1
|
Joshi AA, Hegde MV, Zanwar AA. Potential of flaxseed oil blends to modulate tissue fatty acid composition and determination of safety parameters in Wistar rats. Lipids 2025; 60:125-141. [PMID: 39797657 DOI: 10.1002/lipd.12431] [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: 04/02/2024] [Revised: 11/26/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025]
Abstract
Non-communicable diseases (NCD) are associated with inflammation and oxidative stress which is further associated with omega-6 (ω6) and omega-3 (ω3) fatty acid (FA) imbalance favoring ω6 FA. By improving ω3 FA consumption, this imbalance can be altered to control NCD. Previously we have reported blends of flaxseed oil (FSO, ω3 FA) with palm olein (PO) or coconut oil (CO) were thermo-oxidatively stable with good storage stability and could improve ω6:ω3 ratio in cell lines. In the present study safety of these blends along with their efficacy to improve tissue FA composition particularly ω6:ω3 ratio was evaluated in Wistar rats. Institutional ethics committee approval was obtained initially. Wistar rats were supplemented with individual oils or blends (FSO with PO or CO, 20:80 by volume, 1.0 mL/day/200 gm body weight) for 3 months. Throughout the study period, there were no adverse effect of blends on feed intake and body weight gain. After 3 months, blood and serum were subjected for hematological, biochemical assessment. Vital organs were harvested for histopathological and FA composition investigations. Hematological, biochemical, and tissue histopathological parameters were comparable with Control (group receiving only normal diet). Interestingly serum lipid profile was improved by the blend supplementation. Except brain, FA composition was altered in liver, heart, adipose tissue, and RBC with lowering of ω6:ω3 ratio but there was no favorable effect on inflammatory markers and adipokines in the blend supplemented groups. Thus, to conclude, FSO blends with PO or CO were able to lower tissue ω6:ω3 ratio without adverse effects.
Collapse
Affiliation(s)
- Asavari Anirudha Joshi
- Centre for Innovation in Nutrition Health Disease, Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Mahabaleshwar Vishnu Hegde
- Centre for Innovation in Nutrition Health Disease, Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Anand Arvind Zanwar
- Centre for Innovation in Nutrition Health Disease, Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune, India
| |
Collapse
|
2
|
Shang D, Zhao S. Molecular mechanisms of obesity predisposes to atopic dermatitis. Front Immunol 2024; 15:1473105. [PMID: 39564133 PMCID: PMC11574713 DOI: 10.3389/fimmu.2024.1473105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/15/2024] [Indexed: 11/21/2024] Open
Abstract
Obesity is a prevalent metabolic disease that reduces bacterial diversity, colonizes the epidermis with lipophilic bacteria, and increases intestinal pro-inflammatory species, all of which lead to impaired epithelial barriers. Adipose tissue secretes immunomodulatory molecules, such as adipokines, leptin, and adiponectin, which alters the morphology of adipocytes and macrophages as well as modulates T cell differentiation and peripheral Th2-dominated immune responses. Atopic dermatitis (AD) and obesity have similar pathological manifestations, including inflammation as well as insulin and leptin resistance. This review examines the major mechanisms between obesity and AD, which focus on the effect on skin and gut microbiota, immune responses mediated by the toll like receptor (TLR) signaling pathway, and changes in cytokine levels (TNF-a, IL-6, IL-4, and IL13). Moreover, we describe the potential effects of adipokines on AD and finally mechanisms by which PPAR-γ suppresses and regulates type 2 immunity.
Collapse
Affiliation(s)
- Dajin Shang
- School of China Medical University, Shenyang, Liaoning, China
| | - Shengnan Zhao
- School of China Medical University, Shenyang, Liaoning, China
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
3
|
Baral A, Park PH. Interleukin-1β Signaling Contributes to Cell Cycle Arrest and Apoptotic Cell Death by Leptin via Modulation of AKT and p38MAPK in Hepatocytes. Biomol Ther (Seoul) 2024; 32:611-626. [PMID: 39091024 PMCID: PMC11392659 DOI: 10.4062/biomolther.2023.232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/17/2024] [Accepted: 02/06/2024] [Indexed: 08/04/2024] Open
Abstract
Leptin, an adipose tissue-derived hormone, has exhibited the potent hepatotoxic effects. However, the underlying molecular mechanisms are not fully understood. In this study, we have elucidated the mechanisms by which leptin exerts cytotoxic effects in hepatocytes, particularly focusing on the role of interleukin-1β (IL-1β) signaling. Leptin significantly induced maturation and secretion of IL-1β in cultured rat hepatocytes. Interestingly, inhibition of IL-1β signaling by pretreatment with an IL-1 receptor antagonist (IL-1Ra) or gene silencing of type I IL-1 receptor (IL-1R1) markedly abrogated leptin-induced cell cycle arrest. The critical role of IL-1β signaling in leptin-induced cell cycle arrest is mediated via upregulation of p16, which acts as an inhibitor of cyclin-dependent kinase. In addition, leptin-induced apoptotic cell death was relieved by inhibition of IL-1β signaling, as determined by annexin V/7-AAD binding assay. Mechanistically, IL-1β signaling contributes to apoptotic cell death and cell cycle arrest by suppressing AKT and activation of p38 mitogen-activated protein kinase (p38MAPK) signaling pathways. Involvement of IL-1β signaling in cytotoxic effect of leptin was further confirmed in vivo using hepatocyte specific IL-1R1 knock out (IL-1R1 KO) mice. Essentially similar results were obtained in vivo, where leptin administration caused the upregulation of apoptotic markers, dephosphorylation of AKT, and p38MAPK activation were observed in wild type mice liver without significant effects in the livers of IL-1R1 KO mice. Taken together, these results demonstrate that IL-1β signaling critically contributes to leptin-induced cell cycle arrest and apoptosis, at least in part, by modulating p38MAPK and AKT signaling pathways.
Collapse
Affiliation(s)
- Ananda Baral
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
4
|
Turpin T, Thouvenot K, Gonthier MP. Adipokines and Bacterial Metabolites: A Pivotal Molecular Bridge Linking Obesity and Gut Microbiota Dysbiosis to Target. Biomolecules 2023; 13:1692. [PMID: 38136564 PMCID: PMC10742113 DOI: 10.3390/biom13121692] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/13/2023] [Accepted: 11/19/2023] [Indexed: 12/24/2023] Open
Abstract
Adipokines are essential mediators produced by adipose tissue and exert multiple biological functions. In particular, adiponectin, leptin, resistin, IL-6, MCP-1 and PAI-1 play specific roles in the crosstalk between adipose tissue and other organs involved in metabolic, immune and vascular health. During obesity, adipokine imbalance occurs and leads to a low-grade pro-inflammatory status, promoting insulin resistance-related diabetes and its vascular complications. A causal link between obesity and gut microbiota dysbiosis has been demonstrated. The deregulation of gut bacteria communities characterizing this dysbiosis influences the synthesis of bacterial substances including lipopolysaccharides and specific metabolites, generated via the degradation of dietary components, such as short-chain fatty acids, trimethylamine metabolized into trimethylamine-oxide in the liver and indole derivatives. Emerging evidence suggests that these bacterial metabolites modulate signaling pathways involved in adipokine production and action. This review summarizes the current knowledge about the molecular links between gut bacteria-derived metabolites and adipokine imbalance in obesity, and emphasizes their roles in key pathological mechanisms related to oxidative stress, inflammation, insulin resistance and vascular disorder. Given this interaction between adipokines and bacterial metabolites, the review highlights their relevance (i) as complementary clinical biomarkers to better explore the metabolic, inflammatory and vascular complications during obesity and gut microbiota dysbiosis, and (ii) as targets for new antioxidant, anti-inflammatory and prebiotic triple action strategies.
Collapse
Affiliation(s)
| | | | - Marie-Paule Gonthier
- Université de La Réunion, INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), 97410 Saint-Pierre, La Réunion, France; (T.T.); (K.T.)
| |
Collapse
|
5
|
Phospholipase D1 Attenuation Therapeutics Promotes Resilience against Synaptotoxicity in 12-Month-Old 3xTg-AD Mouse Model of Progressive Neurodegeneration. Int J Mol Sci 2023; 24:ijms24043372. [PMID: 36834781 PMCID: PMC9967100 DOI: 10.3390/ijms24043372] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Abrogating synaptotoxicity in age-related neurodegenerative disorders is an extremely promising area of research with significant neurotherapeutic implications in tauopathies including Alzheimer's disease (AD). Our studies using human clinical samples and mouse models demonstrated that aberrantly elevated phospholipase D1 (PLD1) is associated with amyloid beta (Aβ) and tau-driven synaptic dysfunction and underlying memory deficits. While knocking out the lipolytic PLD1 gene is not detrimental to survival across species, elevated expression is implicated in cancer, cardiovascular conditions and neuropathologies, leading to the successful development of well-tolerated mammalian PLD isoform-specific small molecule inhibitors. Here, we address the importance of PLD1 attenuation, achieved using repeated 1 mg/kg of VU0155069 (VU01) intraperitoneally every alternate day for a month in 3xTg-AD mice beginning only from ~11 months of age (with greater influence of tau-driven insults) compared to age-matched vehicle (0.9% saline)-injected siblings. A multimodal approach involving behavior, electrophysiology and biochemistry corroborate the impact of this pre-clinical therapeutic intervention. VU01 proved efficacious in preventing in later stage AD-like cognitive decline affecting perirhinal cortex-, hippocampal- and amygdala-dependent behaviors. Glutamate-dependent HFS-LTP and LFS-LTD improved. Dendritic spine morphology showed the preservation of mushroom and filamentous spine characteristics. Differential PLD1 immunofluorescence and co-localization with Aβ were noted.
Collapse
|
6
|
Sahu B, Bal NC. Adipokines from white adipose tissue in regulation of whole body energy homeostasis. Biochimie 2023; 204:92-107. [PMID: 36084909 DOI: 10.1016/j.biochi.2022.09.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/08/2022] [Accepted: 09/01/2022] [Indexed: 02/06/2023]
Abstract
Diseases originating from altered energy homeostasis including obesity, and type 2 diabetes are rapidly increasing worldwide. Research in the last few decades on animal models and humans demonstrates that the white adipose tissue (WAT) is critical for energy balance and more than just an energy storage site. WAT orchestrates the whole-body metabolism through inter-organ crosstalk primarily mediated by cytokines named "Adipokines". The adipokines influence metabolism and fuel selection of the skeletal muscle and liver thereby fine-tuning the load on WAT itself in physiological conditions like starvation, exercise and cold. In addition, adipokine secretion is influenced by various pathological conditions like obesity, inflammation and diabetes. In this review, we have surveyed the current state of knowledge on important adipokines and their significance in regulating energy balance and metabolic diseases. Furthermore, we have summarized the interplay of pro-inflammatory and anti-inflammatory adipokines in the modulation of pathological conditions.
Collapse
Affiliation(s)
- Bijayashree Sahu
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India.
| | - Naresh C Bal
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
7
|
Jiménez-Cortegana C, Hontecillas-Prieto L, García-Domínguez DJ, Zapata F, Palazón-Carrión N, Sánchez-León ML, Tami M, Pérez-Pérez A, Sánchez-Jiménez F, Vilariño-García T, de la Cruz-Merino L, Sánchez-Margalet V. Obesity and Risk for Lymphoma: Possible Role of Leptin. Int J Mol Sci 2022; 23:15530. [PMID: 36555171 PMCID: PMC9779026 DOI: 10.3390/ijms232415530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Obesity, which is considered a pandemic due to its high prevalence, is a risk factor for many types of cancers, including lymphoma, through a variety of mechanisms by promoting an inflammatory state. Specifically, over the last few decades, obesity has been suggested not only to increase the risk of lymphoma but also to be associated with poor clinical outcomes and worse responses to different treatments for those diseases. Within the extensive range of proinflammatory mediators that adipose tissue releases, leptin has been demonstrated to be a key adipokine due to its pleotropic effects in many physiological systems and diseases. In this sense, different studies have analyzed leptin levels and leptin/leptin receptor expressions as a probable bridge between obesity and lymphomas. Since both obesity and lymphomas are prevalent pathophysiological conditions worldwide and their incidences have increased over the last few years, here we review the possible role of leptin as a promising proinflammatory mediator promoting lymphomas.
Collapse
Affiliation(s)
- Carlos Jiménez-Cortegana
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY 10065, USA
- Oncology Service, Department of Medicines, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Lourdes Hontecillas-Prieto
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Department of Medicines, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Daniel J. García-Domínguez
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Department of Medicines, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Fernando Zapata
- Oncology Service, Department of Medicines, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Natalia Palazón-Carrión
- Oncology Service, Department of Medicines, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - María L. Sánchez-León
- Oncology Service, Department of Medicines, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Malika Tami
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Department of Medicines, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Antonio Pérez-Pérez
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Flora Sánchez-Jiménez
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Teresa Vilariño-García
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Luis de la Cruz-Merino
- Oncology Service, Department of Medicines, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| |
Collapse
|
8
|
Chen AC, Fang TJ, Ho HH, Chen JF, Kuo YW, Huang YY, Tsai SY, Wu SF, Lin HC, Yeh YT. A multi-strain probiotic blend reshaped obesity-related gut dysbiosis and improved lipid metabolism in obese children. Front Nutr 2022; 9:922993. [PMID: 35990345 PMCID: PMC9386160 DOI: 10.3389/fnut.2022.922993] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/01/2022] [Indexed: 11/15/2022] Open
Abstract
Background and aims Obese children are more prone to becoming obese adults, and excess adiposity consequently increases the risk of many complications, such as metabolic syndromes, non-alcoholic fatty liver disease, cardiovascular disease, etc. This study aimed to evaluate the effects of multi-strain probiotics on the gut microbiota and weight control in obese children. Methods A double-blind, randomized, placebo-controlled trial was carried out on overweight and obese children. Subjects received 12 weeks of treatment with supplementary probiotics that contained three strains: Lactobacillus salivarius AP-32, L. rhamnosus bv-77, and Bifidobacterium animalis CP-9, plus diet and exercise guidance. A total of 82 children were enrolled, and 53 children completed the study. Results The supplementation of multi-strain probiotics resulted in a significant effect demonstrating high-density lipoprotein (HDL) and adiponectin elevation. At the same time, body mass index (BMI) and serum total cholesterol, low-density lipoprotein (LDL), leptin, and tumor necrosis factor-alpha (TNF-α) levels were reduced. Lactobacillus spp. and B. animalis were particularly increased in subjects who received probiotic supplements. The abundance of Lactobacillus spp. was inversely correlated with the ether lipid metabolism pathway, while that of B. animalis was positively correlated with serum adiponectin levels. Conclusion Our results show that obesity-related gut dysbiosis can be reshaped by the supplementation of a multi-strain probiotic to improve lipid metabolism. The regular administration of a multi-strain probiotic supplement may be helpful for weight control and health management in overweight and obese children.
Collapse
Affiliation(s)
- An-Chyi Chen
- Division of Pediatric Gastroenterology, China Medical University Children's Hospital, Taichung City, Taiwan.,School of Medicine, China Medical University, Taichung City, Taiwan
| | - Tzu-Jung Fang
- College of Medicine, Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung City, Taiwan.,Division of Geriatrics and Gerontology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung City, Taiwan
| | - Hsieh-Hsun Ho
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan City, Taiwan
| | - Jui-Fen Chen
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan City, Taiwan
| | - Yi-Wei Kuo
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan City, Taiwan
| | - Yen-Yu Huang
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan City, Taiwan
| | - Shin-Yu Tsai
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan City, Taiwan
| | - Shu-Fen Wu
- Division of Pediatric Gastroenterology, China Medical University Children's Hospital, Taichung City, Taiwan.,School of Medicine, China Medical University, Taichung City, Taiwan
| | - Hung-Chih Lin
- Division of Neonatology, China Medical University Children's Hospital, Taichung City, Taiwan.,School of Chinese Medicine, China Medical University, Taichung City, Taiwan.,Asia University Hospital, Asia University, Taichung City, Taiwan
| | - Yao-Tsung Yeh
- Aging and Disease Prevention Research Center, Fooyin University, Kaohsiung City, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Fooyin University, Kaohsiung City, Taiwan
| |
Collapse
|
9
|
Monteiro LDB, Prodonoff JS, Favero de Aguiar C, Correa-da-Silva F, Castoldi A, Bakker NVT, Davanzo GG, Castelucci B, Pereira JADS, Curtis J, Büscher J, Reis LMD, Castro G, Ribeiro G, Virgílio-da-Silva JV, Adamoski D, Dias SMG, Consonni SR, Donato J, Pearce EJ, Câmara NOS, Moraes-Vieira PM. Leptin Signaling Suppression in Macrophages Improves Immunometabolic Outcomes in Obesity. Diabetes 2022; 71:1546-1561. [PMID: 35377454 DOI: 10.2337/db21-0842] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 03/13/2022] [Indexed: 11/13/2022]
Abstract
Obesity is a major concern for global health care systems. Systemic low-grade inflammation in obesity is a major risk factor for insulin resistance. Leptin is an adipokine secreted by the adipose tissue that functions by controlling food intake, leading to satiety. Leptin levels are increased in obesity. Here, we show that leptin enhances the effects of LPS in macrophages, intensifying the production of cytokines, glycolytic rates, and morphological and functional changes in the mitochondria through an mTORC2-dependent, mTORC1-independent mechanism. Leptin also boosts the effects of IL-4 in macrophages, leading to increased oxygen consumption, expression of macrophage markers associated with a tissue repair phenotype, and wound healing. In vivo, hyperleptinemia caused by diet-induced obesity increases the inflammatory response by macrophages. Deletion of leptin receptor and subsequently of leptin signaling in myeloid cells (ObR-/-) is sufficient to improve insulin resistance in obese mice and decrease systemic inflammation. Our results indicate that leptin acts as a systemic nutritional checkpoint to regulate macrophage fitness and contributes to obesity-induced inflammation and insulin resistance. Thus, specific interventions aimed at downstream modulators of leptin signaling may represent new therapeutic targets to treat obesity-induced systemic inflammation.
Collapse
Affiliation(s)
- Lauar de Brito Monteiro
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Juliana Silveira Prodonoff
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Cristhiane Favero de Aguiar
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Felipe Correa-da-Silva
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Angela Castoldi
- Laboratory Keizo Asami, Immunopathology Laboratory, Federal University of Pernambuco, Pernambuco, Brazil
| | - Nikki van Teijlingen Bakker
- Department of Immunometabolism, Max Planck Institute of Epigenetics and Immunobiology, Freiburg im Breisgau, Germany
| | - Gustavo Gastão Davanzo
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Bianca Castelucci
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Jéssica Aparecida da Silva Pereira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
- Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo, São Paulo, Brazil
| | - Jonathan Curtis
- Department of Immunometabolism, Max Planck Institute of Epigenetics and Immunobiology, Freiburg im Breisgau, Germany
- Bloomberg Kimmel Institute and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jörg Büscher
- Department of Immunometabolism, Max Planck Institute of Epigenetics and Immunobiology, Freiburg im Breisgau, Germany
| | - Larissa Menezes Dos Reis
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Gisele Castro
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Guilherme Ribeiro
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - João Victor Virgílio-da-Silva
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Douglas Adamoski
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Sandra Martha Gomes Dias
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Silvio Roberto Consonni
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Edward J Pearce
- Department of Immunometabolism, Max Planck Institute of Epigenetics and Immunobiology, Freiburg im Breisgau, Germany
- Bloomberg Kimmel Institute and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Niels Olsen Saraiva Câmara
- Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo, São Paulo, Brazil
| | - Pedro M Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
- Experimental Medicine Research Cluster, University of Campinas, São Paulo, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, São Paulo, Brazil
| |
Collapse
|
10
|
Mechanisms contributing to adverse outcomes of COVID-19 in obesity. Mol Cell Biochem 2022; 477:1155-1193. [PMID: 35084674 PMCID: PMC8793096 DOI: 10.1007/s11010-022-04356-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/07/2022] [Indexed: 01/08/2023]
Abstract
A growing amount of epidemiological data from multiple countries indicate an increased prevalence of obesity, more importantly central obesity, among hospitalized subjects with COVID-19. This suggests that obesity is a major factor contributing to adverse outcome of the disease. As it is a metabolic disorder with dysregulated immune and endocrine function, it is logical that dysfunctional metabolism contributes to the mechanisms behind obesity being a risk factor for adverse outcome in COVID-19. Emerging data suggest that in obese subjects, (a) the molecular mechanisms of viral entry and spread mediated through ACE2 receptor, a multifunctional host cell protein which links to cellular homeostasis mechanisms, are affected. This includes perturbation of the physiological renin-angiotensin system pathway causing pro-inflammatory and pro-thrombotic challenges (b) existent metabolic overload and ER stress-induced UPR pathway make obese subjects vulnerable to severe COVID-19, (c) host cell response is altered involving reprogramming of metabolism and epigenetic mechanisms involving microRNAs in line with changes in obesity, and (d) adiposopathy with altered endocrine, adipokine, and cytokine profile contributes to altered immune cell metabolism, systemic inflammation, and vascular endothelial dysfunction, exacerbating COVID-19 pathology. In this review, we have examined the available literature on the underlying mechanisms contributing to obesity being a risk for adverse outcome in COVID-19.
Collapse
|
11
|
Ren Y, Zhao H, Yin C, Lan X, Wu L, Du X, Griffiths HR, Gao D. Adipokines, Hepatokines and Myokines: Focus on Their Role and Molecular Mechanisms in Adipose Tissue Inflammation. Front Endocrinol (Lausanne) 2022; 13:873699. [PMID: 35909571 PMCID: PMC9329830 DOI: 10.3389/fendo.2022.873699] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/17/2022] [Indexed: 11/18/2022] Open
Abstract
Chronic low-grade inflammation in adipose tissue (AT) is a hallmark of obesity and contributes to various metabolic disorders, such as type 2 diabetes and cardiovascular diseases. Inflammation in ATs is characterized by macrophage infiltration and the activation of inflammatory pathways mediated by NF-κB, JNK, and NLRP3 inflammasomes. Adipokines, hepatokines and myokines - proteins secreted from AT, the liver and skeletal muscle play regulatory roles in AT inflammation via endocrine, paracrine, and autocrine pathways. For example, obesity is associated with elevated levels of pro-inflammatory adipokines (e.g., leptin, resistin, chemerin, progranulin, RBP4, WISP1, FABP4, PAI-1, Follistatin-like1, MCP-1, SPARC, SPARCL1, and SAA) and reduced levels of anti-inflammatory adipokines such as adiponectin, omentin, ZAG, SFRP5, CTRP3, vaspin, and IL-10. Moreover, some hepatokines (Fetuin A, DPP4, FGF21, GDF15, and MANF) and myokines (irisin, IL-6, and DEL-1) also play pro- or anti-inflammatory roles in AT inflammation. This review aims to provide an updated understanding of these organokines and their role in AT inflammation and related metabolic abnormalities. It serves to highlight the molecular mechanisms underlying the effects of these organokines and their clinical significance. Insights into the roles and mechanisms of these organokines could provide novel and potential therapeutic targets for obesity-induced inflammation.
Collapse
Affiliation(s)
- Yakun Ren
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
| | - Hao Zhao
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xi Lan
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Litao Wu
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Xiaojuan Du
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Helen R. Griffiths
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| | - Dan Gao
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Center, Xi’an, China
- *Correspondence: Dan Gao,
| |
Collapse
|
12
|
Umar MI, Hassan W, Murtaza G, Buabeid M, Arafa E, Irfan HM, Asmawi MZ, Huang X. The Adipokine Component in the Molecular Regulation of Cancer Cell Survival, Proliferation and Metastasis. Pathol Oncol Res 2021; 27:1609828. [PMID: 34588926 PMCID: PMC8473628 DOI: 10.3389/pore.2021.1609828] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/09/2021] [Indexed: 12/22/2022]
Abstract
A hormonal imbalance may disrupt the rigorously monitored cellular microenvironment by hampering the natural homeostatic mechanisms. The most common example of such hormonal glitch could be seen in obesity where the uprise in adipokine levels is in virtue of the expanding bulk of adipose tissue. Such aberrant endocrine signaling disrupts the regulation of cellular fate, rendering the cells to live in a tumor supportive microenvironment. Previously, it was believed that the adipokines support cancer proliferation and metastasis with no direct involvement in neoplastic transformations and tumorigenesis. However, the recent studies have reported discrete mechanisms that establish the direct involvement of adipokine signaling in tumorigenesis. Moreover, the individual adipokine profile of the patients has never been considered in the prognosis and staging of the disease. Hence, the present manuscript has focused on the reported extensive mechanisms that culminate the basis of poor prognosis and diminished survival rate in obese cancer patients.
Collapse
Affiliation(s)
| | - Waseem Hassan
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Manal Buabeid
- Department of Clinical Sciences, Ajman University, Ajman, United Arab Emirates.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | - Elshaimaa Arafa
- Department of Clinical Sciences, Ajman University, Ajman, United Arab Emirates.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | | | - Mohd Zaini Asmawi
- School of Pharmaceutical Sciences, University of Science Malaysia, Pulau Pinang, Malaysia
| | - Xianju Huang
- College of Pharmacy, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
13
|
Mintoff D, Benhadou F, Pace NP, Frew JW. Metabolic syndrome and hidradenitis suppurativa: epidemiological, molecular, and therapeutic aspects. Int J Dermatol 2021; 61:1175-1186. [PMID: 34530487 DOI: 10.1111/ijd.15910] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/28/2021] [Indexed: 12/12/2022]
Abstract
Hidradenitis suppurativa (HS) is a chronic, suppurative condition of the pilosebaceous unit. Patients suffering from HS demonstrate a molecular profile in keeping with a state of systemic inflammation and are often found to fit the criteria for a diagnosis of metabolic syndrome (MetS). In this paper, we review the literature with regards to established data on the prevalence of MetS in HS patients and revise the odds ratio of comorbid disease. Furthermore, we attempt to draw parallels between inflammatory pathways in HS and MetS and evaluate how convergences may explain the risk of comorbid disease, necessitating the need for multidisciplinary care.
Collapse
Affiliation(s)
- Dillon Mintoff
- Department of Dermatology, Mater Dei Hospital, Msida, Malta.,European Hidradenitis Suppurativa Foundation e.V, Dessau, Germany.,Department of Pathology, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Farida Benhadou
- European Hidradenitis Suppurativa Foundation e.V, Dessau, Germany.,Department of Dermatology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Nikolai P Pace
- Department of Pathology, Faculty of Medicine and Surgery, University of Malta, Msida, Malta.,Centre for Molecular Medicine and Biobanking, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - John W Frew
- Department of Dermatology, Liverpool Hospital, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
14
|
Pereira S, Cline DL, Chan M, Chai K, Yoon JS, O'Dwyer SM, Ellis CE, Glavas MM, Webber TD, Baker RK, Erener S, Covey SD, Kieffer TJ. Role of myeloid cell leptin signaling in the regulation of glucose metabolism. Sci Rep 2021; 11:18394. [PMID: 34526546 PMCID: PMC8443652 DOI: 10.1038/s41598-021-97549-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/23/2021] [Indexed: 11/17/2022] Open
Abstract
Although innate immunity is linked to metabolic health, the effect of leptin signaling in cells from the innate immune system on glucose homeostasis has not been thoroughly investigated. We generated two mouse models using Cre-lox methodology to determine the effect of myeloid cell-specific leptin receptor (Lepr) reconstitution and Lepr knockdown on in vivo glucose metabolism. Male mice with myeloid cell-specific Lepr reconstitution (Lyz2Cre+LeprloxTB/loxTB) had better glycemic control as they aged compared to male mice with whole-body transcriptional blockade of Lepr (Lyz2Cre−LeprloxTB/loxTB). In contrast, Lyz2Cre+LeprloxTB/loxTB females only had a trend for diminished hyperglycemia after a prolonged fast. During glucose tolerance tests, Lyz2Cre+LeprloxTB/loxTB males had a mildly improved plasma glucose profile compared to Cre− controls while Lyz2Cre+LeprloxTB/loxTB females had a similar glucose excursion to their Cre− controls. Myeloid cell-specific Lepr knockdown (Lyz2Cre+Leprflox/flox) did not significantly alter body weight, blood glucose, insulin sensitivity, or glucose tolerance in males or females. Expression of the cytokine interleukin 10 (anti-inflammatory) tended to be higher in adipose tissue of male Lyz2Cre+LeprloxTB/loxTB mice (p = 0.0774) while interleukin 6 (pro-inflammatory) was lower in male Lyz2Cre+Leprflox/flox mice (p < 0.05) vs. their respective controls. In conclusion, reconstitution of Lepr in cells of myeloid lineage has beneficial effects on glucose metabolism in male mice.
Collapse
Affiliation(s)
- Sandra Pereira
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Daemon L Cline
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Melissa Chan
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Kalin Chai
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Ji Soo Yoon
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Shannon M O'Dwyer
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Cara E Ellis
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Maria M Glavas
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Travis D Webber
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Robert K Baker
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Suheda Erener
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Scott D Covey
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Timothy J Kieffer
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada. .,Department of Surgery, University of British Columbia, 2775 Laurel Street, Vancouver, BC, V5Z 1M9, Canada. .,School of Biomedical Engineering, University of British Columbia, 251-2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
15
|
Dinari Ghozhdi H, Heidarianpour A, Keshvari M, Tavassoli H. Exercise training and de-training effects on serum leptin and TNF-α in high fat induced diabetic rats. Diabetol Metab Syndr 2021; 13:57. [PMID: 34039404 PMCID: PMC8157711 DOI: 10.1186/s13098-021-00676-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/18/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Adipocytokines, which are secreted by the adipose tissue, contribute to the pathogenesis of obesity-related complications. To evaluate this assumption, we investigated the effects of aerobic exercise training (AET), resistance exercise training (RET), and 4 weeks of de-training on serum leptin and TNF-α levels in diabetic rats. METHOD 36 Wistar rats were divided into normal diet (ND) (control, RET, AET) and high-fat diet (HFD) + STZ (control, RET, AET) groups. Serum insulin, leptin, and TNF-α levels were assessed by commercial ELISA kits. Also fasting blood glucose (FBG), low-density lipoprotein cholesterol (LDL-C), and triglycerides (TG) levels were measured by the colorimetric kits. RESULTS Diabetes induction increased body weight (BW) and FBG, and decreased insulin compared to the ND rats' groups (p < 0.001). 12-weeks of AET and RET programs in the trained diabetic rats led to a decrease in TG, LDL-C, leptin, TNF-α, and FBG, and an increase in insulin compared to the HFD + STZ-C group (p < 0.001). Besides, there was no difference between AET and RET in improving the variables studied (p > 0.05). Also, de-training led to increased BW, TG, leptin, and TNF-α compared to the end of the exercise training (p < 0.05). The correlation between the variables studied was established at different stages of the study (p < 0.05), and only BW was not correlated with insulin during exercise training and de-training (p > 0.05). CONCLUSION These findings indicate that both AET and RET are useful in reducing levels of serum adipocytokines (TNF-α, leptin) in diabetic and non-diabetic rats. At the same time, 4 weeks of de-training was sufficient to lose the metabolic adaptations.
Collapse
Affiliation(s)
- Hamideh Dinari Ghozhdi
- Department of Exercise Physiology, Faculty of Sport Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Ali Heidarianpour
- Department of Exercise Physiology, Faculty of Sport Sciences, Bu-Ali Sina University, Hamedan, Iran.
| | - Maryam Keshvari
- Department of Physical Education and Sport Sciences, Faculty of Literature and Human Sciences, Lorestan University, Khorramabad, Iran
| | - Hassan Tavassoli
- Department of Exercise Physiology, Faculty of Sport Sciences, Bu-Ali Sina University, Hamedan, Iran
| |
Collapse
|
16
|
Kulkarni A, Bowers LW. The role of immune dysfunction in obesity-associated cancer risk, progression, and metastasis. Cell Mol Life Sci 2021; 78:3423-3442. [PMID: 33464384 PMCID: PMC11073382 DOI: 10.1007/s00018-020-03752-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/10/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023]
Abstract
Obesity has been linked to an increased risk of and a worse prognosis for several types of cancer. A number of interrelated mediators contribute to obesity's pro-tumor effects, including chronic adipose inflammation and other perturbations of immune cell development and function. Here, we review studies examining the impact of obesity-induced immune dysfunction on cancer risk and progression. While the role of adipose tissue inflammation in obesity-associated cancer risk has been well characterized, the effects of obesity on immune cell infiltration and activity within the tumor microenvironment are not well studied. In this review, we aim to highlight the impact of both adipose-mediated inflammatory signaling and intratumoral immunosuppressive signaling in obesity-induced cancer risk, progression, and metastasis.
Collapse
Affiliation(s)
- Aneesha Kulkarni
- Department of Nutrition Science, College of Health and Human Sciences, Purdue University, West Lafayette, IN, 47906, USA
| | - Laura W Bowers
- Department of Nutrition Science, College of Health and Human Sciences, Purdue University, West Lafayette, IN, 47906, USA.
| |
Collapse
|
17
|
Pereira S, Cline DL, Glavas MM, Covey SD, Kieffer TJ. Tissue-Specific Effects of Leptin on Glucose and Lipid Metabolism. Endocr Rev 2021; 42:1-28. [PMID: 33150398 PMCID: PMC7846142 DOI: 10.1210/endrev/bnaa027] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Indexed: 12/18/2022]
Abstract
The discovery of leptin was intrinsically associated with its ability to regulate body weight. However, the effects of leptin are more far-reaching and include profound glucose-lowering and anti-lipogenic effects, independent of leptin's regulation of body weight. Regulation of glucose metabolism by leptin is mediated both centrally and via peripheral tissues and is influenced by the activation status of insulin signaling pathways. Ectopic fat accumulation is diminished by both central and peripheral leptin, an effect that is beneficial in obesity-associated disorders. The magnitude of leptin action depends upon the tissue, sex, and context being examined. Peripheral tissues that are of particular relevance include the endocrine pancreas, liver, skeletal muscle, adipose tissues, immune cells, and the cardiovascular system. As a result of its potent metabolic activity, leptin is used to control hyperglycemia in patients with lipodystrophy and is being explored as an adjunct to insulin in patients with type 1 diabetes. To fully understand the role of leptin in physiology and to maximize its therapeutic potential, the mechanisms of leptin action in these tissues needs to be further explored.
Collapse
Affiliation(s)
- Sandra Pereira
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Daemon L Cline
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Maria M Glavas
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Scott D Covey
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Timothy J Kieffer
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada.,Department of Surgery, University of British Columbia, Vancouver, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| |
Collapse
|
18
|
Iwabuchi T, Takahashi N, Nishimura T, Rahman MS, Harada T, Okumura A, Kuwabara H, Takagai S, Nomura Y, Matsuzaki H, Ozaki N, Tsuchiya KJ. Associations Among Maternal Metabolic Conditions, Cord Serum Leptin Levels, and Autistic Symptoms in Children. Front Psychiatry 2021; 12:816196. [PMID: 35185642 PMCID: PMC8851349 DOI: 10.3389/fpsyt.2021.816196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/31/2021] [Indexed: 01/03/2023] Open
Abstract
INTRODUCTION Accumulating evidence has shown that maternal metabolic conditions, such as pre-pregnancy overweight, diabetes mellitus, and hypertensive disorders of pregnancy (HDP) are potential risk factors of autism spectrum disorder (ASD). However, it remains unclear how these maternal conditions lead to neurodevelopmental outcomes in the offspring, including autistic symptoms. Leptin, an adipokine that has pro-inflammatory effects and affects fetal neurodevelopment, is a candidate mediator of the association between maternal metabolic factors and an increased risk of ASD. However, whether prenatal exposure to leptin mediates the association between maternal metabolic conditions and autistic symptoms in children has not been investigated yet. METHODS This study investigated the associations between mothers' metabolic conditions (pre-pregnancy overweight, diabetes mellitus during or before pregnancy, and HDP), leptin concentrations in umbilical cord serum, and autistic symptoms among 762 children from an ongoing cohort study, using generalized structural equation modeling. We used the Social Responsive Scale, Second Edition (SRS-2) at 8-9 years old to calculate total T-scores. Additionally, we used the T-scores for two subdomains: Social Communication and Interaction (SCI) and Restricted Interests and Repetitive Behavior (RRB). RESULTS Umbilical cord leptin levels were associated with pre-pregnancy overweight [coefficient = 1.297, 95% confidence interval (CI) 1.081-1.556, p = 0.005] and diabetes mellitus (coefficient = 1.574, 95% CI 1.206-2.055, p = 0.001). Furthermore, leptin levels were significantly associated with SRS-2 total T-scores (coefficient = 1.002, 95% CI 1.000-1.004, p = 0.023), SCI scores (coefficient = 1.002, 95% CI 1.000-1.004, p = 0.020), and RRB scores (coefficient = 1.001, 95% CI 1.000-1.003, p = 0.044) in children. Associations between maternal metabolic factors and autistic symptoms were not significant. DISCUSSION The present study uncovered an association between cord leptin levels and autistic symptoms in children, while maternal metabolic conditions did not have an evident direct influence on the outcome. These results imply that prenatal pro-inflammatory environments affected by maternal metabolic conditions may contribute to the development of autistic symptoms in children. The findings warrant further investigation into the role of leptin in the development of autistic symptoms.
Collapse
Affiliation(s)
- Toshiki Iwabuchi
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan.,United Graduate School of Child Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Nagahide Takahashi
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan.,United Graduate School of Child Development, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Child and Adolescent Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoko Nishimura
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan.,United Graduate School of Child Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Md Shafiur Rahman
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan.,United Graduate School of Child Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Taeko Harada
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan.,United Graduate School of Child Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Akemi Okumura
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan.,United Graduate School of Child Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hitoshi Kuwabara
- United Graduate School of Child Development, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Psychiatry, Saitama Medical University, Saitama, Japan
| | - Shu Takagai
- United Graduate School of Child Development, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Child and Adolescent Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yoko Nomura
- Queens College and Graduate Center, City University of New York, New York City, NY, United States
| | - Hideo Matsuzaki
- Research Center for Child Mental Development, University of Fukui, Fukui, Japan.,United Graduate School of Child Development, University of Fukui, Fukui, Japan
| | - Norio Ozaki
- Department of Child and Adolescent Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenji J Tsuchiya
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan.,United Graduate School of Child Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
19
|
Leptin Receptor Deficiency Protects Mice against Chronic Cerebral Hypoperfusion-Induced Neuroinflammation and White Matter Lesions. Mediators Inflamm 2021; 2020:7974537. [PMID: 33380900 PMCID: PMC7762643 DOI: 10.1155/2020/7974537] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 11/30/2020] [Accepted: 12/04/2020] [Indexed: 12/19/2022] Open
Abstract
Leptin participates in the inflammatory responses in multiple cell types and animal models. Chronic cerebral hypoperfusion (CCH) induces inflammation in the central nervous system (CNS), which acts as one of the main reasons for CCH-induced white matter lesions (WMLs). But whether leptin participates in the pathogenesis of CCH-induced WMLs remains unknown. Therefore, we performed bilateral common carotid artery stenosis (BCAS) to induce CCH on the leptin receptor- (LepR-) deficient db/db mice, aiming to evaluate the possible involvement of leptin in CCH-induced cognitive impairment, WMLs, and neuroinflammation, and further explore the effect of leptin on chronic hypoxia-induced inflammation using the BV2 microglial cell line. After four weeks of BCAS, wild-type mice showed significant working memory deficits, WMLs, activation of microglia and astrocytes, decrease in the number of oligodendrocytes, downregulation of myelin basic protein expression, and increase in the expression of TNF-α and IL-1β; however, four weeks of BCAS failed to induce significant changes in the LepR-deficient db/db mice but elevated the production of anti-inflammatory cytokines and activated the M2 microglia. We further confirmed that leptin would aggravate the hypoxia-induced proinflammatory cytokine expression in the BV2 microglia cell line. These results suggested that LepR deficiency would protect mice against the CCH-induced cognitive impairment and WMLs by inhibiting glial activation and suppressing proinflammatory responses as well as promoting anti-inflammatory cytokine expression and M2 microglia activation in the white matter.
Collapse
|
20
|
Raut PK, Park PH. Globular adiponectin antagonizes leptin-induced growth of cancer cells by modulating inflammasomes activation: Critical role of HO-1 signaling. Biochem Pharmacol 2020; 180:114186. [DOI: 10.1016/j.bcp.2020.114186] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/16/2022]
|
21
|
Wójcik M, Herman AP, Zieba DA, Krawczyńska A. The Impact of Photoperiod on the Leptin Sensitivity and Course of Inflammation in the Anterior Pituitary. Int J Mol Sci 2020; 21:ijms21114153. [PMID: 32532062 PMCID: PMC7312887 DOI: 10.3390/ijms21114153] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 12/16/2022] Open
Abstract
Leptin has a modulatory impact on the course of inflammation, affecting the expression of proinflammatory cytokines and their receptors. Pathophysiological leptin resistance identified in humans occurs typically in sheep during the long-day photoperiod. This study aimed to determine the effect of the photoperiod with relation to the leptin-modulating action on the expression of the proinflammatory cytokines and their receptors in the anterior pituitary under physiological or acute inflammation. Two in vivo experiments were conducted on 24 blackface sheep per experiment in different photoperiods. The real-time PCR analysis for the expression of the genes IL1B, IL1R1, IL1R2, IL6, IL6R, IL6ST, TNF, TNFR1, and TNFR2 was performed. Expression of all examined genes, except IL1β and IL1R2, was higher during short days. The leptin injection increased the expression of all examined genes during short days. In short days the synergistic effect of lipopolysaccharide and leptin increased the expression of IL1B, IL1R1, IL1R2, IL6, TNF, and TNFR2, and decreased expression of IL6ST. This mechanism was inhibited during long days for the expression of IL1R1, IL6, IL6ST, and TNFR1. The obtained results suggest the occurrence of leptin resistance during long days and suggest that leptin modulates the course of inflammation in a photoperiod-dependent manner in the anterior pituitary.
Collapse
Affiliation(s)
- Maciej Wójcik
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, ul. Instytucka 3, 05-110 Jabłonna, Poland; (A.P.H.); (A.K.)
- Correspondence:
| | - Andrzej Przemysław Herman
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, ul. Instytucka 3, 05-110 Jabłonna, Poland; (A.P.H.); (A.K.)
| | - Dorota Anna Zieba
- Laboratory of Biotechnology and Genomics, Department of Nutrition, Animal Biotechnology and Fisheries, Agricultural University of Krakow, 30-248 Krakow, Poland;
| | - Agata Krawczyńska
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, ul. Instytucka 3, 05-110 Jabłonna, Poland; (A.P.H.); (A.K.)
| |
Collapse
|
22
|
The Influence of Photoperiod on the Action of Exogenous Leptin on Gene Expression of Proinflammatory Cytokines and Their Receptors in the Thoracic Perivascular Adipose Tissue (PVAT) in Ewes. Mediators Inflamm 2019; 2019:7129476. [PMID: 31780867 PMCID: PMC6875191 DOI: 10.1155/2019/7129476] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/02/2019] [Accepted: 10/09/2019] [Indexed: 12/31/2022] Open
Abstract
Leptin resistance is either a condition induced by human obesity or a natural phenomenon associated with seasonality in ruminants. In the cardiovascular system, the leptin resistance state presence is a complex issue. Moreover, the perivascular adipose tissue (PVAT) appears to be crucial as a source of proinflammatory cytokines and as a site of interaction for leptin contributing to endothelium dysfunction and atherosclerosis progression. So the aim of this study was to examine the influence of the photoperiod on the action of exogenous leptin on gene expression of selected proinflammatory cytokines and their receptors in thoracic PVAT of ewe with or without prior lipopolysaccharide (LPS) stimulation. The experiment was conducted on 48 adult, female ewes divided into 4 group (n = 6 in each): control, with LPS intravenous (iv.) injection (400 ng/kg of BW), with leptin iv. injection (20 μg/kg BW), and with LPS and 30-minute-later leptin injection, during short-day (SD) and long-day (LD) seasons. Three hours after LPS/control treatment, animals were euthanized to collect the PVAT adherent to the aorta wall. The leptin injection enhanced IL1B gene expression only in the LD season; however, in both seasons leptin injection intensified LPS-induced increase in IL1B gene expression. IL1R2 gene expression was increased by leptin injection only in the SD season. Neither IL6 nor its receptor and signal transducer gene expressions were influenced by leptin administration. Leptin injection increased TNFA gene expression regardless of photoperiodic conditions. Only in the SD season did leptin treatment increase the gene expression of both TNFα receptors. To conclude, leptin may modulate the inflammatory reaction progress in PVAT. In ewe, the sensitivity of PVAT on leptin action is dependent upon the photoperiodic condition with stronger effects stated in the SD season.
Collapse
|
23
|
Monteiro L, Pereira JADS, Palhinha L, Moraes-Vieira PMM. Leptin in the regulation of the immunometabolism of adipose tissue-macrophages. J Leukoc Biol 2019; 106:703-716. [PMID: 31087711 DOI: 10.1002/jlb.mr1218-478r] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/19/2019] [Accepted: 04/26/2019] [Indexed: 01/03/2025] Open
Abstract
Obesity is a pandemic disease affecting around 15% of the global population. Obesity is a major risk factor for other conditions, such as type 2 diabetes and cardiovascular diseases. The adipose tissue is the main secretor of leptin, an adipokine responsible for the regulation of food intake and energy expenditure. Obese individuals become hyperleptinemic due to increased adipogenesis. Leptin acts through the leptin receptor and induces several immunometabolic changes in different cell types, including adipocytes and Mϕs. Adipose tissue resident Mϕs (ATMs) are the largest leukocyte population in the adipose tissue and these ATMs are in constant contact with the excessive leptin levels secreted in obese conditions. Leptin activates both the JAK2-STAT3 and the PI3K-AKT-mTOR pathways. The activation of these pathways leads to intracellular metabolic changes, with increased glucose uptake, upregulation of glycolytic enzymes, and disruption of mitochondrial function, as well as immunologic alterations, such as increased phagocytic activity and proinflammatory cytokines secretion. Here, we discuss the immunometabolic effects of leptin in Mϕs and how hyperleptinemia can contribute to the low-grade systemic inflammation in obesity.
Collapse
Affiliation(s)
- Lauar Monteiro
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Sao Paulo, Brazil
| | - Jéssica Aparecida da Silva Pereira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Sao Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Lohanna Palhinha
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Manoel M Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Sao Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
24
|
Krawczyńska A, Antushevich H, Bochenek J, Wojtulewicz K, Pawlina B, Herman A, Zięba D. Photoperiodic conditions as a factor modulating leptin influence
on pro-inflammatory cytokines
and their receptors gene expression in ewe’s aorta. JOURNAL OF ANIMAL AND FEED SCIENCES 2019. [DOI: 10.22358/jafs/110022/2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
25
|
Ren X, Keeney JTR, Miriyala S, Noel T, Powell DK, Chaiswing L, Bondada S, St Clair DK, Butterfield DA. The triangle of death of neurons: Oxidative damage, mitochondrial dysfunction, and loss of choline-containing biomolecules in brains of mice treated with doxorubicin. Advanced insights into mechanisms of chemotherapy induced cognitive impairment ("chemobrain") involving TNF-α. Free Radic Biol Med 2019; 134:1-8. [PMID: 30593843 PMCID: PMC6588453 DOI: 10.1016/j.freeradbiomed.2018.12.029] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 12/23/2018] [Indexed: 10/27/2022]
Abstract
Cancer treatments are developing fast and the number of cancer survivors could arise to 20 million in United State by 2025. However, a large fraction of cancer survivors demonstrate cognitive dysfunction and associated decreased quality of life both shortly, and often long-term, after chemotherapy treatment. The etiologies of chemotherapy induced cognitive impairment (CICI) are complicated, made more so by the fact that many anti-cancer drugs cannot cross the blood-brain barrier (BBB). Multiple related factors and confounders lead to difficulties in determining the underlying mechanisms. Chemotherapy induced, oxidative stress-mediated tumor necrosis factor-alpha (TNF-α) elevation was considered as one of the main candidate mechanisms underlying CICI. Doxorubicin (Dox) is a prototypical reactive oxygen species (ROS)-generating chemotherapeutic agent used to treat solid tumors and lymphomas as part of multi-drug chemotherapeutic regimens. We previously reported that peripheral Dox-administration leads to plasma protein damage and elevation of TNF-α in plasma and brain of mice. In the present study, we used TNF-α null (TNFKO) mice to investigate the role of TNF-α in Dox-induced, oxidative stress-mediated alterations in brain. We report that Dox-induced oxidative stress in brain is ameliorated and brain mitochondrial function assessed by the Seahorse-determined oxygen consumption rate (OCR) is preserved in brains of TNFKO mice. Further, we show that Dox-decreased the level of hippocampal choline-containing compounds and brain phospholipases activity are partially protected in TNFKO group in MRS study. Our results provide strong evidence that Dox-targeted mitochondrial damage and levels of brain choline-containing metabolites, as well as phospholipases changes decreased in the CNS are associated with oxidative stress mediated by TNF-α. These results are consistent with the notion that oxidative stress and elevated TNF-α in brain underlie the damage to mitochondria and other pathological changes that lead to CICI. The results are discussed with reference to our identifying a potential therapeutic target to protect against cognitive problems after chemotherapy.
Collapse
Affiliation(s)
- Xiaojia Ren
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Jeriel T R Keeney
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Sumitra Miriyala
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Teresa Noel
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - David K Powell
- Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky Medical Center, Lexington, KY 40536, USA
| | - Luksana Chaiswing
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Subbarao Bondada
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536 USA, USA; Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Daret K St Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536, USA; Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536 USA, USA
| | - D Allan Butterfield
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536 USA, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
26
|
Żelechowska P, Kozłowska E, Pastwińska J, Agier J, Brzezińska-Błaszczyk E. Adipocytokine Involvement in Innate Immune Mechanisms. J Interferon Cytokine Res 2018; 38:527-538. [PMID: 30431386 DOI: 10.1089/jir.2018.0102] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The innate immune response is defined as an immensely complex and sophisticated process aimed at defending the organism against any disturbance in the body homeostasis, including invading pathogens. It requires a close cooperation of a vast amount of different cell types, recognized as inflammatory migrating cells, as well as stationary cells that form tissues. Moreover, innate immune mechanisms require an efficient functioning of various humoral components that exert a significant impact on physiological and pathological processes. Apart from commonly mentioned humoral factors, this group also includes a family of proteins known as adipocytokines that may act as pro- or anti-inflammatory agents or act both ways. Leptin, predominantly characterized as a proinflammatory adipokine, plays a crucial role in endothelium remodeling and regulation, as well as in cell survival and production of numerous cytokines. Adiponectin, similar to leptin, acts on the endothelial cells and the phagocytic properties of immune cells; however, it exerts an anti-inflammatory impact. Resistin has a documented role in the control of angiogenesis and stimulation of proinflammatory mediator generation and release. Furthermore, there are adipokines, ie, visfatin and chemerin, whose participation in the inflammatory processes is ambiguous. This review focuses on the current knowledge on the extensive role of selected adipokines in innate immune response.
Collapse
Affiliation(s)
- Paulina Żelechowska
- Department of Experimental Immunology, Medical University of Lodz, Lodz, Poland
| | - Elżbieta Kozłowska
- Department of Experimental Immunology, Medical University of Lodz, Lodz, Poland
| | - Joanna Pastwińska
- Department of Experimental Immunology, Medical University of Lodz, Lodz, Poland
- Laboratory of Cellular Immunology, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Justyna Agier
- Department of Experimental Immunology, Medical University of Lodz, Lodz, Poland
| | | |
Collapse
|
27
|
Targeted disruption of the iNOS gene improves adipose tissue inflammation and fibrosis in leptin-deficient ob/ob mice: role of tenascin C. Int J Obes (Lond) 2018; 42:1458-1470. [PMID: 29449623 DOI: 10.1038/s41366-018-0005-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 12/12/2017] [Accepted: 12/21/2017] [Indexed: 01/14/2023]
Abstract
BACKGROUND/OBJECTIVES Obesity is related to a dynamic extracellular matrix (ECM) remodeling, which involves the synthesis and degradation of different proteins, such as tenascin C (TNC) in the adipose tissue (AT). Given the functional relationship between leptin and inducible nitric oxide synthase (iNOS), our aim was to analyze the impact of the absence of the iNOS gene in AT inflammation and ECM remodeling in ob/ob mice. SUBJECTS/METHODS The expression of genes involved in inflammation and ECM remodeling was evaluated in 10-week-old male double knockout (DBKO) mice simultaneously lacking the ob and iNOS genes as well as in ob/ob mice classified into three groups [control, leptin-treated (1 mg kg-1 day-1) and pair-fed]. RESULTS Leptin deficiency increased inflammation and fibrosis in AT. As expected, leptin treatment improved the obesity phenotype. iNOS deficiency in ob/ob mice improved insulin sensitivity, AT inflammation, and ECM remodeling, as evidenced by lower AT macrophage infiltration and collagen deposition, a downregulation of proinflammatory and profibrogenic genes Tnf, Emr1, Hif1a, Col6a1, Col6a3, and Tnc, as well as lower circulating TNC levels. Interestingly, leptin upregulated TNC expression and release in 3T3-L1 adipocytes, and iNOS knockdown in 3T3-L1 fat cells produced a significant decrease in basal and leptin-induced Tnc expression. CONCLUSIONS Ablation of iNOS in leptin-deficient mice improved AT inflammation and ECM remodeling-related genes, attenuating fibrosis, and metabolic dysfunction. The activation of iNOS by leptin is necessary for the synthesis and secretion of TNC in adipocytes, suggesting an important role of this alarmin in the development of AT inflammation and fibrosis.
Collapse
|
28
|
Souza-Almeida G, D'Avila H, Almeida PE, Luna-Gomes T, Liechocki S, Walzog B, Hepper I, Castro-Faria-Neto HC, Bozza PT, Bandeira-Melo C, Maya-Monteiro CM. Leptin Mediates In Vivo Neutrophil Migration: Involvement of Tumor Necrosis Factor-Alpha and CXCL1. Front Immunol 2018; 9:111. [PMID: 29467755 PMCID: PMC5808117 DOI: 10.3389/fimmu.2018.00111] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/15/2018] [Indexed: 11/13/2022] Open
Abstract
Leptin directly activates macrophages and lymphocytes, but the role of leptin in neutrophil activation and migration is still controversial. Here, we investigate the in vivo mechanisms of neutrophil migration induced by leptin. The intraperitoneal injection of leptin (1 mg/kg) induces a time- and concentration-dependent neutrophil influx. We did not observe the enhancement of lipid bodies/droplets in neutrophils, after leptin treatment, as we had observed previously in peritoneal macrophages. The participation of leukotriene B4 (LTB4) in neutrophil recruitment triggered by leptin was investigated using different strategies. Leptin-induced neutrophil recruitment occurs both in the absence of 5-lipoxygenase activity in 5-lipoxygenase (5-LO)-/- mice and after the administration of either 5-LO inhibitor (Zileuton) or the LTB4 receptor antagonist (U-75302). Moreover, no direct induction of LTB4 by leptin could be observed. Neutrophil influx could not be prevented by the mammalian target of rapamycin (mTOR) inhibitor, rapamycin, contrasting with the leptin-induced signaling for lipid body formation in macrophage that is mTOR-dependent. Leptin administration led to tumor necrosis factor-alpha (TNFα) production by the peritoneal cells both in vivo and in vitro. In addition, neutrophil recruitment was inhibited in tumor necrosis factor receptor 1 (TNFR1-/-) mice, indicating a role for TNF in leptin-induced neutrophil recruitment to the peritoneal cavity. Leptin-induced neutrophil influx was PI3Kγ-dependent, as it was absent in PI3Kγ-/- mice. Accordingly, leptin induced the peritoneal cells to produce CXCL1, both in vivo and in vitro, and the neutrophil influx was ablated after using an antibody against CXCL1. Our results establish TNFα/TNFR1- and CXCL1-dependent signaling as important pathways for leptin-induced neutrophil migration in vivo.
Collapse
Affiliation(s)
- Glaucia Souza-Almeida
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Heloisa D'Avila
- Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Patricia E Almeida
- Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Tatiana Luna-Gomes
- Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil.,Departamento de Ciências da Natureza, Instituto de Aplicação Fernando Rodrigues da Silveira, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sally Liechocki
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Barbara Walzog
- Walter Brendel Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ingrid Hepper
- Walter Brendel Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Patricia T Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Christianne Bandeira-Melo
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarissa M Maya-Monteiro
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
29
|
Rastogi D, Nico J, Johnston AD, Tobias TAM, Jorge Y, Macian F, Greally JM. CDC42-related genes are upregulated in helper T cells from obese asthmatic children. J Allergy Clin Immunol 2018; 141:539-548.e7. [PMID: 28479334 PMCID: PMC5671374 DOI: 10.1016/j.jaci.2017.04.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 04/04/2017] [Accepted: 04/18/2017] [Indexed: 01/21/2023]
Abstract
BACKGROUND Pediatric obesity-related asthma is more severe and less responsive to medications than asthma in normal-weight children. Obese asthmatic children have nonatopic TH1-polarized systemic inflammation that correlates with pulmonary function deficits, but the pathways underlying TH1-polarized inflammation are not well understood. OBJECTIVE We compared the CD4+ T-cell transcriptome in obese children with asthma with that in normal-weight children with asthma to identify key differentially expressed genes associated with TH1-polarized inflammation. METHODS CD4+ T-cell transcriptome-wide differential gene expression was compared between 21 obese and 21 normal-weight children by using directional RNA sequencing. High-confidence differentially expressed genes were verified in the first cohort and validated in a second cohort of 20 children (10 obese and 10 normal-weight children) by using quantitative RT-PCR. RESULTS Transcriptome-wide differential gene expression among obese asthmatic children was enriched for genes, including VAV2, DOCK5, PAK3, PLD1, CDC42EP4, and CDC42PBB, which are associated with CDC42, a small guanosine triphosphate protein linked to T-cell activation. Upregulation of MLK3 and PLD1, genes downstream of CDC42 in the mitogen-activated protein kinase and mammalian target of rapamycin pathways and the inverse correlation of CDC42EP4 and DOCK5 transcript counts with FEV1/FVC ratio together support a role of CDC42 in the TH1 polarization and pulmonary function deficits found in patients with obesity-related asthma. CONCLUSIONS Our study identifies the CDC42 pathway as a novel target that is upregulated in TH cells of obese asthmatic children, suggesting its role in nonatopic TH1-polarized systemic inflammation and pulmonary function deficits found in patients with pediatric obesity-related asthma.
Collapse
Affiliation(s)
- Deepa Rastogi
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY.
| | - John Nico
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY
| | - Andrew D Johnston
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY
| | | | - Yurydia Jorge
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY
| | - Fernando Macian
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY
| | - John M Greally
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
30
|
To M, Kono Y, Ogura N, Mikami S, Honda N, Hitani A, Kano I, Haruki K, To Y. Obesity-related systemic oxidative stress: An important factor of poor asthma control. Allergol Int 2018. [PMID: 28648979 DOI: 10.1016/j.alit.2017.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
31
|
Kim JH, Choi HJ, Oh CH, Oh JW, Han JS. PLD1 activation mediates Amb a 1-induced Th2-associated cytokine expression via the JNK/ATF-2 pathway in BEAS-2B cells. Cell Immunol 2015; 298:9-17. [PMID: 26302934 DOI: 10.1016/j.cellimm.2015.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 07/21/2015] [Accepted: 08/13/2015] [Indexed: 01/06/2023]
Abstract
The purpose of this study was to identify the role of phospholipase D1 (PLD1) in Amb a 1-induced IL-5 and IL-13 expression. When BEAS-2B cells were stimulated with Amb a 1, PLD activity increased, and knockdown of PLD1 decreased Amb a 1-induced IL-5 and IL-13 expression. Amb a 1 also activated the PLCγ/p70S6K/JNK pathway. Furthermore, Amb a 1-induced PLD activation was also attenuated by PLCγ inhibition, and knockdown of PLD1 decreased Amb a 1-induced activation of P70S6K and JNK. When ATF-2 activity was blocked with ATF-2 siRNA, Amb a 1-induced IL-5 and IL-13 expression was completely abolished, indicating that ATF-2 is a transcriptional factor required for the expression of IL-5 and IL-13 in response to Amb a 1. Taken together, we suggest that PLD1 acts as an important regulator in Amb a 1-induced expression of IL-5 and IL-13 via a PLCγ/p70S6K/JNK/ATF-2 pathway in BEAS-2B cells.
Collapse
Affiliation(s)
- Joo-Hwa Kim
- Department of Pediatrics, College of Medicine, Hanyang University, Seoul 133-791, Republic of Korea
| | - Hye-Jin Choi
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Republic of Korea
| | - Cheong-Hae Oh
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Republic of Korea
| | - Jae-Won Oh
- Department of Pediatrics, College of Medicine, Hanyang University, Seoul 133-791, Republic of Korea.
| | - Joong-Soo Han
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Republic of Korea.
| |
Collapse
|
32
|
Wan Z, Mah D, Simtchouk S, Kluftinger A, Little JP. Role of amyloid β in the induction of lipolysis and secretion of adipokines from human adipose tissue. Adipocyte 2015; 4:212-6. [PMID: 26257989 DOI: 10.4161/21623945.2014.985020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 10/19/2014] [Accepted: 11/03/2014] [Indexed: 11/19/2022] Open
Abstract
Amyloid-β (Aβ) peptide, which is generated from proteolytic cleavage of amyloid precursor protein (APP), is a key molecule involved in the pathology of Alzheimer disease. Both APP and Aβ peptides are expressed in adipose tissue, however it is currently unclear whether Aβ can affect the key functions of adipose tissue. We aimed to explore whether Aβ affected lipolysis and adipokine secretion in cultured human adipose tissue. We found that Aβ25-35, which contains the main functional domain of the Aβ, stimulated lipolysis via PKA and ERK1/2-dependent pathways and that Aβ25-35 induced leptin and IL-6 secretion. It is concluded that Aβ peptide exerts functional effects on adipose tissue that may lead to increased release of free fatty acids and pro-inflammatory adipokines.
Collapse
|
33
|
Polyzos SA, Mantzoros CS. Leptin in health and disease: facts and expectations at its twentieth anniversary. Metabolism 2015; 64:5-12. [PMID: 25467841 DOI: 10.1016/j.metabol.2014.10.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 10/20/2014] [Accepted: 10/20/2014] [Indexed: 02/07/2023]
Affiliation(s)
- Stergios A Polyzos
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Boston VA Healthcare system and Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
34
|
Jia G, Aroor AR, Martinez-Lemus LA, Sowers JR. Overnutrition, mTOR signaling, and cardiovascular diseases. Am J Physiol Regul Integr Comp Physiol 2014; 307:R1198-206. [PMID: 25253086 DOI: 10.1152/ajpregu.00262.2014] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The prevalence of obesity and associated medical disorders has increased dramatically in the United States and throughout much of the world in the past decade. Obesity, induced by excess intake of carbohydrates and fats, is a major cause of Type 2 diabetes, hypertension, and the cardiorenal metabolic syndrome. There is emerging evidence that excessive nutrient intake promotes signaling through the mammalian target of rapamycin (mTOR), which, in turn, may lead to alterations of cellular metabolic signaling leading to insulin resistance and obesity-related diseases, such as diabetes, cardiovascular and kidney disease, as well as cancer. While the pivotal role of mTOR signaling in regulating metabolic stress, autophagy, and adaptive immune responses has received increasing attention, there remain many gaps in our knowledge regarding this important nutrient sensor. For example, the precise cellular signaling mechanisms linking excessive nutrient intake and enhanced mTOR signaling with increased cardiovascular and kidney disease, as well as cancer, are not well understood. In this review, we focus on the effects that the interaction between excess intake of nutrients and enhanced mTOR signaling have on the promotion of obesity-associated diseases and potential therapeutic strategies involving targeting mTOR signaling.
Collapse
Affiliation(s)
- Guanghong Jia
- Divisions of Endocrinology, Diabetes, Hypertension and Metabolism, Diabetes Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri; Harry S. Truman Memorial Veterans Hospital, University of Missouri School of Medicine, Columbia, Missouri; and
| | - Annayya R Aroor
- Divisions of Endocrinology, Diabetes, Hypertension and Metabolism, Diabetes Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri; Harry S. Truman Memorial Veterans Hospital, University of Missouri School of Medicine, Columbia, Missouri; and
| | - Luis A Martinez-Lemus
- Departments of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; Harry S. Truman Memorial Veterans Hospital, University of Missouri School of Medicine, Columbia, Missouri; and Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri
| | - James R Sowers
- Divisions of Endocrinology, Diabetes, Hypertension and Metabolism, Diabetes Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri; Departments of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; Harry S. Truman Memorial Veterans Hospital, University of Missouri School of Medicine, Columbia, Missouri; and Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri
| |
Collapse
|