51
|
Silveira-Rodrigues JG, Pires W, Gomes PF, Ogando PHM, Melo BP, Aleixo IMS, Soares DD. Combined exercise training improves specific domains of cognitive functions and metabolic markers in middle-aged and older adults with type 2 diabetes mellitus. Diabetes Res Clin Pract 2021; 173:108700. [PMID: 33600868 DOI: 10.1016/j.diabres.2021.108700] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 01/12/2021] [Accepted: 02/01/2021] [Indexed: 12/15/2022]
Abstract
AIM To investigate the effects of 8-weeks of CT on specific domains of cognitive function, metabolic and cardiovascular parameters of subjects with Type 2 Diabetes Mellitus (T2DM). METHODS 31 sedentary T2DM adults and older divided into CT (3x/week, during 8-week, n = 16) or Control group (CONT, n = 15). Before and after the intervention, a cognitive task battery, blood samples, and functional tests were assessed. RESULTS CT improved inhibitory control (d = 0.89), working memory (d = 0.88), cognitive flexibility (d = 0.67) and attention/concentration (d = 0.64) in T2DM subjects. However, memory, verbal fluency, and processing speed (d < 0.1, p > 0.05 for all) were not changed. The CT-induced improvements on global cognitive z-score (r = -0.51; p < 0.001) were inversely correlated to cognitive screening scores. Moreover, CT improved functional performance (p < 0.05) and reduced insulin levels (p = 0.04). Although there was no statistical significance, there were a clinically relevant reduction of peripheral insulin sensitivity (d = 0.51, p = 0.09), resistin levels (d = 0.53, p = 0.08), diastolic (d = 0.63, p = 0.09) and mean blood pressure (d = 0.50, p = 0.09). Conversely, no changes were observed for glucose, fructosamine and blood lipids (d < 0.2 for all). CONCLUSION CT partially reversed the negative effects of T2DM on specific cognitive domains possibly by amelioration of metabolic regulation. Moreover, lower cognitive scores may modulate the responsivity of cognitive function to CT.
Collapse
Affiliation(s)
- João Gabriel Silveira-Rodrigues
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627 Belo Horizonte, MG 31270-901, Brazil.
| | - Washington Pires
- Sports Center of Federal University of Ouro Preto, St. Two, MG 35400-000, Brazil
| | - Patrícia Ferreira Gomes
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627 Belo Horizonte, MG 31270-901, Brazil
| | - Pedro Henrique Madureira Ogando
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627 Belo Horizonte, MG 31270-901, Brazil
| | - Bruno Pereira Melo
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627 Belo Horizonte, MG 31270-901, Brazil
| | - Ivana Montandon Soares Aleixo
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627 Belo Horizonte, MG 31270-901, Brazil
| | - Danusa Dias Soares
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627 Belo Horizonte, MG 31270-901, Brazil
| |
Collapse
|
52
|
Land Lail H, Feresin RG, Hicks D, Stone B, Price E, Wanders D. Berries as a Treatment for Obesity-Induced Inflammation: Evidence from Preclinical Models. Nutrients 2021; 13:nu13020334. [PMID: 33498671 PMCID: PMC7912458 DOI: 10.3390/nu13020334] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/15/2021] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammation that accompanies obesity is associated with the infiltration of metabolically active tissues by inflammatory immune cells. This propagates a chronic low-grade inflammation associated with increased signaling of common inflammatory pathways such as NF-κB and Toll-like receptor 4 (TLR4). Obesity-associated inflammation is linked to an increased risk of chronic diseases, including type 2 diabetes, cardiovascular disease, and cancer. Preclinical rodent and cell culture studies provide robust evidence that berries and their bioactive components have beneficial effects not only on inflammation, but also on biomarkers of many of these chronic diseases. Berries contain an abundance of bioactive compounds that have been shown to inhibit inflammation and to reduce reactive oxygen species. Therefore, berries represent an intriguing possibility for the treatment of obesity-induced inflammation and associated comorbidities. This review summarizes the anti-inflammatory properties of blackberries, blueberries, strawberries, and raspberries. This review highlights the anti-inflammatory mechanisms of berries and their bioactive components that have been elucidated through the use of preclinical models. The primary mechanisms mediating the anti-inflammatory effects of berries include a reduction in NF-κB signaling that may be secondary to reduced oxidative stress, a down-regulation of TLR4 signaling, and an increase in Nrf2.
Collapse
|
53
|
Ma Q, Deng P, Lin M, Yang L, Li L, Guo L, Zhang L, He M, Lu Y, Pi H, Zhang Y, Yu Z, Chen C, Zhou Z. Long-term bisphenol A exposure exacerbates diet-induced prediabetes via TLR4-dependent hypothalamic inflammation. JOURNAL OF HAZARDOUS MATERIALS 2021; 402:123926. [PMID: 33254826 DOI: 10.1016/j.jhazmat.2020.123926] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 06/12/2023]
Abstract
Bisphenol A (BPA), an environmental endocrine-disrupting compound, has been revealed associated with metabolic disorders such as obesity, prediabetes, and type 2 diabetes (T2D). However, its underlying mechanisms are still not fully understood. Here, we provide new evidence that BPA is a risk factor for T2D from a case-control study. To explore the detailed mechanisms, we used two types of diet models, standard diet (SD) and high-fat diet (HFD), to study the effects of long-term BPA exposure on prediabetes in 4-week-old mice. We found that BPA exposure for 12 weeks exacerbated HFD-induced prediabetic symptoms. Female mice showed increased body mass, serum insulin level, and impaired glucose tolerance, while male mice only exhibited impaired glucose tolerance. No change was found in SD-fed mice. Besides, BPA exposure enhanced astrocyte-dependent hypothalamic inflammation in both male and female mice, which impaired proopiomelanocortin (POMC) neuron functions. Moreover, eliminating inflammation by toll-like receptor 4 (TLR4) knockout significantly abolished the effects of BPA on the hypothalamus and diet-induced prediabetes. Taken together, our data establish a key role for TLR4-dependent hypothalamic inflammation in regulating the effects of BPA on prediabetes.
Collapse
Affiliation(s)
- Qinlong Ma
- Department of Occupational Health, Army Medical University (Former Name: Third Military Medical University), Chongqing 400038, People's Republic of China
| | - Ping Deng
- Department of Occupational Health, Army Medical University (Former Name: Third Military Medical University), Chongqing 400038, People's Republic of China
| | - Min Lin
- Department of Occupational Health, Army Medical University (Former Name: Third Military Medical University), Chongqing 400038, People's Republic of China
| | - Lingling Yang
- Department of Occupational Health, Army Medical University (Former Name: Third Military Medical University), Chongqing 400038, People's Republic of China
| | - Le Li
- Department of Health Management Center, Southwest Hospital, Army Medical University (Former Name: Third Military Medical University), Chongqing 400038, People's Republic of China
| | - Lu Guo
- Department of Neurology, Daping Hospital, Army Medical University (Former Name: Third Military Medical University), Chongqing 400042, People's Republic of China
| | - Lei Zhang
- Department of Occupational Health, Army Medical University (Former Name: Third Military Medical University), Chongqing 400038, People's Republic of China
| | - Mindi He
- Department of Occupational Health, Army Medical University (Former Name: Third Military Medical University), Chongqing 400038, People's Republic of China
| | - Yonghui Lu
- Department of Occupational Health, Army Medical University (Former Name: Third Military Medical University), Chongqing 400038, People's Republic of China
| | - Huifeng Pi
- Department of Occupational Health, Army Medical University (Former Name: Third Military Medical University), Chongqing 400038, People's Republic of China
| | - Yanwen Zhang
- Department of Occupational Health, Army Medical University (Former Name: Third Military Medical University), Chongqing 400038, People's Republic of China
| | - Zhengping Yu
- Department of Occupational Health, Army Medical University (Former Name: Third Military Medical University), Chongqing 400038, People's Republic of China
| | - Chunhai Chen
- Department of Occupational Health, Army Medical University (Former Name: Third Military Medical University), Chongqing 400038, People's Republic of China.
| | - Zhou Zhou
- Department of Environmental Medicine, and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.
| |
Collapse
|
54
|
Wang X, Chen S, He J, Chen W, Ding Y, Huang J, Huang J. Histone methyltransferases G9a mediated lipid-induced M1 macrophage polarization through negatively regulating CD36. Metabolism 2021; 114:154404. [PMID: 33069810 DOI: 10.1016/j.metabol.2020.154404] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/30/2020] [Accepted: 10/11/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Recent studies have considered the obesity-related lipid environment as the potential cause for M1 macrophage polarization in type 2 diabetes. However, the specific regulatory mechanism is still unclear. Here, we investigated the role and molecular mechanism of histone methyltransferases G9a in lipids-induced M1 macrophage polarization in type 2 diabetes. METHODS We used saturated fatty acid palmitate to induce macrophage polarization, and performed real-time PCR, western blot, flow cytometry and CHIP assay to study the function and molecular mechanism of G9a. Additionally, we isolated the peripheral blood mononuclear cells (PBMCs) from 187 patients with type 2 diabetes and 68 healthy individuals, and analyzed the expression level of G9a. RESULTS The palmitate treatment induced the macrophage M1 polarization, and decreased the expression of G9a. The deficiency of G9a could promote the palmitate-induced M1 macrophage polarization, whereas, over-expressing G9a notably suppressed this process. Meanwhile, we observed the regulatory role of G9a on the ER stress which could contribute to M1 macrophage. Furthermore, we identified the fatty acid transport protein CD36 as the potential target of G9a. Dependent on the methyltransferase activity, G9a could negatively regulate the expression of CD36 induced by palmitate. The CD36 inhibitor SSO could significantly attenuate the regulatory effect of G9a on M1 macrophage polarization and ER stress. Importantly, G9a was decreased, and suppressed CD36 and M1 macrophage genes in the PBMCs from individuals with type 2 diabetes. CONCLUSIONS Our studies demonstrate that G9a plays critical roles in lipid-induced M1 macrophage polarization via negatively regulating CD36.
Collapse
Affiliation(s)
- Xiuling Wang
- Department of Medical Laboratory, The Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, China
| | - Shanshan Chen
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinrong He
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiqun Chen
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Ding
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Huang
- Department of Nephrology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jin Huang
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
55
|
Li X, Sun F, Lu J, Zhang J, Wang J, Zhu H, Gu M, Ma J. Osteoclasts May Affect Glucose Uptake-Related Insulin Resistance by Secreting Resistin. Diabetes Metab Syndr Obes 2021; 14:3461-3470. [PMID: 34366677 PMCID: PMC8336992 DOI: 10.2147/dmso.s316964] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/06/2021] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVES Bone may play a role in the modulation of insulin sensitivity. Insulin resistance can be caused by increased resistin. However, whether osteoclasts affect the insulin resistance via resistin remains unclear. In the present study, we show the expression of resistin in osteoclasts and the possible underlying role of resistin on glucose uptake-related insulin resistance in vitro. METHODS Conditioned mediums (CM) were collected from Raw264.7 cells treated without (CCM) or with RANKL (CM3, treated with RANKL for 3 days; CM5, treated with RANKL for 5 days) and transfected with control or resistin siRNA (CMsiRNA). The osteoclast formation was examined by tartrate resistant acid phosphatase (TRAP) staining. C2C12 myoblasts were cultured with the CM or CMsiRNA. Glucose uptake was evaluated by 2-NBDG fluorescence intensity. Resistin expression was evaluated by quantitative real-time polymerase chain reaction (qRT-PCR) and enzyme-linked immunosorbent assay. Statistical analysis was performed by an independent two sample t-test or one-way ANOVA. RESULTS The 2-NBDG fluorescence intensity was higher in C2C12 cells treated with CCM compared to those that received CM3 and CM5 (p < 0.05). Resistin mRNA and protein expressions were both increased in RAW264.7 cells treated with RANKL for 3 days and 5 days compared with those cells without RANKL administration. The 2-NBDG fluorescence intensities in C2C12 cells treated with CMsiRNA and CM5+Anti-resistin antibody were significantly higher than those cultured with CM5 (p < 0.05). CONCLUSION Osteoclasts may promote glucose uptake-related insulin resistance by secreting resistin.
Collapse
Affiliation(s)
- Xiangqi Li
- Department of Endocrinology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People’s Republic of China
| | - Fei Sun
- Department of Endocrinology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People’s Republic of China
| | - Jiancan Lu
- Department of Endocrinology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People’s Republic of China
| | - Jichen Zhang
- Department of Endocrinology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People’s Republic of China
| | - Jingnan Wang
- Department of Endocrinology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People’s Republic of China
| | - Hongling Zhu
- Department of Endocrinology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People’s Republic of China
| | - Mingjun Gu
- Department of Endocrinology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People’s Republic of China
| | - Junhua Ma
- Department of Endocrinology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People’s Republic of China
- Correspondence: Junhua Ma Department of Endocrinology, Shanghai Gongli Hospital, The Second Military Medical University, Miaopu Road 219, Shanghai, 200135, People’s Republic of ChinaTel +86 21 58858730 Email
| |
Collapse
|
56
|
Gater DR, Farkas GJ, Tiozzo E. Pathophysiology of Neurogenic Obesity After Spinal Cord Injury. Top Spinal Cord Inj Rehabil 2021; 27:1-10. [PMID: 33814879 PMCID: PMC7983633 DOI: 10.46292/sci20-00067] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Individuals with a spinal cord injury (SCI) have a unique physiology characterized by sarcopenia, neurogenic osteoporosis, neurogenic anabolic deficiency, sympathetic dysfunction, and blunted satiety associated with their SCI, all of which alter energy balance and subsequently body composition. The distinct properties of "neurogenic obesity" place this population at great risk for metabolic dysfunction, including systemic inflammation, hyperglycemia, dyslipidemia, and hypertension. The purpose of this article is to demonstrate the relationship between neurogenic obesity and the metabolic syndrome after SCI, highlighting the mechanisms associated with adipose tissue pathology and those respective comorbidities. Additionally, representative studies of persons with SCI will be provided to elucidate the severity of the problem and to prompt greater vigilance among SCI specialists as well as primary care providers in order to better manage the epidemic from a public health perspective.
Collapse
Affiliation(s)
- David R. Gater
- Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, Florida
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| | - Gary J. Farkas
- Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, Florida
| | - Eduard Tiozzo
- Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
57
|
Abstract
Diabetes is on the rise across the globe affecting more than 463 million people and crucially increasing morbidities of diabetes-associated diseases. Urgent and immense actions are needed to improve diabetes prevention and treatment. Regarding the correlation of diabetes with many associated diseases, inhibition of the disease progression is more crucial than controlling symptoms. Currently, anti-diabetic drugs are accompanied by undesirable side-effects and target confined types of biomolecules. Thus, extensive research is demanding to identify novel disease mechanisms and molecular targets as probable candidates for effective treatment of diabetes. This review discusses the conventional molecule targets that have been applied for their therapeutic rationale in treatment of diabetes. Further, the emerging and prospective molecular targets for the future focus of library screenings are presented.
Collapse
Affiliation(s)
- Faezeh Almasi
- Pharmaceutical Biotechnology Lab, Department of Microbial Biotechnology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
| | - Fatemeh Mohammadipanah
- Pharmaceutical Biotechnology Lab, Department of Microbial Biotechnology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
58
|
Gudise V, Chowdhury B. Molecular mechanisms and the vital roles of resistin, TLR 4, and NF-κB in treating type 2 diabetic complications. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2020. [DOI: 10.1186/s43088-020-00078-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Abstract
Background
Type 2 diabetes in obese (≥ 25 and ≥ 30 kg/m2) patients is the foremost cause of cardiovascular complications like stroke, osteoarthritis, cancers (endometrial, breast, ovarian, liver, kidney, colon, and prostate), and vascular complications like diabetic neuropathy, diabetic and retinopathy, and diabetic nephropathy. It is recognized as a global burden disorder with high prevalence in middle-income nations which might lead to a double burden on health care professionals. Hence, this review emphasizes on understanding the complexity and vital signaling tracts involved in diabetic complications for effective treatment.
Main body
Type 2 diabetes in overweight patients induces the creation of specific ROS that further leads to changes in cellular proliferation, hypothalamus, and fringe. The resistin, TLR4, and NF-κB signalings are mainly involved in the progression of central and fringe changes such as insulin resistance and inflammation in diabetic patients. The overexpression of these signals might lead to the rapid progression of diabetic vascular complications induced by the release of proinflammatory cytokines, chemokines, interleukins, and cyclooxygenase-mediated chemicals. Until now, there has been no curative treatment for diabetes. Therefore, to effectively treat complications of type 2 diabetes, the researchers need to concentrate on the molecular mechanisms and important signaling tracts involved.
Conclusion
In this review, we suggested the molecular mechanism of STZ-HFD induced type 2 diabetes and the vital roles of resistin, TLR4, and NF-κB signalings in central, fringe changes, and development diabetic complications for its effective treatment.
Graphical abstract
Collapse
|
59
|
Rawal K, Purohit KM, Patel TP, Karont N, Gupta S. Resistin mitigates stemness and metabolic profile of human adipose-derived mesenchymal stem cells via insulin resistance. Cytokine 2020; 138:155374. [PMID: 33271386 DOI: 10.1016/j.cyto.2020.155374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 10/31/2020] [Accepted: 11/15/2020] [Indexed: 01/23/2023]
Abstract
During obesity adipose tissue abundantly secrete pro-inflammatory adipokines like Tumour Necrosis factor-alpha (TNFα), resistin, leptin, etc. but reduced anti-inflammatory adipokines like adiponectin, interleukin (IL)-10, and IL-4. In our recent clinical study, it was observed that both gene expressions and stored levels of resistin were elevated in adipose tissue of metabolically obese Indians. Resistin profoundly increases obesity, mitigates lipid metabolism, and causes peripheral insulin resistance. It dysregulates the metabolism of human adipocytes but, its effects on human adipose-derived mesenchymal stem cells (hADSC) are sparsely explored. Therefore, the present study was designed to explore the repercussion of resistin on stemness and metabolic profile of hADSC. hADSC were isolated from a healthy individual followed by immunophenotyping. Purified cells were treated with resistin and proliferation was monitored by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) and Cell Cycle experiments. Gene expressions of pluripotent markers, inflammatory mediators, and lipogenic genes were scrutinized. Insulin sensitivity was examined by western blot and glucose uptake assay. Further, consequences of resistin on differentiation potentials of hADSC were examined by temporal expressions of phospho (p)SMAD1/5/8 protein complex, non-phosphorylated beta (β) catenin, and their dependent adipogenic transcription factors (ATF) and osteogenic transcription factors (OTF). MTT and cell cycle analysis revealed that resistin hampered proliferation of hADSC. Expressions of inflammatory markers and lipogenic genes were elevated. Resistin impaired insulin sensitivity and thus embarked insulin resistance in hADSC. Resistin increased adipogenesis and osteogenesis by altering expressions of activated pSMAD1/5/8 complex, activated β catenin, ATF and OTF temporally. Downregulation of CCAAT/enhancer-binding proteins (C/EBP)α and adiponectin in adipocytes and Sirtuin (SIRT)1 in osteocytes denote that resistin induces immaturity and insulin resistance in adipocytes and osteocytes. This is the first study which, reports that resistin mitigates the stemness of hADSC by reducing proliferation, inducing insulin resistance, and hampering maturation of adipocyte and osteocyte which could lead to metabolic disorders.
Collapse
Affiliation(s)
- Komal Rawal
- Molecular Endocrinology and Stem Cell Research Laboratory, Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India
| | - Kishan M Purohit
- Molecular Endocrinology and Stem Cell Research Laboratory, Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India
| | - Tushar P Patel
- Molecular Endocrinology and Stem Cell Research Laboratory, Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India
| | - Neeta Karont
- Molecular Endocrinology and Stem Cell Research Laboratory, Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India
| | - Sarita Gupta
- Molecular Endocrinology and Stem Cell Research Laboratory, Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India.
| |
Collapse
|
60
|
Lizcano F, Arroyave F. Control of Adipose Cell Browning and Its Therapeutic Potential. Metabolites 2020; 10:metabo10110471. [PMID: 33227979 PMCID: PMC7699191 DOI: 10.3390/metabo10110471] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/20/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022] Open
Abstract
Adipose tissue is the largest endocrine organ in humans and has an important influence on many physiological processes throughout life. An increasing number of studies have described the different phenotypic characteristics of fat cells in adults. Perhaps one of the most important properties of fat cells is their ability to adapt to different environmental and nutritional conditions. Hypothalamic neural circuits receive peripheral signals from temperature, physical activity or nutrients and stimulate the metabolism of white fat cells. During this process, changes in lipid inclusion occur, and the number of mitochondria increases, giving these cells functional properties similar to those of brown fat cells. Recently, beige fat cells have been studied for their potential role in the regulation of obesity and insulin resistance. In this context, it is important to understand the embryonic origin of beige adipocytes, the response of adipocyte to environmental changes or modifications within the body and their ability to transdifferentiate to elucidate the roles of these cells for their potential use in therapeutic strategies for obesity and metabolic diseases. In this review, we discuss the origins of the different fat cells and the possible therapeutic properties of beige fat cells.
Collapse
Affiliation(s)
- Fernando Lizcano
- Center of Biomedical Investigation, (CIBUS), Universidad de La Sabana, 250008 Chia, Colombia
- Correspondence:
| | - Felipe Arroyave
- Doctoral Program in Biociencias, Universidad de La Sabana, 250008 Chia, Colombia
| |
Collapse
|
61
|
AlZaim I, Hammoud SH, Al-Koussa H, Ghazi A, Eid AH, El-Yazbi AF. Adipose Tissue Immunomodulation: A Novel Therapeutic Approach in Cardiovascular and Metabolic Diseases. Front Cardiovasc Med 2020; 7:602088. [PMID: 33282920 PMCID: PMC7705180 DOI: 10.3389/fcvm.2020.602088] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue is a critical regulator of systemic metabolism and bodily homeostasis as it secretes a myriad of adipokines, including inflammatory and anti-inflammatory cytokines. As the main storage pool of lipids, subcutaneous and visceral adipose tissues undergo marked hypertrophy and hyperplasia in response to nutritional excess leading to hypoxia, adipokine dysregulation, and subsequent low-grade inflammation that is characterized by increased infiltration and activation of innate and adaptive immune cells. The specific localization, physiology, susceptibility to inflammation and the heterogeneity of the inflammatory cell population of each adipose depot are unique and thus dictate the possible complications of adipose tissue chronic inflammation. Several lines of evidence link visceral and particularly perivascular, pericardial, and perirenal adipose tissue inflammation to the development of metabolic syndrome, insulin resistance, type 2 diabetes and cardiovascular diseases. In addition to the implication of the immune system in the regulation of adipose tissue function, adipose tissue immune components are pivotal in detrimental or otherwise favorable adipose tissue remodeling and thermogenesis. Adipose tissue resident and infiltrating immune cells undergo metabolic and morphological adaptation based on the systemic energy status and thus a better comprehension of the metabolic regulation of immune cells in adipose tissues is pivotal to address complications of chronic adipose tissue inflammation. In this review, we discuss the role of adipose innate and adaptive immune cells across various physiological and pathophysiological states that pertain to the development or progression of cardiovascular diseases associated with metabolic disorders. Understanding such mechanisms allows for the exploitation of the adipose tissue-immune system crosstalk, exploring how the adipose immune system might be targeted as a strategy to treat cardiovascular derangements associated with metabolic dysfunctions.
Collapse
Affiliation(s)
- Ibrahim AlZaim
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Safaa H. Hammoud
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon
| | - Houssam Al-Koussa
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Alaa Ghazi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Pharmacology and Therapeutics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Basic Medical Sciences, College of Medicine, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
62
|
Sahukari R, Punabaka J, Bhasha S, Ganjikunta VS, Ramudu SK, Kesireddy SR. Plant Compounds for the Treatment of Diabetes, a Metabolic Disorder: NF-κB as a Therapeutic Target. Curr Pharm Des 2020; 26:4955-4969. [DOI: 10.2174/1381612826666200730221035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022]
Abstract
Background:
The prevalence of diabetes in the world population hás reached 8.8 % and is expected to
rise to 10.4% by 2040. Hence, there is an urgent need for the discovery of drugs against therapeutic targets to
sojourn its prevalence. Previous studies proved that NF-κB serves as a central agent in the development of diabetic
complications.
Objectives:
This review intended to list the natural plant compounds that would act as inhibitors of NF-κB signalling
in different organs under the diabetic condition with their possible mechanism of action.
Methods:
Information on NF-κB, diabetes, natural products, and relation in between them, was gathered from
scientific literature databases such as Pubmed, Medline, Google scholar, Science Direct, Springer, Wiley online
library.
Results and Conclusion:
NF-κB plays a crucial role in the development of diabetic complications because of its
link in the expression of genes that are responsible for organs damage such as kidney, brain, eye, liver, heart,
muscle, endothelium, adipose tissue and pancreas by inflammation, apoptosis and oxidative stress. Activation of
PPAR-α, SIRT3/1, and FXR through many cascades by plant compounds such as terpenoids, iridoids, flavonoids,
alkaloids, phenols, tannins, carbohydrates, and phytocannabinoids recovers diabetic complications. These compounds
also exhibit the prevention of NF-κB translocation into the nucleus by inhibiting NF-κB activators, such
as VEGFR, RAGE and TLR4 receptors, which in turn, prevent the activation of many genes involved in tissue
damage. Current knowledge on the treatment of diabetes by targeting NF-κB is limited, so future studies would
enlighten accordingly.
Collapse
Affiliation(s)
- Ravi Sahukari
- Division of Molecular Biology and Ethnopharmacology, Department of Zoology, Sri Venkateswara University, Tirupati, India
| | - Jyothi Punabaka
- Division of Molecular Biology and Ethnopharmacology, Department of Zoology, Sri Venkateswara University, Tirupati, India
| | - Shanmugam Bhasha
- Division of Molecular Biology and Ethnopharmacology, Department of Zoology, Sri Venkateswara University, Tirupati, India
| | - Venkata S. Ganjikunta
- Division of Molecular Biology and Ethnopharmacology, Department of Zoology, Sri Venkateswara University, Tirupati, India
| | - Shanmugam K. Ramudu
- Division of Molecular Biology and Ethnopharmacology, Department of Zoology, Sri Venkateswara University, Tirupati, India
| | - Sathyavelu R. Kesireddy
- Division of Molecular Biology and Ethnopharmacology, Department of Zoology, Sri Venkateswara University, Tirupati, India
| |
Collapse
|
63
|
Belsham DD, Dalvi PS. Insulin signalling in hypothalamic neurones. J Neuroendocrinol 2020; 33:e12919. [PMID: 33227171 DOI: 10.1111/jne.12919] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 12/15/2022]
Abstract
Subsequent to the discovery of insulin by Banting and Best in the Department of Physiology at the University of Toronto 100 years ago, the field of insulin signalling and action has grown at a remarkable pace. Yet, the recognition that insulin action in the brain is critical for whole body homeostasis has only recently been appreciated. The hypothalamus is a key region in the brain that responds to circulating insulin by engaging a complex signalling cascade resulting in the ultimate release of neuropeptides that control hunger and feeding. Disruption of this important feedback system can lead to a phenomenon called cellular insulin resistance, where the neurones cease to sense insulin. The factors contributing to insulin resistance, as well as the resulting detrimental effects, include the induction of neuroinflammation, endoplasmic reticulum stress and alterations in the architecture of the blood-brain barrier that allow transport of insulin into the brain. These manifestations usually change energy balance, causing weight gain, often resulting in obesity and its deadly comorbidities, including type 2 diabetes mellitus, cardiovascular disease and metabolic syndrome. Nonetheless, there is still hope because the signal transduction pathways can be targeted at a number of levels by neurone-specific therapeutics. With the advent of unique cell models for investigating the mechanisms involved in these processes, the discovery of novel targets is increasingly possible. Although we are still looking for a cure for diabetes, Banting and Best would be impressed at how far their discovery has advanced and the contemporary knowledge that has been accumulated based on insulin action.
Collapse
Affiliation(s)
- Denise D Belsham
- Departments of Physiology, Obstetrics and Gynaecology and Medicine, University of Toronto, Toronto, ON, Canada
| | - Prasad S Dalvi
- Biology Department, Morosky College of Health Professions and Sciences, Gannon University, Erie, PA, USA
| |
Collapse
|
64
|
Zieba DA, Biernat W, Barć J. Roles of leptin and resistin in metabolism, reproduction, and leptin resistance. Domest Anim Endocrinol 2020; 73:106472. [PMID: 32265081 DOI: 10.1016/j.domaniend.2020.106472] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/28/2020] [Accepted: 02/29/2020] [Indexed: 12/20/2022]
Abstract
Increased adipose mass can cause insulin resistance and type 2 diabetes mellitus. This phenomenon is related to adipocyte-secreted signaling molecules that affect glucose balance, such as fatty acids, adiponectin, leptin, interleukin-6, tumor necrosis factor-α, and resistin. Among these hormones, leptin and resistin play important roles in regulating weight and glucose metabolism. Leptin and resistin work in both similar and opposite ways, and they interact with each other. Circulating concentrations of leptin and resistin are elevated in models of obesity and rodents fed a high-fat diet. In addition, leptin and resistin are similarly regulated by nutritional status: they are reduced by fasting and increased by feeding. This effect is mediated partially through insulin receptors and glucose transporters. Our latest data provided the first indication that in sheep, intravenous infusion of resistin increases the mean circulating concentrations of leptin and decreases luteinizing hormone in a dose-dependent manner during both the long-day (LD) and short-day seasons. Furthermore, exogenous resistin increased suppressor of cytokine signaling (SOCS)-3 mRNA expression only during the LD season, when the leptin resistance/insensitivity phenomenon was observed in the arcuate nucleus, preoptic area, and anterior pituitary. We concluded that one factor contributing to central leptin resistance is autosuppression, via which leptin and resistin stimulate the expression of SOCS-3, which inhibits leptin signaling. The increased expression of SOCS-3 in response to leptin and resistin may be a pivotal cause of leptin resistance/insensitivity, a pathological situation in obese individuals and a physiological occurrence in sheep during the LD season.
Collapse
Affiliation(s)
- D A Zieba
- Department of Animal Nutrition and Biotechnology, and Fisheries, Agricultural University of Krakow, Al. Mickiewicza 24/28, 30-059 Krakow, Poland.
| | - W Biernat
- Department of Animal Nutrition and Biotechnology, and Fisheries, Agricultural University of Krakow, Al. Mickiewicza 24/28, 30-059 Krakow, Poland
| | - J Barć
- Department of Animal Nutrition and Biotechnology, and Fisheries, Agricultural University of Krakow, Al. Mickiewicza 24/28, 30-059 Krakow, Poland
| |
Collapse
|
65
|
Zhou C, Qin Y, Chen R, Gao F, Zhang J, Lu F. Fenugreek attenuates obesity-induced inflammation and improves insulin resistance through downregulation of iRhom2/TACE. Life Sci 2020; 258:118222. [PMID: 32768577 DOI: 10.1016/j.lfs.2020.118222] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/29/2020] [Accepted: 08/03/2020] [Indexed: 11/16/2022]
Abstract
AIMS We previously reported that fenugreek-derived 4-hydroxyisoleucine ameliorates insulin resistance via regulation of TNF-α converting enzyme (TACE) expression. In the present study, we further investigate the effects and mechanisms of fenugreek on obesity-induced inflammation and insulin signaling in the high-fat diet (HFD)-challenged obese mice. MAIN METHODS After 12 weeks of HFD intervention, mice were treated with the low or high dosages of fenugreek. Serum levels of glucose, insulin, lipid profile, inflammation cytokines, and adipokines were detected. Macrophage infiltration and adipose tissue morphology were observed. Western blot was conducted to investigate the expressions of inactive rhomboid 2 (iRhom2) and TACE as well as other signaling pathways in subcutaneous adipose tissue. KEY FINDINGS We showed that fenugreek significantly suppressed body weight gain and fat accumulation in HFD-challenged obese mice. Meanwhile, fasting glucose, insulin, and HOMA-IR in fenugreek-treated mice were remarkably decreased, which were properly explained by fenugreek-induced activation of the insulin receptor signaling pathway. Moreover, the anti-inflammatory properties of fenugreek were shown by the decrease of systemic and local expressions of pro-inflammatory cytokines as well as reduced macrophage infiltration into adipose tissue. Additionally, fenugreek markedly deactivated NF-κB and JNK pathways. Finally, we demonstrated that fenugreek strikingly repressed the transcriptions and expressions of iRhom2 and TACE. SIGNIFICANCE Fenugreek shows an encouraging and promising property in ameliorating insulin resistance and suppressing inflammation in obesity, which might be realized by fenugreek-mediated inhibition of iRhom2/TACE axis-facilitated TNF-α release from adipocytes.
Collapse
Affiliation(s)
- Cong Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - You Qin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Rui Chen
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Feng Gao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiaoyue Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Furong Lu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
66
|
Tang N, Liu Y, Tian Z, Xu S, Wang M, Chen H, Wang B, Li Y, Wang Y, Yang S, Zhao L, Chen D, Li Z. Characterization, tissue distribution of resistin gene and the effect of fasting and refeeding on resistin mRNA expression in Siberian sturgeon (Acipenser baerii). JOURNAL OF FISH BIOLOGY 2020; 97:508-514. [PMID: 32447775 DOI: 10.1111/jfb.14406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/15/2020] [Accepted: 05/21/2020] [Indexed: 06/11/2023]
Abstract
Resistin as an adipokine identified from rodents in 2001 is involved in many biological processes. However, little is known about this gene in fish. We cloned Siberian sturgeon (Acipenser baerii) resistin cDNA of 795 base pairs, encoding 107 amino acids, which showed 38-40% identity to human and rodents. Real-time quantitative PCR showed that the resistin gene was widely distributed in tissues of Siberian sturgeon, with the highest expression in liver. After fasting for 1, 3, 6 and 10 days, the expression of the resistin gene in the liver of Siberian sturgeon decreased significantly, and after refeeding on the 10 days of fasting the resistin mRNA expression increased rapidly, suggesting that resistin may play an important role in liver in response to starvation. Taken together, these results suggest that resistin may be involved in the regulation of energy homeostasis in liver.
Collapse
Affiliation(s)
- Ni Tang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yanlin Liu
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Zhengzhi Tian
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Shaoqi Xu
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Mei Wang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Hu Chen
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Bin Wang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Ya Li
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yan Wang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Song Yang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Liulan Zhao
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Defang Chen
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Zhiqiong Li
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
67
|
Short-term exposure to air pollution (PM 2.5) induces hypothalamic inflammation, and long-term leads to leptin resistance and obesity via Tlr4/Ikbke in mice. Sci Rep 2020; 10:10160. [PMID: 32576879 PMCID: PMC7311527 DOI: 10.1038/s41598-020-67040-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 05/18/2020] [Indexed: 12/21/2022] Open
Abstract
A previous study demonstrated that a high-fat diet (HFD), administered for one-three-days, induces hypothalamic inflammation before obesity’s established, and the long term affects leptin signaling/action due to inflammation. We investigate whether exposure to particulate matter of a diameter of ≤2.5 μm (PM2.5) in mice fed with a chow diet leads to similar metabolic effects caused by high-fat feeding. Compared to the filtered air group (FA), one-day-exposure-PM2.5 did not affect adiposity. However, five-days-exposure-PM2.5 increased hypothalamic microglia density, toll-like-receptor-4 (Tlr4), and the inhibitor-NF-kappa-B-kinase-epsilon (Ikbke) expression. Concurrently, fat mass, food intake (FI), and ucp1 expression in brown adipose tissue were also increased. Besides, decreased hypothalamic STAT3-phosphorylation and Pomc expression were found after twelve-weeks-exposure-PM2.5. These were accompanied by increased FI and lower energy expenditure (EE), leading to obesity, along with increased leptin and insulin levels and HOMA. Mechanistically, the deletion of Tlr4 or knockdown of the Ikbke gene in the hypothalamus was sufficient to reverse the metabolic outcomes of twelve-weeks-exposure-PM2.5. These data demonstrated that short-term exposure-PM2.5 increases hypothalamic inflammation, similar to a HFD. Long-term exposure-PM2.5 is even worse, leading to leptin resistance, hyperphagia, and decreased EE. These effects are most likely due to chronic hypothalamic inflammation, which is regulated by Tlr4 and Ikbke signaling.
Collapse
|
68
|
Elieh Ali Komi D, Shafaghat F, Christian M. Crosstalk Between Mast Cells and Adipocytes in Physiologic and Pathologic Conditions. Clin Rev Allergy Immunol 2020; 58:388-400. [PMID: 32215785 PMCID: PMC7244609 DOI: 10.1007/s12016-020-08785-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Excessive fatty acids and glucose uptake support the infiltration of adipose tissue (AT) by a variety of immune cells including neutrophils, pro-inflammatory M1 macrophages, and mast cells (MCs). These cells promote inflammation by releasing pro-inflammatory mediators. The involvement of MCs in AT biology is supported by their accumulation in the AT of obese individuals along with significantly higher serum levels of MC-derived tryptase. AT-resident MCs under the influence of locally derived adipokines such as leptin become activated and release pro-inflammatory cytokines including TNFα that worsens the inflammatory state. MCs support angiogenesis in AT by releasing chymase and inducing preadipocyte differentiation and also the proliferation of adipocytes through 15-deoxy-delta PGJ2/PPARγ interaction. Additionally, they contribute to the remodeling of the AT extracellular matrix (ECM) and play a role in the recruitment and activation of leukocytes. MC degranulation has been linked to brown adipocyte activation, and evidence indicates an important link between MCs and the appearance of BRITE/beige adipocytes in white AT. Cell crosstalk between MCs and AT-resident cells, mainly adipocytes and immune cells, shows that these cells play a critical role in the regulation of AT homeostasis and inflammation.
Collapse
Affiliation(s)
- Daniel Elieh Ali Komi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farzaneh Shafaghat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mark Christian
- School of Science and Technology, Nottingham, NG11 8NS, UK.
| |
Collapse
|
69
|
Shook LL, Kislal S, Edlow AG. Fetal brain and placental programming in maternal obesity: A review of human and animal model studies. Prenat Diagn 2020; 40:1126-1137. [PMID: 32362000 DOI: 10.1002/pd.5724] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 03/26/2020] [Accepted: 04/21/2020] [Indexed: 12/13/2022]
Abstract
Both human epidemiologic and animal model studies demonstrate that prenatal and lactational exposure to maternal obesity and high-fat diet are associated with adverse neurodevelopmental outcomes in offspring. Neurodevelopmental outcomes described in offspring of obese women include cognitive impairment, autism spectrum disorder (ASD), attention deficit hyperactivity disorder, anxiety and depression, disordered eating, and propensity for reward-driven behavior, among others. This review synthesizes human and animal data linking maternal obesity and high-fat diet consumption to abnormal fetal brain development, and neurodevelopmental and psychiatric morbidity in offspring. It highlights key mechanisms by which maternal obesity and maternal diet impact fetal and offspring development, and sex differences in offspring programming. In addition, we review placental effects of maternal obesity, and the role the placenta might play as an indicator vs mediator of fetal programming.
Collapse
Affiliation(s)
- Lydia L Shook
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sezen Kislal
- Massachusetts General Hospital Research Institute, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Andrea G Edlow
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Massachusetts General Hospital Research Institute, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
70
|
Guevara-Aguirre J, Teran E, Lescano D, Guevara A, Guevara C, Longo V, Gavilanes AWD. Growth hormone receptor deficiency in humans associates to obesity, increased body fat percentage, a healthy brain and a coordinated insulin sensitivity. Growth Horm IGF Res 2020; 51:58-64. [PMID: 32145513 DOI: 10.1016/j.ghir.2020.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/15/2020] [Accepted: 02/17/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND We have shown that subjects with Laron syndrome (LS) due to growth hormone receptor deficiency (GHRD) and their relatives have comparable brain structure and function; moreover, the brain of individuals affected with GHRD appears like those of younger people. While the functionally absent growth hormone receptor and the diminished concentrations of the insulin-like growth factor-I have been associated to these findings, the role of the insulin-glucose axis is emerging as an unavoidable consideration when determining the aetiology of these observations. In consequence, we decided to search for the potential and discrete associations between the neurological findings and several parameters of carbohydrate metabolism that might exist in the subjects affected with GHRD. SUBJECTS AND METHODS Individuals affected with GHRD were compared to relative controls. Besides standard measures of anthropometry, body composition and brain characteristics, the elements of the carbohydrate metabolism (CHO), including glucose, insulin, triacylglycerol and the free insulin growth factor binding protein 1 (IGFBP1) concentrations were determined. In addition, the correlations existing between the parameters of CHO and brain characteristics were established. RESULTS Besides the phenotypical characteristics of GHRD subjects, including those of brain structure and function, enhanced insulin sensitivity, and other minor, we observed that the insulin-regulated IGFBP1 had a consistent negative correlation with the main elements of the carbohydrate metabolism only in the individuals affected with the disease, and not in their relatives. CONCLUSIONS When compared to their relatives, subjects with GHRD who lack the counter-regulatory effects of GH on the insulin axis, despite their increased risk factor profile due to obesity and increased body fat percentage, have a healthy and younger looking brain associated to an enhanced and coordinated insulin sensitivity. Furthermore, it was observed that in the GHRD subjects IGFBP1 negatively correlates, in a constant and systematic manner, with the main elements of the CHO metabolism. These observations suggest a direct relationship between an efficient insulin sensitivity and a healthy brain.
Collapse
Affiliation(s)
- Jaime Guevara-Aguirre
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito, Diego de Robles s/n y Pampite, Cumbaya, Quito, Ecuador; Instituto de Endocrinología IEMYR, Quito, Ecuador; Maastricht University, Maastricht, The Netherlands..
| | - Enrique Teran
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito, Diego de Robles s/n y Pampite, Cumbaya, Quito, Ecuador
| | - Daniela Lescano
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito, Diego de Robles s/n y Pampite, Cumbaya, Quito, Ecuador
| | | | | | - Valter Longo
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | | |
Collapse
|
71
|
Role of c-Jun N-terminal Kinase (JNK) in Obesity and Type 2 Diabetes. Cells 2020; 9:cells9030706. [PMID: 32183037 PMCID: PMC7140703 DOI: 10.3390/cells9030706] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/16/2020] [Accepted: 03/11/2020] [Indexed: 12/13/2022] Open
Abstract
Obesity has been described as a global epidemic and is a low-grade chronic inflammatory disease that arises as a consequence of energy imbalance. Obesity increases the risk of type 2 diabetes (T2D), by mechanisms that are not entirely clarified. Elevated circulating pro-inflammatory cytokines and free fatty acids (FFA) during obesity cause insulin resistance and ß-cell dysfunction, the two main features of T2D, which are both aggravated with the progressive development of hyperglycemia. The inflammatory kinase c-jun N-terminal kinase (JNK) responds to various cellular stress signals activated by cytokines, free fatty acids and hyperglycemia, and is a key mediator in the transition between obesity and T2D. Specifically, JNK mediates both insulin resistance and ß-cell dysfunction, and is therefore a potential target for T2D therapy.
Collapse
|
72
|
Lorigo M, Mariana M, Oliveira N, Lemos MC, Cairrao E. Vascular Pathways of Testosterone: Clinical Implications. J Cardiovasc Transl Res 2019; 13:55-72. [DOI: 10.1007/s12265-019-09939-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 11/15/2019] [Indexed: 12/17/2022]
|
73
|
Resistin in Human Seminal Plasma: Relationship with Lipid Peroxidation, CAT Activity, GSH/GSSG Ratio, and Semen Parameters. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2192093. [PMID: 31772701 PMCID: PMC6854241 DOI: 10.1155/2019/2192093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/26/2019] [Accepted: 09/07/2019] [Indexed: 12/14/2022]
Abstract
Resistin is an adipokine involved in inflammation and able to induce the expression of other proinflammatory cytokines. It is known that, in human semen, resistin is correlated with inflammatory cytokines and sperm quality. The aim of this prospective study was to explore the potential relationship between resistin, lipid peroxidation (LPO), catalase (CAT) activity, and reduced and oxidized glutathione (GSH/GSSG) ratio in semen samples of infertile patients with leukocytospermia (no. 19), infertile patients with varicocele (no. 17), and fertile men (no. 17). Semen analysis was performed following the WHO guidelines, and sperm apoptosis and necrosis were evaluated with annexin V/propidium iodide assay. Seminal plasma samples were used to determine resistin levels by an immunological method, MDA concentration by a HPLC analysis with UV detection, GSH/GSSG ratio by an enzymatic method, CAT activity by a spectrophotometric method. The results showed that, in both groups of infertile patients, semen parameters were significantly reduced (P < 0.001) and sperm apoptosis and necrosis percentages were increased. Resistin levels were significantly higher in leukocytospermia and varicocele groups (P < 0.001 and P < 0.01, respectively) as well as MDA concentration (P < 0.001) compared to controls. The MDA level was also significantly increased in the leukocytospermia group versus the varicocele group (P < 0.05). The GSH/GSSG ratio was higher in fertile controls than the leukocytospermia group (P < 0.05) and the varicocele group (P < 0.001) and in the leukocytospermia group versus the varicocele group (P < 0.05). Both the leukocytospermia and varicocele groups showed increased values of CAT activities (P < 0.001) than controls. Briefly, the correlation between variables, calculated in the whole patient population, showed that resistin levels positively correlated with MDA levels, CAT activity, sperm apoptosis, and necrosis and negatively with sperm parameters and GSH/GSSG ratio. These results support an active role of resistin in an inflammatory process causing LPO, increase of CAT activity, and decrease of GSH/GSSG ratio in seminal plasma of infertile men vs. fertile controls.
Collapse
|
74
|
Pattern Recognition Receptor-Mediated Chronic Inflammation in the Development and Progression of Obesity-Related Metabolic Diseases. Mediators Inflamm 2019; 2019:5271295. [PMID: 31582899 PMCID: PMC6754942 DOI: 10.1155/2019/5271295] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022] Open
Abstract
Obesity-induced chronic inflammation is known to promote the development of many metabolic diseases, especially insulin resistance, type 2 diabetes mellitus, nonalcoholic fatty liver disease, and atherosclerosis. Pattern recognition receptor-mediated inflammation is an important determinant for the initiation and progression of these metabolic diseases. Here, we review the major features of the current understanding with respect to obesity-related chronic inflammation in metabolic tissues, focus on Toll-like receptors and nucleotide-binding oligomerization domain-like receptors with an emphasis on how these receptors determine metabolic disease progression, and provide a summary on the development and progress of PRR antagonists for therapeutic intervention.
Collapse
|
75
|
Hausmann J, Waechtershaeuser A, Behnken I, Aksan A, Blumenstein I, Brenner M, Loitsch SM, Stein J. The role of adipokines in the improvement of diabetic and cardiovascular risk factors within a 52-week weight-loss programme for obesity. Obes Res Clin Pract 2019; 13:440-447. [PMID: 31591082 DOI: 10.1016/j.orcp.2019.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/17/2019] [Accepted: 09/23/2019] [Indexed: 01/01/2023]
|
76
|
García-Jacobo RE, Uresti-Rivera EE, Portales-Pérez DP, González-Amaro R, Lara-Ramírez EE, Enciso-Moreno JA, García-Hernández MH. Circulating miR-146a, miR-34a and miR-375 in type 2 diabetes patients, pre-diabetic and normal-glycaemic individuals in relation to β-cell function, insulin resistance and metabolic parameters. Clin Exp Pharmacol Physiol 2019; 46:1092-1100. [PMID: 31355469 DOI: 10.1111/1440-1681.13147] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 12/16/2022]
Abstract
The pathogenesis of type 2 diabetes (T2D) is associated with a progressive loss of pancreatic β-cell mass. It is known that miR-146a, miR-34a, and miR-375 are involved in β-cell functionality. In this work, we evaluated the levels of these miRNAs in normal-glycaemic individuals, pre-diabetic, and T2D patients in relation to β-cell functionality, insulin resistance, and metabolic parameters. The relative expression of the miRNAs was evaluated in serum samples by real-time polymerase chain reaction. In a principal component analysis, we observed that T2D patients and pre-diabetic individuals were not associated with β-cell functionality. However, in a correlation matrix analysis, we detected that miR-34a was related to miR-146a and insulin resistance. The relative expression of miR-375 was correlated with cholesterol and low-density lipoprotein levels. A decrease of β-cell function in pre-diabetic individuals and T2D patients was observed. The insulin resistance was higher in pre-diabetic individuals and T2D patients. The relative expression of miR-146a in pre-diabetic individuals, T2D patients with insulin treatment, and T2D patients with nephropathy and diabetic foot was decreased. In addition, miR-34a was increased in T2D patients who were overweight and obese. The relative expression of miR-375 was increased in T2D patients with poor glycaemic control, while a decrease was seen in T2D patients with nephropathy and diabetic foot. Circulating miR-375, miR-34a, and miR-146a were not associated with β-cell functionality, but their expression was differentially affected by glycaemia, obesity, insulin treatment, and the presence of nephropathy and diabetic foot.
Collapse
Affiliation(s)
- Rocío E García-Jacobo
- Unidad de Investigación Biomédica, Delegación Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas, México
| | - Edith E Uresti-Rivera
- Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autonóma de San Luis Potosí, San Luis Potosí, México
| | - Diana P Portales-Pérez
- Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autonóma de San Luis Potosí, San Luis Potosí, México
| | - Roberto González-Amaro
- Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autonóma de San Luis Potosí, San Luis Potosí, México
| | - Edgar E Lara-Ramírez
- Unidad de Investigación Biomédica, Delegación Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas, México
| | - José A Enciso-Moreno
- Unidad de Investigación Biomédica, Delegación Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas, México
| | - Mariana H García-Hernández
- Unidad de Investigación Biomédica, Delegación Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas, México
| |
Collapse
|