stroke. Metab Brain Dis 2020; 35:1201-1210. [PMID: 32632665 DOI: 10.1007/s11011-020-00596-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 06/29/2020] [Indexed: 12/20/2022]
Abstract
Hyperglycemia is a well-known indicator of stroke prognosis, and one-third of nondiabetic patients develop postischemic hyperglycemia during the acute phase of stroke; this is related to relatively poor prognosis, high mortality, and impaired neurological recovery. Interleukin-13 (IL-13), a member of the Th2 cytokine family, is involved in both the regulation of immune response and glucose metabolism. Thus, we investigated the mechanism of postischemic hyperglycemia and the role of IL-13 by using a permanent middle cerebral artery occlusion (MCAO) rat model. Our results indicated that postischemic hyperglycemia was accompanied with hyperinsulinemia and increased HOMA-IR, elevated hepatic gluconeogenesis, and suppressed insulin signaling. A shift towards inflammatory response was evident with results of elevated proinflammatory cytokines and increased expression of negative regulatory proteins, suggesting an ongoing vicious cycle of inflammatory-induced insulin-resistant hyperglycemia. IL-13 treatment counteracted the proinflammatory states and abolished the vicious cycle through enhancing STAT6 and STAT3, which mediated the immune and metabolic pathways respectively; these effects resolved the formerly described pathological changes of postischemic hyperglycemia and reduced infarction size in the MCAO rats. Our findings demonstrated the importance of Th1-Th2 balance in the peripheral glucose metabolism affected by acute ischemic stroke, which provides a new perspective for the prevention and control of postischemic hyperglycemia.
Collapse
Affiliation(s)
- Keng-Ying Liao
- Department of Veterinary Medicine, National Chung Hsing University, 145 Xingda Rd., Taichung City, South Dist., 402, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Sheng-Kuo Hsieh
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Ping-Ho Pan
- Department of Veterinary Medicine, National Chung Hsing University, 145 Xingda Rd., Taichung City, South Dist., 402, Taiwan
- Department of Pediatrics, Tungs' Taichung Metro Harbor Hospital, Taichung, Taiwan
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, 145 Xingda Rd., Taichung City, South Dist., 402, Taiwan.
| |
Collapse
|
20
|
Moghbeli M, Khedmatgozar H, Yadegari M, Avan A, Ferns GA, Ghayour Mobarhan M. Cytokines and the immune response in obesity-related disorders. Adv Clin Chem 2020; 101:135-168. [PMID: 33706888 DOI: 10.1016/bs.acc.2020.06.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The increasing prevalence of obesity and the associated morbidity and mortality are important public health problems globally. There is an important relationship between an unhealthy lifestyle and increased serum inflammatory cytokines. Adipocytes secrete several pro-inflammatory cytokines involved in the recruitment and activation of macrophages resulting in chronic low-grade inflammation. Increased cytokines in obese individual are related to the progression of several disorders including cardiovascular disease, hypertension, and insulin resistance. In present review we have summarized the crucial roles of cytokines and their inflammatory functions in obesity-related immune disorders.
Collapse
Affiliation(s)
- Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Khedmatgozar
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehran Yadegari
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee and Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Brighton, United Kingdom
| | - Majid Ghayour Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
21
|
Johansson EK, Ivert LU, Bradley B, Lundqvist M, Bradley M. Weight gain in patients with severe atopic dermatitis treated with dupilumab: a cohort study. BMC DERMATOLOGY 2020; 20:8. [PMID: 32962676 PMCID: PMC7510313 DOI: 10.1186/s12895-020-00103-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 09/16/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND Dupilumab, targeting the interleukin-4α receptor and inhibiting the action of interleukin-4 and interleukin-13, was recently approved for treatment of moderate to severe atopic dermatitis. There is limited data on long-term effects and safety among patients with severe atopic dermatitis treated with dupilumab. Weight gain was observed among patients treated with dupilumab in our clinic. The aim was to describe weight change in a cohort study of patients with severe atopic dermatitis treated with dupilumab from baseline to follow-up after 12 months, and to analyze if weight change was associated with effect of treatment, reported appetite, and/or disturbed night sleep due to itching. METHODS All patients with atopic dermatitis receiving systemic treatment at the Unit of Dermatology, Karolinska University Hospital, have been registered and monitored consecutively since January 2017. This cohort constituted all patients who started treatment on dupilumab or methotrexate between 10 January 2017 and 30 June 2019 with at least 6 months of follow-up within the study period. The following variables were monitored at start of and during treatment: Eczema Severity Score Index, Patient-Oriented Eczema Measure, visual analogue scale for pruritus 10 cm, Montgomery-Åsberg Depression Rating Scale, Dermatology Life Quality Index, and weight. Data analyses were performed using two-sample Wilcoxon-Mann-Whitney rank-sum test, or the Wilcoxon matched-pairs sign-rank test with a p-value < 0.05 considered as statistically significant. RESULTS Patients treated with dupilumab (n = 12) gained weight (mean 6.1 kg, range [0.1-18.0], p = 0.002) after 1 year on treatment. The majority of patients showed a good response to treatment with dupilumab (n = 11); at follow-up at 6, 9, or 12 months, they reached EASI-90 (n = 6), EASI-75 (n = 4), or EASI-50 (n = 1). There was no significant association between weight gain and treatment response, reported appetite, or disturbed night-sleep due to itch. Patients treated with methotrexate showed no significant weight change (n = 8). CONCLUSIONS To our knowledge, this is the first report on a possible association between weight gain and dupilumab treatment; the extent of the association is yet to be seen, as is the mechanism behind this finding.
Collapse
Affiliation(s)
- Emma Kristin Johansson
- Dermatology and Venereology Unit, Department of Medicine Solna, Karolinska Institutet, SE-171 77, Stockholm, Sweden. .,Dermatological and Venereal Clinic, Södersjukhuset, SE-118 83, Stockholm, Sweden.
| | - Lina Ulrika Ivert
- Dermatology and Venereology Unit, Department of Medicine Solna, Karolinska Institutet, SE-171 77, Stockholm, Sweden.,Department of Dermatology, Karolinska University Hospital Solna, SE-171 76, Stockholm, Sweden
| | - Baltzar Bradley
- Department of Dermatology, Karolinska University Hospital Solna, SE-171 76, Stockholm, Sweden
| | - Maria Lundqvist
- Department of Dermatology, Karolinska University Hospital Solna, SE-171 76, Stockholm, Sweden
| | - Maria Bradley
- Dermatology and Venereology Unit, Department of Medicine Solna, Karolinska Institutet, SE-171 77, Stockholm, Sweden.,Department of Dermatology, Karolinska University Hospital Solna, SE-171 76, Stockholm, Sweden
| |
Collapse
|
22
|
Ni Y, Ni L, Zhuge F, Fu Z. The Gut Microbiota and Its Metabolites, Novel Targets for Treating and Preventing Non-Alcoholic Fatty Liver Disease. Mol Nutr Food Res 2020; 64:e2000375. [PMID: 32738185 DOI: 10.1002/mnfr.202000375] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Indexed: 12/15/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most prevalent metabolic disorders worldwide, along with obesity and type 2 diabetes. NAFLD involves a series of liver abnormalities from simple hepatic steatosis to non-alcoholic steatohepatitis, which can ultimately lead to liver cirrhosis and cancer. The gut-liver axis plays an important role in the development of NAFLD, which depends mainly on regulation of the gut microbiota and its bacterial products. These intestinal bacterial species and their metabolites, including bile acids, tryptophan catabolites, and branched-chain amino acids, regulate adipose tissue and intestinal homeostasis and contribute to the pathogenesis of NAFLD/non-alcoholic steatohepatitis. In this review, the current evidence regarding the key role of the gut microbiota and its metabolites in the pathogenesis and development of NAFLD is highlighted, and the advances in the progression and applied prospects of gut microbiota-targeted dietary and exercise therapies is also discussed.
Collapse
Affiliation(s)
- Yinhua Ni
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310032, China
| | - Liyang Ni
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310032, China
| | - Fen Zhuge
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, 310015, China
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310032, China
| |
Collapse
|
23
|
Boccardi V, Paolacci L, Croce MF, Baroni M, Ercolani S, Cecchetti R, Villa A, Mecocci P. Lower serum levels of IL-13 is associated with increased carotid intima-media thickness in old age subjects. Aging Clin Exp Res 2020; 32:1289-1294. [PMID: 31468495 DOI: 10.1007/s40520-019-01313-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/07/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Cardiovascular diseases due to atherosclerosis represent the major cause of disability and mortality in old age subjects. The atherosclerotic process is linked to a low grade of systemic inflammation with the involvement of many cytokines and inflammatory proteins. Among them, evidence from animal studies suggests that IL-13 has a protective property. However, the role of IL-13 in the pathogenesis of atherosclerosis in humans is still unknown. AIMS With this study, we aim to investigate a potential association between IL-13 and carotid intima-media thickness (IMT) in old age subjects. METHODS This is a retrospective study conducted among 79 old age subjects (over 75 years old). All subjects underwent IMT assessment by high-resolution B-mode ultrasonography and IL-13 measurement in serum by ELISA. RESULTS Subjects (41 M/38F) had a mean age of 81.0 ± 4.5 years and were mostly overweight. Stratifying the whole cohort by IMT thickness (IMT ≤ 0.9, n = 17; IMT ≥ 1 and ≤ 1.3, n = 50; IMT ≥ 1.4, n = 12) among the main variables explored, only BMI and triglycerides differed among groups, having subjects with higher IMT significantly higher BMI and lower triglycerides. Serum IL-13 levels significantly differed among groups having subjects with IMT ≥ 1.4 lower levels as compared to other groups (p < 0.0001). In all sample population, IMT values significantly correlate with IL-13 levels (r = - 0.454, p < 0.0001). Indeed, a linear regression analysis showed that independent of age, gender, body mass index, triglycerides, systolic blood pressure, statin use and smoking habit, lower IL-13 serum levels were associated with higher IMT values. CONCLUSIONS IL-13, an anti-inflammatory cytokine, may have a protective role in the human atherosclerotic process. It could be used as an effective and promising novel therapeutic target development.
Collapse
|
24
|
Chao R, Li D, Yue Z, Huang C, Kou Y, Zhou Q, Gao Y, Hasegawa T, Guo J, Li M. Interleukin-4 Restores Insulin Sensitivity in Insulin-Resistant Osteoblasts by Increasing the Expression of Insulin Receptor Substrate 1. BIOCHEMISTRY (MOSCOW) 2020; 85:334-343. [PMID: 32564738 DOI: 10.1134/s0006297920030098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Obesity and latent inflammation can give rise to insulin resistance and type 2 diabetes. Here we established an insulin resistance model of osteoblasts to explore the restoration effect of anti-inflammatory interleukin-4 (IL-4) on insulin sensitivity and its mechanism. We found that IL-4 inhibited cell proliferation in a concentration- and time-dependent manner. Insulation resistance significantly reduced the phosphorylation levels of the insulin receptor substrate 1 (IRS1; Tyr612), Akt (Ser473), and AS160 (Ser318) proteins. The addition of IL-4 to the insulin resistance model led to a dose-dependent stimulation of the phosphorylation of IRS1, Akt, and AS160. IL-4 fully restored the activation of the insulin cascade in insulin-resistant cells at the concentration of 50 ng/ml. Additionally, IL-4 promoted the expression of IRS1 in a time-dependent manner. We conjecture that IL-4 restores insulin sensitivity in osteoblasts by upregulating the expression of IRS1. It was also found that IL-4 promoted the expression of osteoprotegerin depending on the time of exposure. This effect may play an important role in the regulation of the energy metabolism in the whole body.
Collapse
Affiliation(s)
- R Chao
- Department of Bone Metabolism, School and Hospital of Stomatology, Shandong University; Shandong Key Laboratory of Oral Tissue Regeneration; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, 250012, China.
| | - D Li
- Department of Bone Metabolism, School and Hospital of Stomatology, Shandong University; Shandong Key Laboratory of Oral Tissue Regeneration; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, 250012, China
| | - Z Yue
- Department of Bone Metabolism, School and Hospital of Stomatology, Shandong University; Shandong Key Laboratory of Oral Tissue Regeneration; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, 250012, China
| | - C Huang
- Department of Bone Metabolism, School and Hospital of Stomatology, Shandong University; Shandong Key Laboratory of Oral Tissue Regeneration; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, 250012, China
| | - Y Kou
- Department of Bone Metabolism, School and Hospital of Stomatology, Shandong University; Shandong Key Laboratory of Oral Tissue Regeneration; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, 250012, China
| | - Q Zhou
- Department of Bone Metabolism, School and Hospital of Stomatology, Shandong University; Shandong Key Laboratory of Oral Tissue Regeneration; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, 250012, China
| | - Y Gao
- Department of Bone Metabolism, School and Hospital of Stomatology, Shandong University; Shandong Key Laboratory of Oral Tissue Regeneration; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, 250012, China
| | - T Hasegawa
- Department of Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586, Japan
| | - J Guo
- Department of Bone Metabolism, School and Hospital of Stomatology, Shandong University; Shandong Key Laboratory of Oral Tissue Regeneration; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, 250012, China
| | - M Li
- Department of Bone Metabolism, School and Hospital of Stomatology, Shandong University; Shandong Key Laboratory of Oral Tissue Regeneration; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, 250012, China.
| |
Collapse
|
25
|
Abstract
Adipose tissue (AT) plays a central role in both metabolic health and pathophysiology. Its expansion in obesity results in increased mortality and morbidity, with contributions to cardiovascular disease, diabetes mellitus, fatty liver disease, and cancer. Obesity prevalence is at an all-time high and is projected to be 50% in the United States by 2030. AT is home to a large variety of immune cells, which are critical to maintain normal tissue functions. For example, γδ T cells are fundamental for AT innervation and thermogenesis, and macrophages are required for recycling of lipids released by adipocytes. The expansion of visceral white AT promotes dysregulation of its immune cell composition and likely promotes low-grade chronic inflammation, which has been proposed to be the underlying cause for the complications of obesity. Interestingly, weight loss after obesity alters the AT immune compartment, which may account for the decreased risk of developing these complications. Recent technological advancements that allow molecular investigation on a single-cell level have led to the discovery of previously unappreciated heterogeneity in many organs and tissues. In this review, we will explore the heterogeneity of immune cells within the visceral white AT and their contributions to homeostasis and pathology.
Collapse
Affiliation(s)
- Ada Weinstock
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Hernandez Moura Silva
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY, 10016, USA
| | - Kathryn J. Moore
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Edward A. Fisher
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| |
Collapse
|
26
|
Banerjee A, Singh J. Remodeling adipose tissue inflammasome for type 2 diabetes mellitus treatment: Current perspective and translational strategies. Bioeng Transl Med 2020; 5:e10150. [PMID: 32440558 PMCID: PMC7237149 DOI: 10.1002/btm2.10150] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/07/2019] [Accepted: 12/03/2019] [Indexed: 12/14/2022] Open
Abstract
Obesity-associated type 2 diabetes mellitus (T2DM) is characterized by low-grade chronic systemic inflammation that arises primarily from the white adipose tissue. The interplay between various adipose tissue-derived chemokines drives insulin resistance in T2DM and has therefore become a subject of rigorous investigation. The adipocytokines strongly associated with glucose homeostasis include tumor necrosis factor-α, various interleukins, monocyte chemoattractant protein-1, adiponectin, and leptin, among others. Remodeling the adipose tissue inflammasome in obesity-associated T2DM is likely to treat the underlying cause of the disease and bring significant therapeutic benefit. Various strategies have been adopted or are being investigated to modulate the serum/tissue levels of pro- and anti-inflammatory adipocytokines to improve glucose homeostasis in T2DM. These include use of small molecule agonists/inhibitors, mimetics, antibodies, gene therapy, and other novel formulations. Here, we discuss adipocytokines that are strongly associated with insulin activity and therapies that are under investigation for modulation of their levels in the treatment of T2DM.
Collapse
Affiliation(s)
- Amrita Banerjee
- Department of Pharmaceutical SciencesNorth Dakota State UniversityFargoNorth Dakota
| | - Jagdish Singh
- Department of Pharmaceutical SciencesNorth Dakota State UniversityFargoNorth Dakota
| |
Collapse
|
27
|
Matsui S, Okabayashi K, Tsuruta M, Shigeta K, Seishima R, Ishida T, Kondo T, Suzuki Y, Hasegawa H, Shimoda M, Sugimoto S, Sato T, Kitagawa Y. Interleukin-13 and its signaling pathway is associated with obesity-related colorectal tumorigenesis. Cancer Sci 2019; 110:2156-2165. [PMID: 31099450 PMCID: PMC6609806 DOI: 10.1111/cas.14066] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 01/10/2023] Open
Abstract
The incidence of colorectal cancer (CRC) has been on the rise, which is linked to the increasing prevalence of obesity, based on global epidemiological evidence. Although chronic inflammation is implicated in tumor development, the mechanisms underlying obesity‐associated CRC remain unknown. Here, we sought to identify the inflammatory cytokines and their roles in obesity‐related colorectal tumorigenesis using cytokine array analyses in a mouse model. Colorectal tumorigenesis was induced through i.p. injection of azoxymethane once a week for 6 weeks in 6‐week‐old female WT C57Black/6J mice and the obesity diabetes model mouse KK/TaJcl, KK‐Ay/TaJcl. The formation of aberrant crypt foci and colorectal tumors were more frequent in obese mice compared with WT mice, and both serum interleukin (IL)‐13 and IL‐13 receptor (R) expression in the normal intestinal mucosal epithelium were significantly increased in the obese mice. Furthermore, addition of IL‐13 to a human CRC cell line and a human colon organoid culture altered the phenotype of intestinal epithelial cells. Knockdown experiments further revealed that IL‐13Rα1 dominantly induced mucosal proliferation. Collectively, These results suggest an association between anti‐inflammatory cytokines and colorectal carcinogenesis, and provide new research directions for cancer prevention strategies. In particular, inflammation provoked by obesity, notably by increased expression of the cytokine IL‐13, could play an important role in the carcinogenesis of obesity‐related CRC.
Collapse
Affiliation(s)
- Shimpei Matsui
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Koji Okabayashi
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masashi Tsuruta
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Kohei Shigeta
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Ryo Seishima
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Ishida
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Takayuki Kondo
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yoshiyuki Suzuki
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hirotoshi Hasegawa
- Department of Surgery, Tokyo Dental College Ichikawa General Hospital, Chiba, Japan
| | - Masayuki Shimoda
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Shinya Sugimoto
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Toshiro Sato
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
28
|
Miranda K, Mehrpouya-Bahrami P, Nagarkatti PS, Nagarkatti M. Cannabinoid Receptor 1 Blockade Attenuates Obesity and Adipose Tissue Type 1 Inflammation Through miR-30e-5p Regulation of Delta-Like-4 in Macrophages and Consequently Downregulation of Th1 Cells. Front Immunol 2019; 10:1049. [PMID: 31134094 PMCID: PMC6523050 DOI: 10.3389/fimmu.2019.01049] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/24/2019] [Indexed: 12/31/2022] Open
Abstract
Obesity is characterized by chronic low-grade inflammation that contributes to development of cardiometabolic disorders. Cannabinoid receptor 1 (CB1) antagonists attenuate diet-induced obesity (DIO) and related inflammation, although the precise anti-inflammatory mechanisms involved have not been fully explored. In the current study we used a mouse model of DIO intervention to determine the microRNA (miRNA, miR)-mediated anti-obesity and anti-inflammatory effects of the CB1 antagonist, AM251. DIO mice that were fed high-fat diet (HFD) for 12 weeks were treated with AM251 (10 mg/kg) for an additional 4 weeks. HFD + AM251 mice experienced rapid and prolonged weight loss and reduced inflammatory M1 adipose tissue macrophage (ATM) infiltration. To investigate miRNA-mediated regulation of ATMs, F4/80+ cells from stromal vascular fractions (SVF) of epididymal fat were subjected to miR microarray analysis. Several miRs were differentially expressed in AM251-treated mice that were independent of calorie restriction. Prominently, miR-30e-5p was upregulated in ATMs from HFD + AM251 mice while the miR-30e-5p target, DLL4, was downregulated. Consistent with a decrease in DLL4-Notch signaling, fat storage and pro-inflammatory cytokine/chemokine expression was reduced following AM251 treatment. Furthermore, we found that AM251-treated macrophages can suppress DLL4-mediated Th1 polarization in CD4+ T cells. Together these data demonstrate that blocking CB1 receptors leads to upregulation of miR-30e-5p and down regulation of DLL4 in ATMs, which in turn suppress DLL4-Notch signaling-induced polarization of inflammatory Th1 cells and adipocyte energy storage. This combined effect of ATMs and T cells leads to an anti-inflammatory state and attenuation of DIO. These data support therapeutic potential of miR-30 in the treatment of cardiometabolic disorders.
Collapse
Affiliation(s)
- Kathryn Miranda
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Pegah Mehrpouya-Bahrami
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Prakash S Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
29
|
Garneau L, Aguer C. Role of myokines in the development of skeletal muscle insulin resistance and related metabolic defects in type 2 diabetes. DIABETES & METABOLISM 2019; 45:505-516. [PMID: 30844447 DOI: 10.1016/j.diabet.2019.02.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/04/2019] [Accepted: 02/25/2019] [Indexed: 12/20/2022]
Abstract
Due to its mass, skeletal muscle is the major site of glucose uptake and an important tissue in the development of type 2 diabetes (T2D). Muscles of patients with T2D are affected with insulin resistance and mitochondrial dysfunction, which result in impaired glucose and fatty acid metabolism. A well-established method of managing the muscle metabolic defects occurring in T2D is physical exercise. During exercise, muscles contract and secrete factors called myokines which can act in an autocrine/paracrine fashion to improve muscle energy metabolism. In patients with T2D, plasma levels as well as muscle levels (mRNA and protein) of some myokines are upregulated, while others are downregulated. The signalling pathways of certain myokines are also altered in skeletal muscle of patients with T2D. Taken together, these findings suggest that myokine secretion is an important factor contributing to the development of muscle metabolic defects during T2D. It is also of interest considering that lack of physical activity is closely linked to the occurrence of this disease. The causal relationships between sedentary behavior, factors secreted by skeletal muscle at rest and during contraction and the development of T2D remain to be elucidated. Many myokines shown to influence muscle energy metabolism still have not been characterized in the context of T2D in skeletal muscle specifically. The purpose of this review is to highlight what is known and what remains to be determined regarding myokine secretion in patients with T2D to uncover potential therapeutic targets for the management of this disease.
Collapse
Affiliation(s)
- L Garneau
- University of Ottawa, Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, Ottawa, ON, K1H 8M5, Canada; Institut du Savoir Montfort - recherche, Ottawa, ON, K1K 0T2, Canada
| | - C Aguer
- University of Ottawa, Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, Ottawa, ON, K1H 8M5, Canada; Institut du Savoir Montfort - recherche, Ottawa, ON, K1K 0T2, Canada.
| |
Collapse
|
30
|
Sigrist-Flores SC, Ponciano-Gómez A, Pedroza-González A, Gallardo-Ortíz IA, Villalobos-Molina R, Pardo-Vázquez JP, Saucedo-Campos AD, Jiménez-Flores R, Méndez-Cruz AR. Chronic intake of moderate fat-enriched diet induces fatty liver and low-grade inflammation without obesity in rabbits. Chem Biol Interact 2019; 300:56-62. [PMID: 30639268 DOI: 10.1016/j.cbi.2019.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/06/2018] [Accepted: 01/02/2019] [Indexed: 12/20/2022]
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) is the cause of chronic liver disease. Even though NAFLD is strongly associated with obesity and metabolic syndrome, there is a proportion of patients who develop this condition in the absence of obesity and the underlying mechanisms are poorly understood. We investigated early events in the pathogenesis of non-obese NAFLD, analyzing the impact of the chronic intake of a moderate fat-enriched diet on hepatic lipid accumulation and their relationship with inflammation. Rabbits fed with a moderate Fatty-Acid- Enriched Diet 3% palmitic acid (FAED), were evaluated for body weight, biochemical parameters, and liver function. Liver samples were analyzed by histology and RT-qPCR to measure lipid accumulation, the expression of inflammation-related genes IL-1β, IL-6, IL-10, IL-13, IL-18, COX-2, TNF-α, and TLR-4. Chronic consumption by 6-months of FAED did not generate metabolic changes, but it induced fatty liver. We also observed the development of low-grade inflammation characterized by the up regulation of TNF-α, IL-13 and IL-18. The consumption by 12-months of FAED caused the overexpression of IL-6, IL-10, IL-13, COX-2, and TLR-4. We show that hepatic steatosis is an early consequence of fat-enriched diets, and that it is accompanied by an immune response that exerts protective effects that prevent the development of metabolic disorders, such as overweight/obesity and metabolic syndrome. However, the excessive intake of fatty acids renders these mechanisms less efficient for delaying the start of metabolic alterations. Rabbits fed with FAED can be used as a model of NAFLD in non-obese and obese groups, especially at early stages of the disease.
Collapse
Affiliation(s)
- S C Sigrist-Flores
- Laboratorio de Inmunología, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México Tlalnepantla, Estado de México, México
| | - A Ponciano-Gómez
- Laboratorio de Inmunología, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México Tlalnepantla, Estado de México, México
| | - A Pedroza-González
- Laboratorio de Inmunología, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México Tlalnepantla, Estado de México, México; Unidad de Biomedicina, Facultad de Estudios Superiores Izatacala, Universidad Nacional Autonoma de México, Tlalnepntla, Estado de México, México
| | - I A Gallardo-Ortíz
- Unidad de Biomedicina, Facultad de Estudios Superiores Izatacala, Universidad Nacional Autonoma de México, Tlalnepntla, Estado de México, México
| | - R Villalobos-Molina
- Unidad de Biomedicina, Facultad de Estudios Superiores Izatacala, Universidad Nacional Autonoma de México, Tlalnepntla, Estado de México, México; Carrera de Enfermeria, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México Tlalnepantla, Estado de México, México
| | - J P Pardo-Vázquez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, México
| | - A D Saucedo-Campos
- Laboratorio de Inmunología, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México Tlalnepantla, Estado de México, México
| | - R Jiménez-Flores
- Laboratorio de Inmunología, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México Tlalnepantla, Estado de México, México.
| | - A R Méndez-Cruz
- Laboratorio de Inmunología, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México Tlalnepantla, Estado de México, México.
| |
Collapse
|
31
|
Murphy KP, Hendley MA, Isely C, Annamalai P, Peña E, Gower RM. Resveratrol Delivery from Porous Poly(lactide- co-glycolide) Scaffolds Promotes an Anti-Inflammatory Environment within Visceral Adipose Tissue. ACS APPLIED MATERIALS & INTERFACES 2018; 10:43363-43374. [PMID: 30462474 PMCID: PMC7076954 DOI: 10.1021/acsami.8b13421] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
As biomaterial therapies emerge to address adipose tissue dysfunction that underlies metabolic disease, the immune response to these systems must be established. As a potential therapy, we are investigating resveratrol delivery from porous poly(lactide- co-glycolide) scaffolds designed to integrate with adipose tissue. Resveratrol was selected for its ability to protect mice and primates from high fat diet and broad anti-inflammatory properties. Herein, we report fabrication of scaffolds with high resveratrol loading that are stable and active for up to one year. In vitro release profiles indicate that drug release is biphasic with a burst release over 3 days followed by a plateau. Surprisingly, we find that PLG scaffolds implanted into adipose tissue of mice promote an anti-inflammatory environment characterized by high arginase-1 and low TNF-α and IL-6 compared to naïve unmanipulated fat. Resveratrol delivery from the scaffold augments this anti-inflammatory environment by decreasing monocyte and lymphocyte numbers at the implant site and increasing expression of IL-10 and IL-13, cytokines that promote healthy adipose tissue. In terms of therapeutic applications, implant of scaffolds designed to release resveratrol into the visceral fat decreases MCP-1 expression in mice fed a high fat diet, a molecule that drives both local and systemic inflammation during obesity. Taken together, resveratrol delivery to adipose tissue using poly(lactide- co-glycolide) scaffolds is a promising therapeutic strategy for the treatment of adipose tissue inflammation that drives metabolic disease.
Collapse
Affiliation(s)
- Kendall P. Murphy
- Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA
| | - Michael A. Hendley
- Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208, USA
| | - Christopher Isely
- Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA
| | - Prakasam Annamalai
- Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA
| | - Edsel Peña
- Department of Statistics, University of South Carolina, Columbia, SC 29208, USA
| | - R. Michael Gower
- Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA
- Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
32
|
Liu Y, Zheng J, Zhang HP, Zhang X, Wang L, Wood L, Wang G. Obesity-Associated Metabolic Signatures Correlate to Clinical and Inflammatory Profiles of Asthma: A Pilot Study. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2018; 10:628-647. [PMID: 30306746 PMCID: PMC6182193 DOI: 10.4168/aair.2018.10.6.628] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/27/2018] [Accepted: 06/01/2018] [Indexed: 02/05/2023]
Abstract
PURPOSE Obesity is associated with metabolic dysregulation, but the underlying metabolic signatures involving clinical and inflammatory profiles of obese asthma are largely unexplored. We aimed at identifying the metabolic signatures of obese asthma. METHODS Eligible subjects with obese (n = 11) and lean (n = 22) asthma underwent body composition and clinical assessment, sputum induction, and blood sampling. Sputum supernatant was assessed for interleukin (IL)-1β, -4, -5, -6, -13, and tumor necrosis factor (TNF)-α, and serum was detected for leptin, adiponectin and C-reactive protein. Untargeted gas chromatography time-of-flight mass spectrometry (GC-TOF-MS)-based metabolic profiles in sputum, serum and peripheral blood monocular cells (PBMCs) were analyzed by orthogonal projections to latent structures-discriminate analysis (OPLS-DA) and pathway topology enrichment analysis. The differential metabolites were further validated by correlation analysis with body composition, and clinical and inflammatory profiles. RESULTS Body composition, asthma control, and the levels of IL-1β, -4, -13, leptin and adiponectin in obese asthmatics were significantly different from those in lean asthmatics. OPLS-DA analysis revealed 28 differential metabolites that distinguished obese from lean asthmatic subjects. The validation analysis identified 18 potential metabolic signatures (11 in sputum, 4 in serum and 2 in PBMCs) of obese asthmatics. Pathway topology enrichment analysis revealed that cyanoamino acid metabolism, caffeine metabolism, alanine, aspartate and glutamate metabolism, phenylalanine, tyrosine and tryptophan biosynthesis, pentose phosphate pathway in sputum, and glyoxylate and dicarboxylate metabolism, glycerolipid metabolism and pentose phosphate pathway in serum are suggested to be significant pathways related to obese asthma. CONCLUSIONS GC-TOF-MS-based metabolomics indicates obese asthma is characterized by a metabolic profile different from lean asthma. The potential metabolic signatures indicated novel immune-metabolic mechanisms in obese asthma with providing more phenotypic and therapeutic implications, which needs further replication and validation.
Collapse
Affiliation(s)
- Ying Liu
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Zheng
- Department of Integrated Traditional Chinese and Western Medicine, Xinqiao Hospital, Third Military University, Chongqing, China
| | - Hong Ping Zhang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Zhang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Wang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lisa Wood
- Center for Asthma and Respiratory Diseases, Department of Respiratory and Sleep Medicine, John Hunter Hospital, Hunter Medical Research Institute, University of Newcastle, New Lambton, NSW, Australia
| | - Gang Wang
- Department of Respiratory and Critical Care Medicine, Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
33
|
Sánchez-Aguilera P, Diaz-Vegas A, Campos C, Quinteros-Waltemath O, Cerda-Kohler H, Barrientos G, Contreras-Ferrat A, Llanos P. Role of ABCA1 on membrane cholesterol content, insulin-dependent Akt phosphorylation and glucose uptake in adult skeletal muscle fibers from mice. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:1469-1477. [PMID: 30254016 DOI: 10.1016/j.bbalip.2018.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 08/23/2018] [Accepted: 09/17/2018] [Indexed: 02/07/2023]
Abstract
The ATP-binding cassette transporter A1 (ABCA1) promotes cellular cholesterol efflux, leading to cholesterol binding to the extracellular lipid-free apolipoprotein A-I. ABCA1 regulates lipid content, glucose tolerance and insulin sensitivity in adipose tissue. In skeletal muscle, most GLUT4-mediated glucose transport occurs in the transverse tubule, a system composed by specialized cholesterol-enriched invaginations of the plasma membrane. We have reported that insulin resistant mice have higher cholesterol levels in transverse tubule from adult skeletal muscle. These high levels correlate with decreased GLUT4 trafficking and glucose uptake; however, the role of ABCA1 on skeletal muscle insulin-dependent glucose metabolism remains largely unexplored. Here, we evaluated the functional role of the ABCA1 on insulin-dependent signaling pathways, glucose uptake and cellular cholesterol content in adult skeletal muscle. Male mice were fed for 8 weeks with normal chow diet (NCD) or high fat diet (HFD). Compared to NCD-fed mice, ABCA1 mRNA levels and protein content were lower in muscle homogenates from HFD-fed mice. In Flexor digitorum brevis muscle from NCD-fed mice, shABCA1-RFP in vivo electroporation resulted in 65% reduction of ABCA1 protein content, 1.6-fold increased fiber cholesterol levels, 74% reduction in insulin-dependent Akt (Ser473) phosphorylation, total suppression of insulin-dependent GLUT4 translocation and decreased 2-NBDG uptake compared to fibers electroporated with the scrambled plasmid. Pre-incubation with methyl-β cyclodextrin reestablished both GLUT4 translocation and 2-NBDG transport. Based on the present results, we suggest that decreased ABCA1 contributes to the anomalous cholesterol accumulation and decreased glucose transport displayed by skeletal muscle membranes in the insulin resistant condition.
Collapse
Affiliation(s)
- Pablo Sánchez-Aguilera
- Institute for Research in Dental Sciences, Facultad de Odontología, Universidad de Chile, Chile
| | - Alexis Diaz-Vegas
- Departamento Ciencias Biológicas, Facultad Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | | | | | - Hugo Cerda-Kohler
- Institute for Research in Dental Sciences, Facultad de Odontología, Universidad de Chile, Chile
| | | | - Ariel Contreras-Ferrat
- ACCDiS, Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Chile
| | - Paola Llanos
- Institute for Research in Dental Sciences, Facultad de Odontología, Universidad de Chile, Chile; CEMC, Facultad de Medicina, Universidad de Chile, Chile.
| |
Collapse
|
34
|
Stafeev IS, Michurina SS, Podkuychenko NV, Vorotnikov AV, Menshikov MY, Parfyonova YV. Interleukin-4 Restores Insulin Sensitivity in Lipid-Induced Insulin-Resistant Adipocytes. BIOCHEMISTRY (MOSCOW) 2018; 83:498-506. [PMID: 29738684 DOI: 10.1134/s0006297918050036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Obesity and latent inflammation in adipose tissue significantly contribute to the development of insulin resistance (IR) and type 2 diabetes. Here we studied whether the antiinflammatory interleukin-4 (IL-4) can restore insulin sensitivity in cultured 3T3-L1 adipocytes. The activity of key components of the insulin signaling cascade was assessed by immunoblotting using phospho-specific antibodies to insulin receptor substrate IRS1 (Tyr612), Akt (Thr308 and Ser473), and AS160 (Ser318) protein that regulates translocation of the GLUT4 glucose transporter to the plasma membrane. IR was induced in mature adipocytes with albumin-conjugated palmitate. IR significantly reduced phosphorylation levels of all the above-mentioned proteins. Addition of IL-4 to the culturing medium during IR induction led to a dose-dependent stimulation of the insulin-promoted phosphorylation of IRS1, Akt, and AS160. At the optimal concentration of 50 ng/ml, IL-4 fully restored activation of the insulin cascade in IR cells, but it did not affect insulin signaling activation in the control cells. IL-4 neither upregulated expression of key adipogenesis markers GLUT4 and PPARγ nor caused lipid accumulation in the adipocytes. These results demonstrate that IL-4 can restore insulin sensitivity in adipocytes via mechanisms not associated with induced adipogenesis or de novo formation of lipid depots.
Collapse
Affiliation(s)
- I S Stafeev
- Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Moscow, 121552, Russia. .,Lomonosov Moscow State University, Faculty of Basic Medicine, Moscow, 117192, Russia
| | - S S Michurina
- Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Moscow, 121552, Russia.,Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
| | - N V Podkuychenko
- Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Moscow, 121552, Russia.,Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
| | - A V Vorotnikov
- Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Moscow, 121552, Russia. .,Lomonosov Moscow State University Medical Center, Moscow, 119991, Russia
| | - M Yu Menshikov
- Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Ye V Parfyonova
- Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Moscow, 121552, Russia.,Lomonosov Moscow State University, Faculty of Basic Medicine, Moscow, 117192, Russia
| |
Collapse
|
35
|
Duffen J, Zhang M, Masek-Hammerman K, Nunez A, Brennan A, Jones JEC, Morin J, Nocka K, Kasaian M. Modulation of the IL-33/IL-13 Axis in Obesity by IL-13Rα2. THE JOURNAL OF IMMUNOLOGY 2018; 200:1347-1359. [DOI: 10.4049/jimmunol.1701256] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
In obesity, IL-13 overcomes insulin resistance by promoting anti-inflammatory macrophage differentiation in adipose tissue. Endogenous IL-13 levels can be modulated by the IL-13 decoy receptor, IL-13Rα2, which inactivates and depletes the cytokine. In this study, we show that IL-13Rα2 is markedly elevated in adipose tissues of obese mice. Mice deficient in IL-13Rα2 had high expression of IL-13 response markers in adipose tissue, consistent with increased IL-13 activity at baseline. Moreover, exposure to the type 2 cytokine-inducing alarmin, IL-33, enhanced serum and tissue IL-13 concentrations and elevated tissue eosinophils, macrophages, and type 2 innate lymphoid cells. IL-33 also reduced body weight, fat mass, and fasting blood glucose levels. Strikingly, however, the IL-33–induced protection was greater in IL-13Rα2–deficient mice compared with wild-type littermates, and these changes were largely attenuated in mice lacking IL-13. Although IL-33 administration improved the metabolic profile in the context of a high fat diet, it also resulted in diarrhea and perianal irritation, which was enhanced in the IL-13Rα2–deficient mice. Weight loss in this group was associated with reduced food intake, which was likely related to the gastrointestinal effects. These findings outline both potentially advantageous and deleterious effects of a type 2–skewed immune response under conditions of metabolic stress, and identify IL-13Rα2 as a critical checkpoint in adipose tissues that limits the protective effects of the IL-33/IL-13 axis in obesity.
Collapse
Affiliation(s)
- Jennifer Duffen
- *Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA 02139
| | - Melvin Zhang
- *Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA 02139
| | | | - Angela Nunez
- ‡Comparative Medicine, Pfizer, Inc., Andover, MA 01810; and
| | - Agnes Brennan
- *Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA 02139
| | | | - Jeffrey Morin
- ‡Comparative Medicine, Pfizer, Inc., Andover, MA 01810; and
| | - Karl Nocka
- *Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA 02139
| | - Marion Kasaian
- *Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA 02139
| |
Collapse
|
36
|
Kim JH, Lee E, Friedline RH, Suk S, Jung DY, Dagdeviren S, Hu X, Inashima K, Noh HL, Kwon JY, Nambu A, Huh JR, Han MS, Davis RJ, Lee AS, Lee KW, Kim JK. Endoplasmic reticulum chaperone GRP78 regulates macrophage function and insulin resistance in diet-induced obesity. FASEB J 2018; 32:2292-2304. [PMID: 29242277 DOI: 10.1096/fj.201701017r] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity-mediated inflammation is a major cause of insulin resistance, and macrophages play an important role in this process. The 78-kDa glucose-regulated protein (GRP78) is a major endoplasmic reticulum chaperone that modulates unfolded protein response (UPR), and mice with GRP78 heterozygosity were resistant to diet-induced obesity. Here, we show that mice with macrophage-selective ablation of GRP78 (Lyz- GRP78-/-) are protected from skeletal muscle insulin resistance without changes in obesity compared with wild-type mice after 9 wk of high-fat diet. GRP78-deficient macrophages demonstrated adapted UPR with up-regulation of activating transcription factor (ATF)-4 and M2-polarization markers. Diet-induced adipose tissue inflammation was reduced, and bone marrow-derived macrophages from Lyz- GRP78-/- mice demonstrated a selective increase in IL-6 expression. Serum IL-13 levels were elevated by >4-fold in Lyz- GRP78-/- mice, and IL-6 stimulated the myocyte expression of IL-13 and IL-13 receptor. Lastly, recombinant IL-13 acutely increased glucose metabolism in Lyz- GRP78-/- mice. Taken together, our data indicate that GRP78 deficiency activates UPR by increasing ATF-4, and promotes M2-polarization of macrophages with a selective increase in IL-6 secretion. Macrophage-derived IL-6 stimulates the myocyte expression of IL-13 and regulates muscle glucose metabolism in a paracrine manner. Thus, our findings identify a novel crosstalk between macrophages and skeletal muscle in the modulation of obesity-mediated insulin resistance.-Kim, J. H., Lee, E., Friedline, R. H., Suk, S., Jung, D. Y., Dagdeviren, S., Hu, X., Inashima, K., Noh, H. L., Kwon, J. Y., Nambu, A., Huh, J. R., Han, M. S., Davis, R. J., Lee, A. S., Lee, K. W., Kim, J. K. Endoplasmic reticulum chaperone GRP78 regulates macrophage function and insulin resistance in diet-induced obesity.
Collapse
Affiliation(s)
- Jong Hun Kim
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA.,Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
| | - Eunjung Lee
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA.,Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
| | - Randall H Friedline
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Sujin Suk
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA.,Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
| | - Dae Young Jung
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Sezin Dagdeviren
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Xiaodi Hu
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Kunikazu Inashima
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Hye Lim Noh
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jung Yeon Kwon
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Aya Nambu
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jun R Huh
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Myoung Sook Han
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Amy S Lee
- Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Ki Won Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea.,Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea.,Wellness Emergence Center, Advanced Institutes of Convergence Technology, Seoul National University, Suwon, South Korea
| | - Jason K Kim
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA.,Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea.,Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
37
|
Martínez-Reyes CP, Gómez-Arauz AY, Torres-Castro I, Manjarrez-Reyna AN, Palomera LF, Olivos-García A, Mendoza-Tenorio E, Sánchez-Medina GA, Islas-Andrade S, Melendez-Mier G, Escobedo G. Serum Levels of Interleukin-13 Increase in Subjects with Insulin Resistance but Do Not Correlate with Markers of Low-Grade Systemic Inflammation. J Diabetes Res 2018; 2018:7209872. [PMID: 29675435 PMCID: PMC5841096 DOI: 10.1155/2018/7209872] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/07/2017] [Accepted: 12/18/2017] [Indexed: 12/29/2022] Open
Abstract
Experimental evidence in mice suggests a role for interleukin- (IL-) 13 in insulin resistance and low-grade systemic inflammation. However, IL-13 serum levels have not been assessed in subjects with insulin resistance, and associations of IL-13 with parameters of low-grade systemic inflammation are still unknown. Our main goal was to examine the systemic levels of IL-13 in patients with insulin resistance, while also studying the relationship of IL-13 with anthropometric, metabolic, and low-grade systemic inflammatory markers. Ninety-two participants were included in the study and divided into insulin-resistant patients and noninsulin-resistant controls. Blood levels of IL-13, glucose, insulin, triglycerides, cholesterol, tumor necrosis factor-alpha (TNF-α), IL-10, proinflammatory (Mon-CD11c+CD206-), and anti-inflammatory (Mon-CD11c-CD206+) monocytes, as well as anthropometric parameters, were measured in all volunteers. Insulin-resistant patients showed 2.5-fold higher serum levels of IL-13 than controls (P < 0.0001) and significantly increased values of TNF-α and Mon-CD11c+CD206-, with concomitant reductions in IL-10 and Mon-CD11c-CD206+. Increased IL-13 was extraordinarily well associated with hyperglycemia (r = 0.7362) and hypertriglyceridemia (r = 0.7632) but unexpectedly exhibited no significant correlations with TNF-α (r = 0.2907), IL-10 (r = -0.3882), Mon-CD11c+CD206- (r = 0.2745) or Mon-CD11c-CD206+ (r = -0.3237). This study demonstrates that IL-13 serum levels are elevated in patients with insulin resistance without showing correlation with parameters of low-grade systemic inflammation.
Collapse
Affiliation(s)
- Camilo P. Martínez-Reyes
- Unit of Experimental Medicine, School of Medicine, National University of Mexico, General Hospital of Mexico “Dr. Eduardo Liceaga”, 06726 Mexico City, Mexico
| | - Angélica Y. Gómez-Arauz
- Unit of Experimental Medicine, School of Medicine, National University of Mexico, General Hospital of Mexico “Dr. Eduardo Liceaga”, 06726 Mexico City, Mexico
| | - Israel Torres-Castro
- Unit of Experimental Medicine, School of Medicine, National University of Mexico, General Hospital of Mexico “Dr. Eduardo Liceaga”, 06726 Mexico City, Mexico
| | - Aarón N. Manjarrez-Reyna
- Unit of Experimental Medicine, School of Medicine, National University of Mexico, General Hospital of Mexico “Dr. Eduardo Liceaga”, 06726 Mexico City, Mexico
| | - León F. Palomera
- Unit of Experimental Medicine, School of Medicine, National University of Mexico, General Hospital of Mexico “Dr. Eduardo Liceaga”, 06726 Mexico City, Mexico
| | - Alfonso Olivos-García
- Departamento de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 México, DF, Mexico
| | - Edith Mendoza-Tenorio
- Departamento de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 México, DF, Mexico
| | | | - Sergio Islas-Andrade
- Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, 06726 Mexico City, Mexico
| | - Guillermo Melendez-Mier
- Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, 06726 Mexico City, Mexico
| | - Galileo Escobedo
- Unit of Experimental Medicine, School of Medicine, National University of Mexico, General Hospital of Mexico “Dr. Eduardo Liceaga”, 06726 Mexico City, Mexico
| |
Collapse
|
38
|
Loss of microRNA-22 prevents high-fat diet induced dyslipidemia and increases energy expenditure without affecting cardiac hypertrophy. Clin Sci (Lond) 2017; 131:2885-2900. [PMID: 29101298 DOI: 10.1042/cs20171368] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 12/17/2022]
Abstract
Obesity is associated with development of diverse diseases, including cardiovascular diseases and dyslipidemia. MiRNA-22 (miR-22) is a critical regulator of cardiac function and targets genes involved in metabolic processes. Previously, we generated miR-22 null mice and we showed that loss of miR-22 blunted cardiac hypertrophy induced by mechanohormornal stress. In the present study, we examined the role of miR-22 in the cardiac and metabolic alterations promoted by high-fat (HF) diet. We found that loss of miR-22 attenuated the gain of fat mass and prevented dyslipidemia induced by HF diet, although the body weight gain, or glucose intolerance and insulin resistance did not seem to be affected. Mechanistically, loss of miR-22 attenuated the increased expression of genes involved in lipogenesis and inflammation mediated by HF diet. Similarly, we found that miR-22 mediates metabolic alterations and inflammation induced by obesity in the liver. However, loss of miR-22 did not appear to alter HF diet induced cardiac hypertrophy or fibrosis in the heart. Our study therefore establishes miR-22 as an important regulator of dyslipidemia and suggests it may serve as a potential candidate in the treatment of dyslipidemia associated with obesity.
Collapse
|
39
|
Hotamisligil GS. Foundations of Immunometabolism and Implications for Metabolic Health and Disease. Immunity 2017; 47:406-420. [PMID: 28930657 DOI: 10.1016/j.immuni.2017.08.009] [Citation(s) in RCA: 318] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/06/2017] [Accepted: 08/16/2017] [Indexed: 02/06/2023]
Abstract
Highly ordered interactions between immune and metabolic responses are evolutionarily conserved and paramount for tissue and organismal health. Disruption of these interactions underlies the emergence of many pathologies, particularly chronic non-communicable diseases such as obesity and diabetes. Here, we examine decades of research identifying the complex immunometabolic signaling networks and the cellular and molecular events that occur in the setting of altered nutrient and energy exposures and offer a historical perspective. Furthermore, we describe recent advances such as the discovery that a broad complement of immune cells play a role in immunometabolism and the emerging evidence that nutrients and metabolites modulate inflammatory pathways. Lastly, we discuss how this work may eventually lead to tangible therapeutic advancements to promote health.
Collapse
Affiliation(s)
- Gökhan S Hotamisligil
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Broad Institute of Harvard and MIT, Boston, MA 02115, USA.
| |
Collapse
|
40
|
BITC and S-Carvone Restrain High-Fat Diet-Induced Obesity and Ameliorate Hepatic Steatosis and Insulin Resistance. Pharm Res 2017; 34:2241-2249. [PMID: 28733781 DOI: 10.1007/s11095-017-2230-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/10/2017] [Indexed: 12/17/2022]
Abstract
PURPOSE To investigate the preventative activity of benzyl isothiocyante and S-carvone against high-fat diet-induced obesity and metabolic complications. METHODS Ten-week-old C57BL/6 male mice were fed a high-fat diet and injected intraperitoneally twice per week with benzyl isothiocyante, S-carvone, or vehicle for 8 weeks. The body weight, food intake, and body composition were monitored, and glucose tolerance and insulin tolerance tests were performed at the end of the experiment. Serum and tissue samples were studied using serum biochemistry, histological, and gene expression analysis to define the effects of benzyl isothiocyante and S-carvone treatments on lipid and glucose metabolism and inflammatory responses. RESULTS Benzyl isothiocyante and S-carvone blocked high-fat diet-induced weight gain, fat accumulation in the liver, and insulin resistance. The beneficial effects were found to be associated with an improvement of expression of macrophage marker genes in white adipose tissue, including F4/80, Cd11b, Cd11c, Cd206, and Tnf-α, and reduced expression of genes (Pparγ2, Scd1, Cd36) responsible for lipid synthesis and transport in the liver. CONCLUSION Benzyl isothiocyante and S-carvone block high-fat diet-induced obesity and metabolism disorders and can be considered for management of the obesity epidemic that affects approximately 36% of adults and 17% of children in the USA.
Collapse
|
41
|
Geisler CE, Renquist BJ. Hepatic lipid accumulation: cause and consequence of dysregulated glucoregulatory hormones. J Endocrinol 2017; 234:R1-R21. [PMID: 28428362 DOI: 10.1530/joe-16-0513] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 04/20/2017] [Indexed: 12/11/2022]
Abstract
Fatty liver can be diet, endocrine, drug, virus or genetically induced. Independent of cause, hepatic lipid accumulation promotes systemic metabolic dysfunction. By acting as peroxisome proliferator-activated receptor (PPAR) ligands, hepatic non-esterified fatty acids upregulate expression of gluconeogenic, beta-oxidative, lipogenic and ketogenic genes, promoting hyperglycemia, hyperlipidemia and ketosis. The typical hormonal environment in fatty liver disease consists of hyperinsulinemia, hyperglucagonemia, hypercortisolemia, growth hormone deficiency and elevated sympathetic tone. These endocrine and metabolic changes further encourage hepatic steatosis by regulating adipose tissue lipolysis, liver lipid uptake, de novo lipogenesis (DNL), beta-oxidation, ketogenesis and lipid export. Hepatic lipid accumulation may be induced by 4 separate mechanisms: (1) increased hepatic uptake of circulating fatty acids, (2) increased hepatic de novo fatty acid synthesis, (3) decreased hepatic beta-oxidation and (4) decreased hepatic lipid export. This review will discuss the hormonal regulation of each mechanism comparing multiple physiological models of hepatic lipid accumulation. Nonalcoholic fatty liver disease (NAFLD) is typified by increased hepatic lipid uptake, synthesis, oxidation and export. Chronic hepatic lipid signaling through PPARgamma results in gene expression changes that allow concurrent activity of DNL and beta-oxidation. The importance of hepatic steatosis in driving systemic metabolic dysfunction is highlighted by the common endocrine and metabolic disturbances across many conditions that result in fatty liver. Understanding the mechanisms underlying the metabolic dysfunction that develops as a consequence of hepatic lipid accumulation is critical to identifying points of intervention in this increasingly prevalent disease state.
Collapse
Affiliation(s)
- Caroline E Geisler
- School of Animal and Comparative Biomedical SciencesUniversity of Arizona, Tucson, Arizona, USA
| | - Benjamin J Renquist
- School of Animal and Comparative Biomedical SciencesUniversity of Arizona, Tucson, Arizona, USA
| |
Collapse
|
42
|
Calton EK, Keane KN, Newsholme P, Zhao Y, Soares MJ. The impact of cholecalciferol supplementation on the systemic inflammatory profile: a systematic review and meta-analysis of high-quality randomized controlled trials. Eur J Clin Nutr 2017; 71:931-943. [PMID: 28488684 DOI: 10.1038/ejcn.2017.67] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 03/10/2017] [Accepted: 04/05/2017] [Indexed: 01/21/2023]
Abstract
Causal links between vitamin D status [25(OH)D] and systemic inflammation were examined through a systematic review of randomized controlled trials (RCTs). Selected RCTs were ⩾12 weeks, conducted in adults free of acute inflammatory disease, and of high-quality (Jadad score ⩾3). Of 14 studies that met our criteria, 9 studies (15 study arms) permitted extraction of data. There was no effect on the weighted mean difference (WMD) of IL-6 (WMD (95% confidence interval)=0.1, (-0.166, 0.366) pg/ml, P=0.462) or C-reactive protein (CRP) (WMD=-0.324, (-1.007, 0.359) mg/l, P=0.352). Subgroup analyses of trials achieving ⩾80 nmol/l indicated a trend for lower CRP (WMD=-0.834, (-1.726, 0.058) mg/l, P=0.067), however heterogeneity was significant (I2=66.7%, P=0.017). Studies employing a low dose (<1000 IU/d) showed increased CRP (WMD=0.615, (0.132, 1.098), P=0.013). In contrast, ⩾1000 IU/d had a favourable effect on CRP (WMD=-0.939, (-1.805, -0.073), P=0.034) but heterogeneity was significant (I2=61.3%, P=0.017). Meta-regression indicated that older age predicted a significant decrease in IL-6 (β=-0.02, (-0.034, -0.006) pg/ml, P=0.013) and CRP (β=-0.06, (-0.103, -0.017), P=0.01), whereas a greater percentage of females (β=0.027, (0.011, 0.044), P=0.004) and longer study duration independently predicted a higher WMD for CRP (β=0.049, (0.018, 0.079), P=0.005). Available high-quality RCTs did not support a beneficial effect of cholecalciferol on systemic IL-6 and CRP. Future studies should consider the confounding effects of age, gender and study duration, while possibly targeting an achieved 25(OH)D ⩾80 nmol/l.
Collapse
Affiliation(s)
- E K Calton
- Food, Nutrition &Health, School of Public Health, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - K N Keane
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - P Newsholme
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Y Zhao
- Occupation and the Environment, School of Public Health, Curtin University, Perth, WA, Australia
| | - M J Soares
- Food, Nutrition &Health, School of Public Health, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| |
Collapse
|
43
|
Surendar J, Indulekha K, Hoerauf A, Hübner MP. Immunomodulation by helminths: Similar impact on type 1 and type 2 diabetes? Parasite Immunol 2017; 39. [PMID: 27862000 DOI: 10.1111/pim.12401] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/04/2016] [Indexed: 12/19/2022]
Abstract
The incidence of both type 1 (T1D) and type 2 diabetes (T2D) is drastically increasing, and it is predicted that the global prevalence of diabetes will reach almost 600 million cases by 2035. Even though the pathogenesis of both types of diabetes is distinct, the immune system is actively involved in both forms of the disease. Genetic and environmental factors determine the risk to develop T1D. On the other hand, sedentary life style, surplus of food intake and other lifestyle changes contribute to the increase of T2D incidence. Improved sanitation with high-quality medical treatment is such an environmental factor that has led to a continuous reduction of infectious diseases including helminth infections over the past decades. Recently, a growing body of evidence has implicated a negative association between helminth infections and diabetes in humans as well as animal models. In this review, we discuss studies that have provided evidence for the beneficial impact of helminth infections on T1D and T2D. Possible mechanisms are presented by which helminths prevent T1D onset by mitigating pancreatic inflammation and confer protection against T2D by improving insulin sensitivity, alleviating inflammation, augmenting browning of adipose tissue and improving lipid metabolism and insulin signalling.
Collapse
Affiliation(s)
- J Surendar
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
| | - K Indulekha
- LIMES Institute, Membrane Biology & Lipid Biochemistry, University of Bonn, Bonn, Germany
| | - A Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - M P Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
| |
Collapse
|
44
|
Shea-Donohue T, Qin B, Smith A. Parasites, nutrition, immune responses and biology of metabolic tissues. Parasite Immunol 2017; 39. [PMID: 28235148 DOI: 10.1111/pim.12422] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 02/21/2017] [Indexed: 02/06/2023]
Abstract
Nutritional immunology, immunometabolism and identification of novel immunotherapeutic targets are areas of active investigation in parasitology. There is a well-documented crosstalk among immune cells and cells in metabolically active tissues that is important for homeostasis. The numbers and function of these cells are altered by obesity leading to inflammation. A variety of helminths spend some part of their life cycle in the gastrointestinal tract and even entirely enteral nematode infections exert beneficial effects on glucose and lipid metabolism. The foundation of this review is the ability of enteric nematode infections to improve obesity-induced type 2 diabetes and the metabolic syndrome, which are significant health issues in developed areas. It considers the impact of nutrition and specific nutritional deficiencies, which are occur in both undeveloped and developed areas, on the host's ability mount a protective immune response against parasitic nematodes. There are a number of proposed mechanisms by which parasitic nematodes can impact metabolism including effects gastrointestinal hormones, altering epithelial function and changing the number and/or phenotype of immune cells in metabolic tissues. Nematodes can also exert their beneficial effects through Th2 cytokines that activate the transcription factor STAT6, which upregulates genes that regulate glucose and lipid metabolism.
Collapse
Affiliation(s)
- T Shea-Donohue
- Department of Radiation Oncology & Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - B Qin
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD, USA
| | - A Smith
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD, USA
| |
Collapse
|