1
|
Liu Z, Li Q, Zhao F, Chen J. A decade review on phytochemistry and pharmacological activities of Cynomorium songaricum Rupr.: Insights into metabolic syndrome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156602. [PMID: 40058318 DOI: 10.1016/j.phymed.2025.156602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/12/2025] [Accepted: 03/01/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Cynomorium songaricum Rupr. (CSR), a perennial herb with a rich history in traditional medicine, has demonstrated therapeutic potential against metabolic syndrome (MetS) through its active compounds, including proanthocyanidins, polysaccharides, and triterpenoids. MetS, a global health concern, encompasses interlinked conditions such as obesity, type 2 diabetes mellitus (T2DM), and inflammation. This review synthesizes recent findings on CSR's pharmacological and phytochemical properties, focusing on its role in ameliorating MetS. METHODS Following Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines, relevant studies were retrieved from PubMed, Web of Science, and CNKI databases up to December 2024. Keywords included "Cynomorium Songaricum Rupr.", "Cynomorii Herba", "Suoyang", "Suo Yang", "Metabolic syndrome", "Proanthocyanidins", "Polysaccharides" and "Triterpenoids" and their combinations. Inclusion criteria emphasized studies exploring CSR's impact on MetS, while duplicate, low-quality studies and studies not written in Chinese, English, or unrelated were excluded. RESULTS A total of 92 studies were analyzed, revealing that CSR's active components exhibit multi-target effects. Proanthocyanidins reduce glucose absorption and oxidative stress, polysaccharides enhance insulin sensitivity and gut microbiota composition, and triterpenoids mitigate obesity and mitochondria damage. These mechanisms collectively contribute to the beneficial effects of CSR against MetS. CONCLUSION CSR presents a promising natural therapy for MetS, utilizing its pharmacologically active compounds to address core metabolic dysfunctions. Future studies should focus on clinical validation and safety assessments to facilitate CSR's integration into modern therapeutic regimens.
Collapse
Affiliation(s)
- Zhihao Liu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China; The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, China
| | - Qihao Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Fu Zhao
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Jihang Chen
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China; The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, China.
| |
Collapse
|
2
|
Shakoor H, Kizhakkayil J, Statsenko Y, Platat C. Separate and Combined Effects of Moderate-Intensity Exercise Training and Detraining with Protocatechuic Acid (PCA) on Myokines and Insulin-Signaling Pathways in Male Wistar Rats: A Preclinical Randomized Study. Metabolites 2025; 15:87. [PMID: 39997712 PMCID: PMC11857820 DOI: 10.3390/metabo15020087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Exercise training positively modulates myokine secretion and improves glucose metabolism. Herein, we analyzed the effect of moderate-intensity training, detraining, and Protocatechuic Acid (PCA) supplementation on myokine secretions and regulation of insulin-signaling pathways. Methods: A five-arm study was conducted on 47 healthy male Wistar rats, trained at a moderate intensity level for four weeks (T0-T4). Animals were randomly classified into groups according to PCA supplementation and exercise durations: four weeks of Aerobic Training with or without PCA (AT4, AT4-PCA), eight weeks of Aerobic Training with or without PCA (AT8, AT8-PCA), and PCA Vehicle Control (VC). The animals were followed up until week 12 (T12). We decapitated six rats at T0 and T4, four rats per group at T8, and three rats per group at T12. Myokines (IGF-1, IL-6, FGF-21, myostatin, and irisin) were analyzed with ELISA. Western blot analysis measured protein expression of insulin-signaling pathways and GLUT-4 in the gastrocnemius muscle. Results: The IL-6 levels increased significantly (p < 0.01) with 8-week training in AT8 by 34% and AT8-PCA by 32%, compared to groups trained for only 4 weeks (AT4 and AT4-PCA). Similarly, the PI3K, and GLUT-4 expression improved in AT8 and AT8-PCA at T8. Training for 4 weeks improved IGF-1 levels, but a further 14% improvement was observed with 8-week training in AT8 at T8. Myostatin level significantly dropped by 27% even with 4-week training (p < 0.001). However, detraining increased the myostatin levels in all groups, but in AT8-PCA with PCA dose, myostatin reduced by 11% compared to AT8 at T12. PCA supplementation reduced the FGF-21 levels by 54% during detraining at T12 in AT8-PCA compared to AT8. However, the irisin level did not change markedly in any group. Conclusions: Physical training (with and without PCA) modulates myokine production and improves glucose metabolism, but the benefits are lost after detraining.
Collapse
Affiliation(s)
- Hira Shakoor
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 15551, United Arab Emirates; (H.S.)
- Department of Nutrition and Exercise Physiology, Elson S Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| | - Jaleel Kizhakkayil
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 15551, United Arab Emirates; (H.S.)
| | - Yauhen Statsenko
- Department of Radiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 15551, United Arab Emirates;
| | - Carine Platat
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 15551, United Arab Emirates; (H.S.)
| |
Collapse
|
3
|
Machado-Pereira NAMM, do Nascimento PS, de Freitas GR, Bobinski F, do Espírito Santo CC, Ilha J. Electrical Stimulation Prevents Muscular Atrophy and the Decrease of Interleukin-6 in Paralyzed Muscles after Spinal Cord Injury in Rats. Rev Bras Ortop 2024; 59:e526-e531. [PMID: 39239572 PMCID: PMC11374404 DOI: 10.1055/s-0044-1787767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/20/2024] [Indexed: 09/07/2024] Open
Abstract
Objective To analyze the muscle trophism and expression of interleukin-6 in the biceps brachii muscle of rats with incomplete cervical spinal cord injury treated with neuromuscular electrical stimulation (NMES). Methods Adult rats underwent C5-C7 spinal cord hemisection and a 5-week NMES protocol. Trophism of the biceps brachii was assessed using muscle weight/body weight ratio and histological analysis. Interleukin-6 expression from biceps brachii was measured using the enzyme-linked immunosorbent assay technique. Results Preservation of the biceps brachii muscle trophism was found in the NMES treated group, along with prevention of the reduction of interleukin-6 levels. Conclusion Spinal cord injury causes muscle atrophy and decreases interleukin-6 levels. These alterations are partially prevented by NMES. The results suggest a possible NMES action mechanism and underscore the clinical use of this therapeutic tool.
Collapse
Affiliation(s)
- Nicolas A M M Machado-Pereira
- Núcleo de Pesquisa em Lesão da Medula Espinal (NULEME), Departamento de Fisioterapia, Centro de Ciências da Saúde e do Esporte (CEFID), Universidade do Estado de Santa Catarina (UDESC), Florianópolis, SC, Brasil
| | - Patrícia S do Nascimento
- Departamento de Fisiologia e Farmacologia, Centro de Ciências da Saúde (CCS), Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, Brasil
| | - Gabriel R de Freitas
- Núcleo de Pesquisa em Lesão da Medula Espinal (NULEME), Departamento de Fisioterapia, Centro de Ciências da Saúde e do Esporte (CEFID), Universidade do Estado de Santa Catarina (UDESC), Florianópolis, SC, Brasil
| | - Franciane Bobinski
- Laboratório Experimental de Neurociências (LANEX), Universidade do Sul de Santa Catarina (UNISUL), Palhoça, SC, Brasil
| | | | - Jocemar Ilha
- Núcleo de Pesquisa em Lesão da Medula Espinal (NULEME), Departamento de Fisioterapia, Centro de Ciências da Saúde e do Esporte (CEFID), Universidade do Estado de Santa Catarina (UDESC), Florianópolis, SC, Brasil
| |
Collapse
|
4
|
Wang D, Wang X, Han J, You C, Liu Z, Wu Z. Effect of Lacticaseibacillus casei LC2W Supplementation on Glucose Metabolism and Gut Microbiota in Subjects at High Risk of Metabolic Syndrome: A Randomized, Double-blinded, Placebo-controlled Clinical Trial. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10312-5. [PMID: 38954305 DOI: 10.1007/s12602-024-10312-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2024] [Indexed: 07/04/2024]
Abstract
Metabolic syndrome (MetS) is a global epidemic complex and will cause serious metabolic comorbidities without treatment. A prevention strategy for MetS development has been proposed to modulate gut microbiota by probiotic administration to improve intestinal dysbiosis and benefit the host. Lacticaseibacillus casei LC2W has exhibited positive effects in preventing colitis and anti-hypertension in vivo. However, the effect of L. casei LC2W on subjects at high risk of MetS is unknown. Here, a randomized, double-blinded, placebo-controlled study was conducted on 60 subjects with high risk of MetS, and the hypoglycemic and hypolipidemic activity and possible pathways of L. casei LC2W were inferred from the correlation analysis with gut microbiome composition, function, and clinical phenotypic indicators. The results showed that oral administration of L. casei LC2W could exert significant benefits on weight control, glucose and lipid metabolism, inflammatory and oxidative stress parameters, and SCFA production, as well as modulate the composition of gut microbiota. The relative abundance of Lacticaseibacillus, Bifidobacterium, Dorea, and Blautia was enriched, and their interaction with other gut microbes was strengthened by oral administration of L. casei LC2W, which was beneficial in ameliorating gut inflammation, promoting glucose and lipids degradation pathways, thus alleviated MetS. The present study confirmed the prevention effects of L. casei LC2W towards MetS from aspects of clinical outcomes and microflora modulation, providing an alternative strategy for people at high risk of MetS.Trial registration: The study was proactively registered in ClinicalTrial.gov with the registration number of ChiCTR2000031833 on April 09, 2020.
Collapse
Affiliation(s)
- Danqi Wang
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, 200436, PR China
| | - Xiaohua Wang
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, 200436, PR China
| | - Jin Han
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, 200436, PR China
| | - Chunping You
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, 200436, PR China
| | - Zhenmin Liu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, 200436, PR China
| | - Zhengjun Wu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, 200436, PR China.
| |
Collapse
|
5
|
Hutchinson AL, Liddle DM, Monk JM, Ma DWL, Robinson LE. n-3 and n-6 Polyunsaturated Fatty Acids Modulate Macrophage-Myocyte Inflammatory Crosstalk and Improve Myocyte Insulin Sensitivity. Nutrients 2024; 16:2086. [PMID: 38999834 PMCID: PMC11243049 DOI: 10.3390/nu16132086] [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: 06/07/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024] Open
Abstract
In obesity, circulating saturated fatty acids (SFAs) and inflammatory cytokines interfere with skeletal muscle insulin signaling, leading to whole body insulin resistance. Further, obese skeletal muscle is characterized by macrophage infiltration and polarization to the inflammatory M1 phenotype, which is central to the development of local inflammation and insulin resistance. While skeletal muscle-infiltrated macrophage-myocyte crosstalk is exacerbated by SFA, the effects of other fatty acids, such as n-3 and n-6 polyunsaturated fatty acids (PUFAs), are less studied. Thus, the objective of this study was to determine the effects of long-chain n-3 and n-6 PUFAs on macrophage M1 polarization and subsequent effects on myocyte inflammation and metabolic function compared to SFA. Using an in vitro model recapitulating obese skeletal muscle cells, differentiated L6 myocytes were cultured for 24 h with RAW 264.7 macrophage-conditioned media (MCM), followed by insulin stimulation (100 nM, 20 min). MCM was generated by pre-treating macrophages for 24 h with 100 μM palmitic acid (16:0, PA-control), arachidonic acid (20:4n-6, AA), or docosahexaenoic acid (22:6n-3, DHA). Next, macrophage cultures were stimulated with a physiological dose (10 ng/mL) of lipopolysaccharide for an additional 12 h to mimic in vivo obese endotoxin levels. Compared to PA, both AA and DHA reduced mRNA expression and/or secreted protein levels of markers for M1 (TNFα, IL-6, iNOS; p < 0.05) and increased those for M2 (IL-10, TGF-β; p < 0.05) macrophage polarization. In turn, AA- and DHA-derived MCM reduced L6 myocyte-secreted cytokines (TNFα, IL-6; p < 0.05) and chemokines (MCP-1, MIP-1β; p < 0.05). Only AA-derived MCM increased L6-myocyte phosphorylation of Akt (p < 0.05), yet this was inconsistent with improved insulin signaling, as only DHA-derived MCM improved L6 myocyte glucose uptake (p < 0.05). In conclusion, dietary n-3 and n-6 PUFAs may be a useful strategy to modulate macrophage-myocyte inflammatory crosstalk and improve myocyte insulin sensitivity in obesity.
Collapse
Affiliation(s)
| | | | | | | | - Lindsay E. Robinson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada (J.M.M.); (D.W.L.M.)
| |
Collapse
|
6
|
Ciryam P, Gerzanich V, Simard JM. Interleukin-6 in Traumatic Brain Injury: A Janus-Faced Player in Damage and Repair. J Neurotrauma 2023; 40:2249-2269. [PMID: 37166354 PMCID: PMC10649197 DOI: 10.1089/neu.2023.0135] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Abstract
Traumatic brain injury (TBI) is a common and often devastating illness, with wide-ranging public health implications. In addition to the primary injury, victims of TBI are at risk for secondary neurological injury by numerous mechanisms. Current treatments are limited and do not target the profound immune response associated with injury. This immune response reflects a convergence of peripheral and central nervous system-resident immune cells whose interaction is mediated in part by a disruption in the blood-brain barrier (BBB). The diverse family of cytokines helps to govern this communication and among these, Interleukin (IL)-6 is a notable player in the immune response to acute neurological injury. It is also a well-established pharmacological target in a variety of other disease contexts. In TBI, elevated IL-6 levels are associated with worse outcomes, but the role of IL-6 in response to injury is double-edged. IL-6 promotes neurogenesis and wound healing in animal models of TBI, but it may also contribute to disruptions in the BBB and the progression of cerebral edema. Here, we review IL-6 biology in the context of TBI, with an eye to clarifying its controversial role and understanding its potential as a target for modulating the immune response in this disease.
Collapse
Affiliation(s)
- Prajwal Ciryam
- Shock Trauma Neurocritical Care, Program in Trauma, R Adams Cowley Shock Trauma Center, University of Maryland Medical System, Baltimore, Maryland, USA
- Department of Neurology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Orange ST, Leslie J, Ross M, Mann DA, Wackerhage H. The exercise IL-6 enigma in cancer. Trends Endocrinol Metab 2023; 34:749-763. [PMID: 37633799 DOI: 10.1016/j.tem.2023.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/28/2023]
Abstract
Interleukin (IL)-6 elicits both anticancer and procancer effects depending on the context, which we have termed the 'exercise IL-6 enigma'. IL-6 is released from skeletal muscles during exercise to regulate short-term energy availability. Exercise-induced IL-6 provokes biological effects that may protect against cancer by improving insulin sensitivity, stimulating the production of anti-inflammatory cytokines, mobilising immune cells, and reducing DNA damage in early malignant cells. By contrast, IL-6 continuously produced by leukocytes in inflammatory sites drives tumorigenesis by promoting chronic inflammation and activating tumour-promoting signalling pathways. How can a molecule have such opposing effects on cancer? Here, we review the roles of IL-6 in chronic inflammation, tumorigenesis, and exercise-associated cancer prevention and define the factors that underpin the exercise IL-6 enigma.
Collapse
Affiliation(s)
- Samuel T Orange
- Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK; School of Biomedical, Nutritional and Sport Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
| | - Jack Leslie
- Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK; Newcastle Fibrosis Research Group, Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Mark Ross
- Institute of Life and Earth Sciences, School of Energy, Geoscience, Infrastructure and Society, Heriot-Watt University, Edinburgh, UK
| | - Derek A Mann
- Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK; Newcastle Fibrosis Research Group, Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Henning Wackerhage
- Department of Sport & Health Science, Technical University of Munich, Munich, Germany
| |
Collapse
|
8
|
Ren J, Wang XQ, Nakao T, Libby P, Shi GP. Differential Roles of Interleukin-6 in Severe Acute Respiratory Syndrome-Coronavirus-2 Infection and Cardiometabolic Diseases. CARDIOLOGY DISCOVERY 2023; 3:166-182. [PMID: 38152628 PMCID: PMC10750760 DOI: 10.1097/cd9.0000000000000096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection can lead to a cytokine storm, unleashed in part by pyroptosis of virus-infected macrophages and monocytes. Interleukin-6 (IL-6) has emerged as a key participant in this ominous complication of COVID-19. IL-6 antagonists have improved outcomes in patients with COVID-19 in some, but not all, studies. IL-6 signaling involves at least 3 distinct pathways, including classic-signaling, trans-signaling, and trans-presentation depending on the localization of IL-6 receptor and its binding partner glycoprotein gp130. IL-6 has become a therapeutic target in COVID-19, cardiovascular diseases, and other inflammatory conditions. However, the efficacy of inhibition of IL-6 signaling in metabolic diseases, such as obesity and diabetes, may depend in part on cell type-dependent actions of IL-6 in controlling lipid metabolism, glucose uptake, and insulin sensitivity owing to complexities that remain to be elucidated. The present review sought to summarize and discuss the current understanding of how and whether targeting IL-6 signaling ameliorates outcomes following SARS-CoV-2 infection and associated clinical complications, focusing predominantly on metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Jingjing Ren
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Xiao-Qi Wang
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Tetsushi Nakao
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Peter Libby
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
9
|
Loss of brain energy metabolism control as a driver for memory impairment upon insulin resistance. Biochem Soc Trans 2023; 51:287-301. [PMID: 36606696 DOI: 10.1042/bst20220789] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023]
Abstract
The pathophysiological mechanisms intersecting metabolic and neurodegenerative disorders include insulin resistance, which has a strong involvement of environmental factors. Besides central regulation of whole-body homeostasis, insulin in the central nervous system controls molecular signalling that is critical for cognitive performance, namely signalling through pathways that modulate synaptic transmission and plasticity, and metabolism in neurons and astrocytes. This review provides an overview on how insulin signalling in the brain might regulate brain energy metabolism, and further identified molecular mechanisms by which brain insulin resistance might impair synaptic fuelling, and lead to cognitive deterioration.
Collapse
|
10
|
Drummond FR, Leite LB, de Miranda DC, Drummond LR, Lavorato VN, Soares LL, Neves CA, Natali AJ. Skeletal muscle dysfunctions in pulmonary arterial hypertension: Effects of aerobic exercise training. Front Physiol 2023; 14:1148146. [PMID: 37035672 PMCID: PMC10076612 DOI: 10.3389/fphys.2023.1148146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Pulmonary arterial hypertension is associated with skeletal muscle myopathy and atrophy and impaired exercise tolerance. Aerobic exercise training has been recommended as a non-pharmacological therapy for deleterious effects imposed by pulmonary arterial hypertension. Aerobic physical training induces skeletal muscle adaptations via reduced inflammation, improved anabolic processes, decreased hypoxia and regulation of mitochondrial function. These benefits improve physical exertion tolerance and quality of life in patients with pulmonary arterial hypertension. However, the mechanisms underlying the therapeutic potential of aerobic exercise to skeletal muscle disfunctions in patients with pulmonary arterial hypertension are not well understood yet. This minireview highlights the pathways involved in skeletal muscle adaptations to aerobic exercise training in patients with pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Filipe Rios Drummond
- Department of General Biology, Laboratory of Structural Biology, Federal University of Viçosa, Viçosa, MG, Brazil
| | - Luciano Bernardes Leite
- Department of Physical Education, Laboratory of Exercise Biology Federal University of Viçosa, Viçosa, MG, Brazil
- *Correspondence: Luciano Bernardes Leite,
| | - Denise Coutinho de Miranda
- Department of Biological Sciences, Laboratory of Cell Signaling, Federal University of Ouro Preto, Viçosa, MG, Brazil
- Department of Physical Education, Governador Ozanam Coelho University Center (UNIFAGOC), Ubá, Brazil
| | - Lucas Rios Drummond
- Department of Physiology and Biophysics, Laboratory of Endocrinology and Metabolism, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Victor Neiva Lavorato
- Department of Physical Education, Governador Ozanam Coelho University Center (UNIFAGOC), Ubá, Brazil
| | - Leôncio Lopes Soares
- Department of Physical Education, Laboratory of Exercise Biology Federal University of Viçosa, Viçosa, MG, Brazil
| | - Clóvis Andrade Neves
- Department of General Biology, Laboratory of Structural Biology, Federal University of Viçosa, Viçosa, MG, Brazil
| | - Antônio José Natali
- Department of Physical Education, Laboratory of Exercise Biology Federal University of Viçosa, Viçosa, MG, Brazil
| |
Collapse
|
11
|
Insulin and Insulin-Like Growth Factor 1 Signaling Preserves Sarcomere Integrity in the Adult Heart. Mol Cell Biol 2022; 42:e0016322. [PMID: 36125265 PMCID: PMC9583714 DOI: 10.1128/mcb.00163-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Insulin and insulin-like growth factor 1 (IGF1) signaling is transduced by insulin receptor substrate 1 (IRS1) and IRS2. To elucidate physiological and redundant roles of insulin and IGF1 signaling in adult hearts, we generated mice with inducible cardiomyocyte-specific deletion of insulin and IGF1 receptors or IRS1 and IRS2. Both models developed dilated cardiomyopathy, and most mice died by 8 weeks post-gene deletion. Heart failure was characterized by cardiomyocyte loss and disarray, increased proapoptotic signaling, and increased autophagy. Suppression of autophagy by activating mTOR signaling did not prevent heart failure. Transcriptional profiling revealed reduced serum response factor (SRF) transcriptional activity and decreased mRNA levels of genes encoding sarcomere and gap junction proteins as early as 3 days post-gene deletion, in concert with ultrastructural evidence of sarcomere disruption and intercalated discs within 1 week after gene deletion. These data confirm conserved roles for constitutive insulin and IGF1 signaling in suppressing autophagic and apoptotic signaling in the adult heart. The present study also identifies an unexpected role for insulin and IGF1 signaling in regulating an SRF-mediated transcriptional program, which maintains expression of genes encoding proteins that support sarcomere integrity in the adult heart, reduction of which results in rapid development of heart failure.
Collapse
|
12
|
Jin L, Dang H, Wu J, Yuan L, Chen X, Yao J. Weizmannia coagulans BC2000 Plus Ellagic Acid Inhibits High-Fat-Induced Insulin Resistance by Remodeling the Gut Microbiota and Activating the Hepatic Autophagy Pathway in Mice. Nutrients 2022; 14:4206. [PMID: 36235858 PMCID: PMC9572659 DOI: 10.3390/nu14194206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/27/2022] [Accepted: 10/07/2022] [Indexed: 11/05/2022] Open
Abstract
(1) Background: Ellagic acid (EA) acts as a product of gut microbiota transformation to prevent insulin resistance, which is limited by high-fat diet (HFD)-induced dysbiosis. The aim of this study was to investigate the synergistic effects and mechanisms of supplementation with the probiotic Weizmannia coagulans (W. coagulans) on the prevention of insulin resistance by EA; (2) Methods: C57BL/6J mice were divided into five groups (n = 10/group): low-fat-diet group, high-fat-diet group, EA intervention group, EA + W. coagulans BC77 group, and EA + W. coagulans BC2000 group; (3) Result: W. coagulans BC2000 showed a synergistic effect on EA's lowering insulin resistance index and inhibiting high-fat diet-induced endotoxemia. The combined effect of BC2000 and EA activated the autophagy pathway in the mouse liver, a urolithin-like effect. This was associated with altered β-diversity of gut microbiota and increased Eggerthellaceae, a potential EA-converting family. Ellagic acid treatment alone and the combined use of ellagic acid and W. coagulans BC77 failed to activate the hepatic autophagy pathway; (4) Conclusions: W. coagulans BC2000 can assist EA in its role of preventing insulin resistance. This study provides a basis for the development of EA-rich functional food supplemented with W. coagulans BC2000.
Collapse
Affiliation(s)
- Long Jin
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- University of Science and Technology of China, Hefei 230026, China
- Probiotics Institute, Hefei 230031, China
| | - Hongyang Dang
- College Life Science & Technology, Xinjiang University, Urumqi 830046, China
- Institute of Nutrition and Health, Qingdao University, Qingdao 266021, China
| | - Jinyong Wu
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Lixia Yuan
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Xiangsong Chen
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Jianming Yao
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
13
|
Antuna-Puente B, Fellahi S, McAvoy C, Fève B, Bastard JP. Interleukins in adipose tissue: Keeping the balance. Mol Cell Endocrinol 2022; 542:111531. [PMID: 34910978 DOI: 10.1016/j.mce.2021.111531] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023]
Abstract
The role of the immune system is to defend the host and preserve the functionality in response to stress. This function is not limited to infection or injury as it also plays a role in the response to overnutrition. Indeed, low-grade chronic activation of the immune system associated with overnutrition may be deleterious, contributing importantly to diabetes and long-term complications, such as cardiovascular disorders. Increasing evidence shows that adipose tissue participates in the obesity-related inflammatory response and that interleukins are one of the key players, either as a pro-inflammatory response to the metabolic dysregulation or to restore homeostasis. The crosstalk between adipocytes and immune cells through some important interleukins and their role in metabolic disruption is the topic of this review.
Collapse
Affiliation(s)
- Barbara Antuna-Puente
- Infection Disease Division, Department of Medicine, Queen's University, Kingston, ON, Canada.
| | - Soraya Fellahi
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Département de Biochimie-pharmacologie-biologie Moléculaire-génétique Médicale, Créteil, France; Sorbonne Université-Inserm, Centre de Recherche Saint-Antoine UMR S_938, 75012, Paris Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Paris, France
| | - Chloé McAvoy
- Unité de Recherche Clinique de L'Est Parisien (URC-Est), Hôpital Saint Antoine, Paris, France
| | - Bruno Fève
- Sorbonne Université-Inserm, Centre de Recherche Saint-Antoine UMR S_938, 75012, Paris Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Paris, France; Assistance Publique- Hôpitaux de Paris -Hôpital Saint-Antoine, Service D'Endocrinologie-Diabétologie, Centre de Référence des Maladies Rares de L'Insulino-Sécrétion et de L'Insulino-Sensibilité (PRISIS), 75012, Paris, France
| | - Jean-Philippe Bastard
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Département de Biochimie-pharmacologie-biologie Moléculaire-génétique Médicale, Créteil, France; FHU-SENEC, INSERM U955 and Université Paris Est (UPEC), UMR U955, Faculté de Santé, Créteil, France
| |
Collapse
|
14
|
Kistner TM, Pedersen BK, Lieberman DE. Interleukin 6 as an energy allocator in muscle tissue. Nat Metab 2022; 4:170-179. [PMID: 35210610 DOI: 10.1038/s42255-022-00538-4] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/21/2022] [Indexed: 12/31/2022]
Abstract
Extensive research has shown that interleukin 6 (IL-6) is a multifunctional molecule that is both proinflammatory and anti-inflammatory, depending on the context. Here, we combine an evolutionary perspective with physiological data to propose that IL-6's context-dependent effects on metabolism reflect its adaptive role for short-term energy allocation. This energy-allocation role is especially salient during physical activity, when skeletal muscle releases large amounts of IL-6. We predict that during bouts of physical activity, myokine IL-6 fulfills the three main characteristics of a short-term energy allocator: it is secreted from muscle in response to an energy deficit, it liberates somatic energy through lipolysis and it enhances muscular energy uptake and transiently downregulates immune function. We then extend this model of energy allocation beyond myokine IL-6 to reinterpret the roles that IL-6 plays in chronic inflammation, as well as during COVID-19-associated hyperinflammation and multiorgan failure.
Collapse
Affiliation(s)
- Timothy M Kistner
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.
| | - Bente K Pedersen
- Centre of Inflammation and Metabolism/Centre for Physical Activity Research (CIM/CFAS), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| | - Daniel E Lieberman
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
15
|
Iheagwam FN, Batiha GES, Ogunlana OO, Chinedu SN. Terminalia catappa Extract Palliates Redox Imbalance and Inflammation in Diabetic Rats by Upregulating Nrf-2 Gene. Int J Inflam 2021; 2021:9778486. [PMID: 34956587 PMCID: PMC8702315 DOI: 10.1155/2021/9778486] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/09/2021] [Accepted: 11/25/2021] [Indexed: 12/30/2022] Open
Abstract
This study aims at evaluating the ameliorative role of Terminalia catappa aqueous leaf extract (TCA) on hyperglycaemia-induced oxidative stress and inflammation in a high-fat, low dose streptozotocin-induced type 2 diabetic rat model. Experimental rats were treated orally with 400 and 800 mg/kg bw TCA daily for four weeks. Antioxidant enzyme activities, plasma glucose concentration, protein concentration, oxidative stress, and inflammation biomarkers were assayed using standard methods. Hepatic relative expressions of tumour necrosis factor-alpha (TNF-α), interleukin-six (IL-6), and nuclear factor-erythroid 2 related factor 2 (Nrf-2) were also assessed. Molecular docking and prediction of major TCA phytoconstituents' biological activity related to T2DM-induced oxidative stress were evaluated in silico. Induction of diabetes significantly (p < 0.05) reduced superoxide dismutase, glutathione-S-transferase, and peroxidase activities. Glutathione and protein stores were significantly (p < 0.05) depleted, while glucose, MDA, interleukin-six (IL-6), and tumour necrosis factor-α (TNF-α) concentrations were significantly (p < 0.05) increased. A significant (p < 0.05) upregulation of hepatic TNF-α and IL-6 expression and downregulation (p < 0.05) of Nrf-2 expression were observed during diabetes onset. TCA treatment significantly (p < 0.05) modulated systemic diabetic-induced oxidative stress and inflammation, mRNA expression dysregulation, and dysregulated macromolecule metabolism. However, only 800 mg/kg TCA treatment significantly (p < 0.05) downregulated hepatic TNF-α expression. 9-Oxabicyclo[3.3.1]nonane-2,6-diol and 1,2,3-Benzenetriol bound comparably to glibenclamide in Nrf-2, IL-6, and TNF-α binding pockets. They were predicted to be GST A and M substrate, JAK2 expression, ribulose-phosphate 3-epimerase, NADPH peroxidase, and glucose oxidase inhibitors. These results suggest that TCA ameliorates hyperglycaemia-induced oxidative stress and inflammation by activating Nrf-2 gene.
Collapse
Affiliation(s)
- Franklyn Nonso Iheagwam
- Department of Biochemistry, Covenant University, P.M.B. 1023 Ota, Ogun State, Nigeria
- Covenant University Public Health and Wellbeing Research Cluster (CUPHWERC), Covenant University, P.M.B. 1023 Ota, Ogun State, Nigeria
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt
| | - Olubanke Olujoke Ogunlana
- Department of Biochemistry, Covenant University, P.M.B. 1023 Ota, Ogun State, Nigeria
- Covenant University Public Health and Wellbeing Research Cluster (CUPHWERC), Covenant University, P.M.B. 1023 Ota, Ogun State, Nigeria
| | - Shalom Nwodo Chinedu
- Department of Biochemistry, Covenant University, P.M.B. 1023 Ota, Ogun State, Nigeria
- Covenant University Public Health and Wellbeing Research Cluster (CUPHWERC), Covenant University, P.M.B. 1023 Ota, Ogun State, Nigeria
| |
Collapse
|
16
|
Lactobacillus reuteri FYNLJ109L1 Attenuating Metabolic Syndrome in Mice via Gut Microbiota Modulation and Alleviating Inflammation. Foods 2021; 10:foods10092081. [PMID: 34574191 PMCID: PMC8469823 DOI: 10.3390/foods10092081] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/28/2021] [Accepted: 08/31/2021] [Indexed: 12/30/2022] Open
Abstract
Metabolic syndrome is caused by an excessive energy intake in a long-term, high-fat and/or high-sugar diet, resulting in obesity and a series of related complications, which has become a global health concern. Probiotics intervention can regulate the gut microbiota and relieve the systemic and chronic low-grade inflammation, which is an alternative to relieving metabolic syndrome. The aim of this work was to explore the alleviation of two different Lactobacillusreuteri strains on metabolic syndrome. Between the two L. reuteri strains, FYNLJ109L1 had a better improvement effect on blood glucose, blood lipid, liver tissue damage and other related indexes than NCIMB 30242. In particular, FYNLJ109L1 reduced weight gain, food intake and fat accumulation. Additionally, it can regulate the gut microbiota, increase IL-10, and reduce IL-6 and tumor necrosis factor-α (TNF-α), as well as liver injury, and further reduce insulin resistance and regulate lipid metabolism disorders. In addition, it could modulate the gut microbiota, particularly a decreased Romboutsia and Clostridium sensu stricto-1, and an increased Acetatifactor. The results indicated that FYNLJ109L1 could improve metabolic syndrome significantly via alleviating inflammation and gut microbiota modulation.
Collapse
|
17
|
Gholami A, Mollanoroozy E, Reza Baradaran H, Hariri M. The efficacy of soy isoflavones combined with soy protein on serum concentration of interleukin-6 and tumour necrosis factor-α among post-menopausal women? A systematic review and meta-analysis of randomized controlled trials. Clin Exp Pharmacol Physiol 2021; 49:10-24. [PMID: 34455600 DOI: 10.1111/1440-1681.13586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 12/15/2022]
Abstract
The post-menopausal stage in women's life is associated with the enhancement of inflammation that may be reduced using soy isoflavones or soy protein. The present study aimed to summarize the effect of soy isoflavones plus soy protein on circulating interleukin-6 (IL-6) and tumour necrosis factor-α (TNF-α) in post-menopausal women. The English-language articles were identified from the databases such as Cochrane Library, clinicaltrials.gov, Web of Science, PubMed, and Scopus until December 2020. The mean change from baseline and its standard deviation (SD) for intervention and comparison groups were used to calculate the effect size. The statistical heterogeneity of the intervention effects was computing by Cochran's Q test and I2 statistic. Nine and seven studies were selected for systematic review and meta-analysis, respectively. The results of our meta-analysis indicated a non-significant effect on the serum concentrations of IL-6 and TNF-α (weighted mean differences [WMD] = 0.07 pg/mL; 95% confidence interval [CI] = -0.03, 0.17 pg/mL; P = 0.190; WMD =0.05 pg/mL; 95% CI = -0.01, 0.12 pg/mL; P = 0.092; respectively). In subgroup analysis, soy isoflavones plus soy protein could increase the serum concentration of IL-6 in studies with soy isoflavones dose ≤87 mg/days, cross-over design, weak quality, and studies on participants who had health risk factors or diseases. The serum concentration of TNF-α increased in studies with cross-over design, intervention duration ≤56 days, and body mass index (BMI) >27, and in studies that were conducted on at-risk or sick participants. In conclusion, our meta-analysis did not confirm any significant effect on serum concentration of IL-6 and TNF-α among post-menopausal women.
Collapse
Affiliation(s)
- Ali Gholami
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.,Department of Epidemiology and Biostatistics, School of Public Health, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Ensiyeh Mollanoroozy
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Hamid Reza Baradaran
- Department of Epidemiology, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.,Ageing Clinical and Experimental Research Team, Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition University of Aberdeen, Aberdeen, UK
| | - Mitra Hariri
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.,Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
18
|
Zheng F, Wang Z, Stanton C, Ross RP, Zhao J, Zhang H, Yang B, Chen W. Lactobacillus rhamnosus FJSYC4-1 and Lactobacillus reuteri FGSZY33L6 alleviate metabolic syndrome via gut microbiota regulation. Food Funct 2021; 12:3919-3930. [PMID: 33977963 DOI: 10.1039/d0fo02879g] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Metabolic syndrome, which includes a series of metabolic disorders such as hyperglycemia, hyperlipidemia, insulin resistance and obesity, has become a catastrophic disease worldwide. Accordingly, probiotic intervention is a new strategy to alleviate metabolic syndrome, which can adjust the gut microbiota to a certain extent. The aim of the current work was to explore the alleviation of metabolic syndrome by Lactobacillus reuteri and L. rhamnosus. Two L. reuteri and two L. rhamnosus strains were administered to mice with a high-fat diet for 12 weeks. All Lactobacillus strains tested significantly slowed weight gain in the mice. Among four strains, L. reuteri FGSZY33L6 and L. rhamnosus FJSYC4-1 showed the strongest ability to relieve blood glucose disorders, blood lipid disorders, tissue damage, and particularly gut microbiota disorders. Thus, our findings indicate that these strains can regulate the gut microbiota and produce short-chain fatty acids (SCFAs), which can induce satiety hormones, inhibit food intake and increase satiety, and thus improve metabolic syndrome.
Collapse
Affiliation(s)
- Fuli Zheng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China. and School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Zhi Wang
- Department of Cardiopulmonary Rehabilitation, Wuxi Tongren Rehabilitation Hospital, Wuxi, China.
| | - Catherine Stanton
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, China and APC Microbiome Ireland, University College Cork, Cork, Ireland and Teagasc Food Research Centre, Moorepark, Co. Cork, Ireland
| | - R Paul Ross
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, China and APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China. and School of Food Science and Technology, Jiangnan University, Wuxi, China and International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China. and School of Food Science and Technology, Jiangnan University, Wuxi, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China and Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China. and School of Food Science and Technology, Jiangnan University, Wuxi, China and International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China. and School of Food Science and Technology, Jiangnan University, Wuxi, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China and Beijing Innovation Center of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing, China
| |
Collapse
|
19
|
Wang CR, Tsai HW. Anti- and non-tumor necrosis factor-α-targeted therapies effects on insulin resistance in rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis. World J Diabetes 2021; 12:238-260. [PMID: 33758645 PMCID: PMC7958474 DOI: 10.4239/wjd.v12.i3.238] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/07/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023] Open
Abstract
In addition to β-cell failure with inadequate insulin secretion, the crucial mechanism leading to establishment of diabetes mellitus (DM) is the resistance of target cells to insulin, i.e. insulin resistance (IR), indicating a requirement of beyond-normal insulin concentrations to maintain euglycemic status and an ineffective strength of transduction signaling from the receptor, downstream to the substrates of insulin action. IR is a common feature of most metabolic disorders, particularly type II DM as well as some cases of type I DM. A variety of human inflammatory disorders with increased levels of proinflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β, have been reported to be associated with an increased risk of IR. Autoimmune-mediated arthritis conditions, including rheumatoid arthritis (RA), psoriatic arthritis (PsA) and ankylosing spondylitis (AS), with the involvement of proinflammatory cytokines as their central pathogenesis, have been demonstrated to be associated with IR, especially during the active disease state. There is an increasing trend towards using biologic agents and small molecule-targeted drugs to treat such disorders. In this review, we focus on the effects of anti-TNF-α- and non-TNF-α-targeted therapies on IR in patients with RA, PsA and AS. Anti-TNF-α therapy, IL-1 blockade, IL-6 antagonist, Janus kinase inhibitor and phospho-diesterase type 4 blocker can reduce IR and improve diabetic hyper-glycemia in autoimmune-mediated arthritis.
Collapse
Affiliation(s)
- Chrong-Reen Wang
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 70403, Taiwan
| | - Hung-Wen Tsai
- Department of Pathology, National Cheng Kung University Hospital, Tainan 70403, Taiwan
| |
Collapse
|
20
|
Testa C, DI Lorenzo A, Parlato A, D'Ambrosio G, Merolla A, Pacileo M, Iannuzzo G, Gentile M, Nugara C, Sarullo FM, DE Gregorio C, D'Andrea A, Vigorito C, Venturini E, Giallauria F. Exercise for slowing the progression of atherosclerotic process: effects on inflammatory markers. Panminerva Med 2021; 63:122-132. [PMID: 33565757 DOI: 10.23736/s0031-0808.21.04266-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Atherosclerosis is a dynamic process driven by all cardiovascular risk factors that can be briefly divided into an early and a late phase. Inflammation is one of the fundamental substrates that initiates the atherosclerotic process in the early stages and promotes and maintains it in the final stages. In the last decades, clinical and experimental data have shown that inflammation is supported by mediators that respond to physical activity. The present review aimed at investigating the effect of physical exercise on inflammatory mediators, both the positive ones that have a proinflammatory effect (interleukin 6, c-reactive protein and tumor necrosis factor α, interferon γ, high-mobility group box-1), and the negative ones which have an anti-inflammatory effect (interleukin 10). Pooled data support the evidence that physical exercise can directly modulate the activity of inflammatory cytokines slowing down or preventing the formation of the atherosclerotic stage.
Collapse
Affiliation(s)
- Crescenzo Testa
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Anna DI Lorenzo
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Alessandro Parlato
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Giuseppe D'Ambrosio
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Aurora Merolla
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Mario Pacileo
- Unit of Cardiology and Intensive Care, "Umberto I" Hospital, Nocera Inferiore, Salerno, Italy
| | - Gabriella Iannuzzo
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Marco Gentile
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Cinzia Nugara
- Unit of Cardiovascular Rehabilitation, Buccheri La Ferla Fatebenefratelli Hospital, Palermo, Italy
| | - Filippo M Sarullo
- Unit of Cardiovascular Rehabilitation, Buccheri La Ferla Fatebenefratelli Hospital, Palermo, Italy
| | - Cesare DE Gregorio
- Unit of Cardiology, Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy.,Post-graduate Residency School in Cardiovascular Diseases, Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy
| | - Antonello D'Andrea
- Unit of Cardiology and Intensive Care, "Umberto I" Hospital, Nocera Inferiore, Salerno, Italy
| | - Carlo Vigorito
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Elio Venturini
- Cardiac Rehabilitation Unit, AUSL Toscana Nord-Ovest, Cecina Civil Hospital, Cecina, Livorno, Italy
| | - Francesco Giallauria
- Department of Translational Medical Sciences, Federico II University, Naples, Italy - .,Faculty of Sciences and Technology, University of New England, Armidale, Australia
| |
Collapse
|
21
|
McNamara EL, Taylor RL, Clayton JS, Goullee H, Dilworth KL, Pinós T, Brull A, Alexander IE, Lisowski L, Ravenscroft G, Laing NG, Nowak KJ. Systemic AAV8-mediated delivery of a functional copy of muscle glycogen phosphorylase (Pygm) ameliorates disease in a murine model of McArdle disease. Hum Mol Genet 2020; 29:20-30. [PMID: 31511858 DOI: 10.1093/hmg/ddz214] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/01/2019] [Accepted: 09/02/2019] [Indexed: 12/12/2022] Open
Abstract
McArdle disease is a disorder of carbohydrate metabolism that causes painful skeletal muscle cramps and skeletal muscle damage leading to transient myoglobinuria and increased risk of kidney failure. McArdle disease is caused by recessive mutations in the muscle glycogen phosphorylase (PYGM) gene leading to absence of PYGM enzyme in skeletal muscle and preventing access to energy from muscle glycogen stores. There is currently no cure for McArdle disease. Using a preclinical animal model, we aimed to identify a clinically translatable and relevant therapy for McArdle disease. We evaluated the safety and efficacy of recombinant adeno-associated virus serotype 8 (rAAV8) to treat a murine model of McArdle disease via delivery of a functional copy of the disease-causing gene, Pygm. Intraperitoneal injection of rAAV8-Pygm at post-natal day 1-3 resulted in Pygm expression at 8 weeks of age, accompanied by improved skeletal muscle architecture, reduced accumulation of glycogen and restoration of voluntary running wheel activity to wild-type levels. We did not observe any adverse reaction to the treatment at 8 weeks post-injection. Thus, we have investigated a highly promising gene therapy for McArdle disease with a clear path to the ovine large animal model endemic to Western Australia and subsequently to patients.
Collapse
Affiliation(s)
- Elyshia L McNamara
- Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia.,Centre for Medical Research, University of Western Australia, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia
| | - Rhonda L Taylor
- Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia.,Centre for Medical Research, University of Western Australia, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia
| | - Joshua S Clayton
- Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia.,Centre for Medical Research, University of Western Australia, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia
| | - Hayley Goullee
- Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia.,Centre for Medical Research, University of Western Australia, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia
| | - Kimberley L Dilworth
- Faculty of Medicine and Health, Vector and Genome Engineering Facility, Children's Medical Research Institute, The University of Sydney, Westmead, NSW 2145, Australia
| | - Tomàs Pinós
- Neuromuscular and Mitochondrial Disorders Laboratory, Vall d'Hebron Institut de Recerca, Universitat Autonoma de Barcelona, Barcelona 08035, Spain.,Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Astrid Brull
- Sorbonne Université, INSERM UMRS_974, Center of Research in Myology, Paris 75013, France
| | - Ian E Alexander
- Gene Therapy Research Unit, Faculty of Medicine and Health, Children's Medical Research Institute, The University of Sydney and Sydney Children's Hospitals Network, Westmead, NSW 2145, Australia.,Discipline of Child and Adolescent Health, Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Westmead, NSW 2145, Australia
| | - Leszek Lisowski
- Faculty of Medicine and Health, Vector and Genome Engineering Facility, Children's Medical Research Institute, The University of Sydney, Westmead, NSW 2145, Australia.,Translational Vectorology Group, Faculty of Medicine and Health, Children's Medical Research Institute, The University of Sydney, Sydney, NSW 2006, Australia.,Military Institute of Hygiene and Epidemiology, The Biological Threats Identification and Countermeasure Centre, Puławy 24-100, Poland
| | - Gianina Ravenscroft
- Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia.,Centre for Medical Research, University of Western Australia, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia
| | - Nigel G Laing
- Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia.,Centre for Medical Research, University of Western Australia, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia
| | - Kristen J Nowak
- Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia.,Faculty of Health and Medical Sciences, School of Biomedical Sciences, University of Western Australia, QEII Medical Centre, Nedlands, WA 6009, Australia.,Public and Aboriginal Health Division, Western Australian Department of Health, Office of Population Health Genomics, East Perth, WA 6004, Australia
| |
Collapse
|
22
|
Chun KH, Cho SJ, Lee JW, Seo JH, Kim KW, Lee SK. Protein kinase C-δ interacts with and phosphorylates ARD1. J Cell Physiol 2020; 236:379-391. [PMID: 32542692 DOI: 10.1002/jcp.29866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 11/07/2022]
Abstract
Protein kinase C-δ (PKCδ) is a diacylglycerol-dependent, calcium-independent novel PKC isoform that is engaged in various cell signaling pathways, such as cell proliferation, apoptosis, inflammation, and oxidative stress. In this study, we searched for proteins that bind PKCδ using a yeast two-hybrid assay and identified murine arrest-defective 1 (mARD1) as a binding partner. The interaction between PKCδ and mARD1 was confirmed by glutathione S-transferase pull-down and co-immunoprecipitation assays. Furthermore, recombinant PKCδ phosphorylated full-length mARD1 protein. The NetPhos online prediction tool suggested PKCδ phosphorylates Ser80 , Ser108 , and Ser114 residues of mARD1 with the highest probability. Based on these results, we synthesized peptides containing these sites and examined their phosphorylations using recombinant PKCδ. Autoradiography confirmed these sites were efficiently phosphorylated. Consequent mass spectrometry and peptide sequencing in combination with MALDI-TOF MS/MS confirmed that Ser80 and Ser108 were major phosphorylation sites. The alanine mutations of Ser80 and Ser108 abolished the phosphorylation of mARD1 by PKCδ in 293T cells supporting these observations. In addition, kinase assays using various PKC isotypes showed that Ser80 of ARD1 was phosphorylated by PKCβI and PKCζ isotypes with the highest selectivity, while Ser108 and/or Ser114 were phosphorylated by PKCγ with activities comparable to that of the PKCδ isoform. Overall, these results suggest the possibility that PKCδ transduces signals by regulating phosphorylation of ARD1.
Collapse
Affiliation(s)
- Kwang-Hoon Chun
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Seung-Ju Cho
- Division of Drug Safety Evaluation, New Drug Development Center, Osong Medical Innovation Foundation, Cheongju, Republic of Korea
| | - Ji-Won Lee
- Preclinical Studies, GlycoMimetics Inc., Rockville, Maryland
- Research Institute for Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Ji Hae Seo
- Department of Biochemistry, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Kyu-Won Kim
- Research Institute for Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Seung-Ki Lee
- Research Institute for Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
23
|
Ahn N, Kim K. Effects of Aerobic and Resistance Exercise on Myokines in High Fat Diet-Induced Middle-Aged Obese Rats. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17082685. [PMID: 32295130 PMCID: PMC7215661 DOI: 10.3390/ijerph17082685] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/10/2020] [Accepted: 04/12/2020] [Indexed: 12/11/2022]
Abstract
The objective of this study was to analyze the effects of aerobic and resistance exercise on myokines expression in the skeletal muscle of middle-aged rats with high fat diet-induced obesity, to investigate the feasibility of using exercise training to reduce inflammation. Male 50-week-old Sprague Dawley rats were divided into normal diet, normal diet + exercise, high fat diet, and high fat diet + exercise groups. After six weeks on a high fat diet to induce obesity, a 12-week exercise program was implemented, which combined aerobic exercise (treadmill running) and resistance exercise (ladder climbing) three times a week for 75 min per session. We analyzed the protein levels of interleukins (IL) 6, 7, and 8, C-X-C motif chemokine receptor 2, and vascular endothelial growth factor in skeletal muscles by western blotting. Body weight decreased significantly during the 12-week exercise program in the exercise groups compared to the non-exercise groups (p < 0.05). The levels of all myokines analyzed were significantly lower in the skeletal muscle of the high fat diet group compared to the normal diet group (p < 0.05). After completing the 12-week exercise program, IL-7, IL-8, C-X-C motif chemokine receptor 2, and vascular endothelial growth factor expressions were significantly higher in the high fat diet + exercise group compared to the high fat diet group (p < 0.05). However, while IL-6 expression was significantly lower in the high fat diet and high fat diet + exercise groups compared to the normal diet group (p < 0.05), it was not significantly affected by exercise. In conclusion, high fat diet-induced obesity resulted in decreased myokines in the skeletal muscles, but combined exercise training of aerobic and resistance exercise increased myokines secretion in the skeletal muscle of obese rats, and is thought to help reduce inflammation.
Collapse
Affiliation(s)
| | - Kijin Kim
- Correspondence: ; Tel.: +82-53-580-5256
| |
Collapse
|
24
|
Long MH, Zhang C, Xu DQ, Fu WL, Gan XD, Li F, Wang Q, Xia W, Xu DG. PM 2.5 aggravates diabetes via the systemically activated IL-6-mediated STAT3/SOCS3 pathway in rats' liver. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 256:113342. [PMID: 31676093 DOI: 10.1016/j.envpol.2019.113342] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 06/10/2023]
Abstract
PM2.5 exposure aggravates type 2 diabetes, in which inflammatory factors play an important role. In this study, we aimed to explore the mechanisms responsible for aggravating diabetes after PM2.5 exposure, and study the roles of inflammatory factors in insulin-resistant type 2 diabetes. Our study indicated that short-time PM2.5 exposure enhances insulin resistance in type 2 diabetic rats and significantly raises inflammatory factors, including IL-6, TNF-α, and MCP-1, in lungs. However, we found that of these inflammatory factors only IL-6 levels are elevated in blood, liver, adipose tissue, and macrophages, but not in skeletal muscle. IL-6 induced activation of the STAT3/SOCS3 pathway in liver, but not other downstream pathways including STAT1, ERK1/2, and PI3K. Both STAT3 inhibition and IL-6 neutralization effectively alleviated the disorders of glucose metabolism after PM2.5 exposure. Taken together, this suggests that the systemic increase in IL-6 may play an important role in the deterioration of the type 2 diabetes via IL-6/STAT3/SOCS3 pathway in liver after short-time exposure to PM2.5. Besides, we unexpectedly found a stronger resistance to the PM2.5 exposure-induced increase in IL-6 in skeleton muscle than those of many other tissues.
Collapse
Affiliation(s)
- Min-Hui Long
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, PR China; The South China Normal University, Guangzhou, 510631, PR China
| | - Chao Zhang
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, PR China
| | - Dong-Qun Xu
- Institute of Products Related with Environment and Health, China Center for Diseases Control, Beijing, 100080, PR China
| | - Wen-Liang Fu
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, PR China
| | - Xiang-Dong Gan
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, PR China; The South China Normal University, Guangzhou, 510631, PR China
| | - Fei Li
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, PR China
| | - Qin Wang
- Institute of Products Related with Environment and Health, China Center for Diseases Control, Beijing, 100080, PR China
| | - Wenrong Xia
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, PR China
| | - Dong-Gang Xu
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, PR China.
| |
Collapse
|
25
|
Su KZ, Li YR, Zhang D, Yuan JH, Zhang CS, Liu Y, Song LM, Lin Q, Li MW, Dong J. Relation of Circulating Resistin to Insulin Resistance in Type 2 Diabetes and Obesity: A Systematic Review and Meta-Analysis. Front Physiol 2019; 10:1399. [PMID: 31803062 PMCID: PMC6877503 DOI: 10.3389/fphys.2019.01399] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/30/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Resistin, a cysteine-rich polypeptide encoded by the RETN gene, which plays an important role in many mechanisms in rodent studies, including lipid metabolism, inflammation and insulin resistance. Nevertheless, the relationship between resistin and insulin resistance in humans is under debate. The present study was designed to clarify the correlation between resistin and insulin resistance. Methods: A systematic literature search was performed using PubMed, Embase and Cochrane Library until March 3, 2019 with the keywords "resistin" and "insulin resistance." Funnel plots and Egger's test were used to detect publication bias. A random-effects model was used to calculate the pooled effect size. Subgroup analysis and meta regression was performed to identify the sources of heterogeneity. Results: Fifteen studies were included in our systematic review. Among them, 10 studies with Pearson coefficients were used for meta-analysis. We found resistin levels were weakly correlated with insulin resistance in those with T2DM and obesity (r = 0.21, 95% CI: 0.06-0.35, I 2 = 59.7%, P = 0.003). Nevertheless, subgroup analysis suggested that circulating resistin levels were significantly positively correlated with insulin resistance in individuals with hyperresistinemia (≥14.8 ng/ml) (r = 0.52, 95% CI: 0.35-0.68, I 2 = 0.0%, P = 0.513). And there was no relationship between circulating resistin and insulin resistance in those with normal circulating resistin levels (<14.8 ng/ml) (r = 0.08, 95% CI: -0.01-0.18, I 2 = 0.0%, P = 0.455). Publication bias was insignificant (Egger's test P = 0.592). Conclusion: In T2DM and obese individuals, resistin levels were positively correlated with insulin resistance in those with hyperresistinemia, but not in those with normal circulating resistin levels.
Collapse
Affiliation(s)
- Kai-zhen Su
- Clinical Medicine Department, Medical College, Qingdao University, Qingdao, China
| | - Yan-run Li
- Clinical Medicine Department, Medical College, Qingdao University, Qingdao, China
| | - Di Zhang
- Special Medicine Department, Medical College, Qingdao University, Qingdao, China
| | - Jun-hua Yuan
- Special Medicine Department, Medical College, Qingdao University, Qingdao, China
| | - Cai-shun Zhang
- Special Medicine Department, Medical College, Qingdao University, Qingdao, China
| | - Yuan Liu
- Special Medicine Department, Medical College, Qingdao University, Qingdao, China
| | - Li-min Song
- Special Medicine Department, Medical College, Qingdao University, Qingdao, China
| | - Qian Lin
- Special Medicine Department, Medical College, Qingdao University, Qingdao, China
| | - Man-wen Li
- Special Medicine Department, Medical College, Qingdao University, Qingdao, China
| | - Jing Dong
- Special Medicine Department, Medical College, Qingdao University, Qingdao, China
- Physiology Department, Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
26
|
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
|
27
|
Tenorio-Jiménez C, Martínez-Ramírez MJ, Del Castillo-Codes I, Arraiza-Irigoyen C, Tercero-Lozano M, Camacho J, Chueca N, García F, Olza J, Plaza-Díaz J, Fontana L, Olivares M, Gil Á, Gómez-Llorente C. Lactobacillus reuteri V3401 Reduces Inflammatory Biomarkers and Modifies the Gastrointestinal Microbiome in Adults with Metabolic Syndrome: The PROSIR Study. Nutrients 2019; 11:1761. [PMID: 31370223 PMCID: PMC6723328 DOI: 10.3390/nu11081761] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/16/2019] [Accepted: 07/29/2019] [Indexed: 01/14/2023] Open
Abstract
UNLABELLED Previous studies have reported that probiotics may improve clinical and inflammatory parameters in patients with obesity and metabolic syndrome (MetS). Lactobacillus (L.) reuteri V3401 has shown promising results on the components of MetS in animal studies. We aimed to evaluate the effects of L. reuteri V3401 together with healthy lifestyle recommendations on adult patients with MetS. METHODS We carried out a randomized, crossover, placebo-controlled, single-center trial in which we included 53 adult patients newly diagnosed with MetS. Patients were block randomly allocated by body mass index (BMI) and sex to receive a capsule containing either the probiotic L. reuteri V3401 (5 × 109 colony-forming units) or a placebo once daily for 12 weeks. Anthropometric variables, biochemical and inflammatory biomarkers, as well as the gastrointestinal microbiome composition were determined. RESULTS There were no differences between groups in the clinical characteristics of MetS. However, we found that interleukin-6 (IL-6) and soluble vascular cell adhesion molecule 1 (sVCAM-1) diminished by effect of the treatment with L. reuteri V3401. Analysis of the gastrointestinal microbiome revealed a rise in the proportion of Verrucomicrobia. CONCLUSIONS Consumption of L. reuteri V3401 improved selected inflammatory parameters and modified the gastrointestinal microbiome. Further studies are needed to ascertain additional beneficial effects of other probiotic strains in MetS as well as the mechanisms by which such effects are exerted.
Collapse
Affiliation(s)
- Carmen Tenorio-Jiménez
- Endocrinology and Nutrition Clinical Management Unit, University Hospital Virgen de las Nieves, 18014 Granada, Spain
| | - María José Martínez-Ramírez
- Endocrinology and Nutrition Clinical Management Unit, University Hospital of Jaén, 23007 Jaén, Spain
- Department of Health Sciences, School of Health Sciences, University of Jaén, 23071 Jaén, Spain
| | | | - Carmen Arraiza-Irigoyen
- Endocrinology and Nutrition Clinical Management Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | | | - José Camacho
- Department of Signal Theory, Networking, and Communications, University of Granada, 18071 Granada, Spain
| | - Natalia Chueca
- Department of Microbiology, University Hospital Campus de la Salud, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, 18012 Granada, Spain
| | - Federico García
- Department of Microbiology, University Hospital Campus de la Salud, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, 18012 Granada, Spain
| | - Josune Olza
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
| | - Julio Plaza-Díaz
- Instituto de Investigación Biosanitaria ibs. GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Institute of Nutrition and Food Technology "José Mataix", Center of Biomedical Research, University of Granada, 18016 Granada, Spain
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Luis Fontana
- Instituto de Investigación Biosanitaria ibs. GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Institute of Nutrition and Food Technology "José Mataix", Center of Biomedical Research, University of Granada, 18016 Granada, Spain
| | | | - Ángel Gil
- Instituto de Investigación Biosanitaria ibs. GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Institute of Nutrition and Food Technology "José Mataix", Center of Biomedical Research, University of Granada, 18016 Granada, Spain
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carolina Gómez-Llorente
- Instituto de Investigación Biosanitaria ibs. GRANADA, 18012 Granada, Spain.
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix", Center of Biomedical Research, University of Granada, 18016 Granada, Spain.
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| |
Collapse
|
28
|
Korbecki J, Bajdak-Rusinek K. The effect of palmitic acid on inflammatory response in macrophages: an overview of molecular mechanisms. Inflamm Res 2019; 68:915-932. [PMID: 31363792 PMCID: PMC6813288 DOI: 10.1007/s00011-019-01273-5] [Citation(s) in RCA: 299] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 02/06/2023] Open
Abstract
Palmitic acid is a saturated fatty acid whose blood concentration is elevated in obese patients. This causes inflammatory responses, where toll-like receptors (TLR), TLR2 and TLR4, play an important role. Nevertheless, palmitic acid is not only a TLR agonist. In the cell, this fatty acid is converted into phospholipids, diacylglycerol and ceramides. They trigger the activation of various signaling pathways that are common for LPS-mediated TLR4 activation. In particular, metabolic products of palmitic acid affect the activation of various PKCs, ER stress and cause an increase in ROS generation. Thanks to this, palmitic acid also strengthens the TLR4-induced signaling. In this review, we discuss the mechanisms of inflammatory response induced by palmitic acid. In particular, we focus on describing its effect on ER stress and IRE1α, and the mechanisms of NF-κB activation. We also present the mechanisms of inflammasome NLRP3 activation and the effect of palmitic acid on enhanced inflammatory response by increasing the expression of FABP4/aP2. Finally, we focus on the consequences of inflammatory responses, in particular, the effect of TNF-α, IL-1β and IL-6 on insulin resistance. Due to the high importance of macrophages and the production of proinflammatory cytokines by them, this work mainly focuses on these cells.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Molecular Biology, School of Medicine in Katowice, Medical University of Silesia, Medyków 18 St., 40-752, Katowice, Poland.
| | - Karolina Bajdak-Rusinek
- Department of Medical Genetics, School of Medicine in Katowice, Medical University of Silesia, Medyków 18 St., 40-752, Katowice, Poland
| |
Collapse
|
29
|
Inflammation in the hippocampus affects IGF1 receptor signaling and contributes to neurological sequelae in rheumatoid arthritis. Proc Natl Acad Sci U S A 2018; 115:E12063-E12072. [PMID: 30509997 PMCID: PMC6305002 DOI: 10.1073/pnas.1810553115] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aberrant insulin-like growth factor 1 receptor (IGF1R)/insulin receptor signaling in brain has recently been linked to neurodegeneration in diabetes mellitus and in Alzheimer’s disease. In this study, we demonstrate that functional disability and pain in patients with rheumatoid arthritis (RA) and in experimental RA are associated with hippocampal inflammation and inhibition of IGF1R/insulin receptor substrate 1 (IRS1) signal, reproducing an IGF1/insulin-resistant state. This restricts formation of new neurons in the hippocampus, reduces hippocampal volume, and predisposes RA patients to develop neurological symptoms. Improving IRS1 function through down-regulation of IGF1R disinhibits neurogenesis and can potentially ameliorate neurological symptoms. This opens perspectives for drugs that revert IGF1/insulin resistance as an essential complement to the antirheumatic and antiinflammatory arsenal. Rheumatoid arthritis (RA) is an inflammatory joint disease with a neurological component including depression, cognitive deficits, and pain, which substantially affect patients’ quality of daily life. Insulin-like growth factor 1 receptor (IGF1R) signaling is one of the factors in RA pathogenesis as well as a known regulator of adult neurogenesis. The purpose of this study was to investigate the association between IGF1R signaling and the neurological symptoms in RA. In experimental RA, we demonstrated that arthritis induced enrichment of IBA1+ microglia in the hippocampus. This coincided with inhibitory phosphorylation of insulin receptor substrate 1 (IRS1) and up-regulation of IGF1R in the pyramidal cell layer of the cornus ammoni and in the dentate gyrus, reproducing the molecular features of the IGF1/insulin resistance. The aberrant IGF1R signaling was associated with reduced hippocampal neurogenesis, smaller hippocampus, and increased immobility of RA mice. Inhibition of IGF1R in experimental RA led to a reduction of IRS1 inhibition and partial improvement of neurogenesis. Evaluation of physical functioning and brain imaging in RA patients revealed that enhanced functional disability is linked with smaller hippocampus volume and aberrant IGF1R/IRS1 signaling. These results point to abnormal IGF1R signaling in the brain as a mediator of neurological sequelae in RA and provide support for the potentially reversible nature of hippocampal changes.
Collapse
|
30
|
Emerging Role of Adipocytokines in Type 2 Diabetes as Mediators of Insulin Resistance and Cardiovascular Disease. Can J Diabetes 2018; 42:446-456.e1. [PMID: 29229313 DOI: 10.1016/j.jcjd.2017.10.040] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 10/06/2017] [Accepted: 10/06/2017] [Indexed: 12/13/2022]
|
31
|
Phillips C, Fahimi A. Immune and Neuroprotective Effects of Physical Activity on the Brain in Depression. Front Neurosci 2018; 12:498. [PMID: 30093853 PMCID: PMC6070639 DOI: 10.3389/fnins.2018.00498] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/03/2018] [Indexed: 12/13/2022] Open
Abstract
Physical activity-a lifestyle factor that is associated with immune function, neuroprotection, and energy metabolism-modulates the cellular and molecular processes in the brain that are vital for emotional and cognitive health, collective mechanisms that can go awry in depression. Physical activity optimizes the stress response, neurotransmitter level and function (e.g., serotonergic, noradrenergic, dopaminergic, and glutamatergic), myokine production (e.g., interleukin-6), transcription factor levels and correlates [e.g., peroxisome proliferator-activated receptor C coactivator-1α [PGC-1α], mitochondrial density, nitric oxide pathway activity, Ca2+ signaling, reactive oxygen specie production, and AMP-activated protein kinase [AMPK] activity], kynurenine metabolites, glucose regulation, astrocytic health, and growth factors (e.g., brain-derived neurotrophic factor). Dysregulation of these interrelated processes can effectuate depression, a chronic mental illness that affects millions of individuals worldwide. Although the biogenic amine model has provided some clinical utility in understanding chronic depression, a need remains to better understand the interrelated mechanisms that contribute to immune dysfunction and the means by which various therapeutics mitigate them. Fortunately, convergent evidence suggests that physical activity improves emotional and cognitive function in persons with depression, particularly in those with comorbid inflammation. Accordingly, the aims of this review are to (1) underscore the link between inflammatory correlates and depression, (2) explicate immuno-neuroendocrine foundations, (3) elucidate evidence of neurotransmitter and cytokine crosstalk in depressive pathobiology, (4) determine the immunomodulatory effects of physical activity in depression, (5) examine protocols used to effectuate the positive effects of physical activity in depression, and (6) highlight implications for clinicians and scientists. It is our contention that a deeper understanding of the mechanisms by which inflammation contributes to the pathobiology of depression will translate to novel and more effective treatments, particularly by identifying relevant patient populations that can benefit from immune-based therapies within the context of personalized medicine.
Collapse
Affiliation(s)
- Cristy Phillips
- Physical Therapy, Arkansas State University, Jonesboro, AR, United States
- Physical Therapy, University of Tennessee Health Science Center, Memphis, TN, United States
| | | |
Collapse
|
32
|
IL-6 signalling pathways and the development of type 2 diabetes. Inflammopharmacology 2018; 26:685-698. [PMID: 29508109 DOI: 10.1007/s10787-018-0458-0] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 02/21/2018] [Indexed: 02/07/2023]
Abstract
Interleukin 6 (IL-6), a multifunctional cytokine, has been implicated in the pathophysiology of type 2 diabetes (T2D). The elevated circulating level of IL-6 is an independent predictor of T2D and is considered to be involved in the development of inflammation, insulin resistance and β-cell dysfunction. On the other hand, an increasing number of evidence suggests that IL-6 has an anti-inflammatory role and improves glucose metabolism. The complex signal transduction mechanism of IL-6 may help explain the pleiotropic nature of the cytokine. IL-6 acts via two distinct signalling pathways called classic signalling and trans-signalling. While both signalling modes lead to activation of the same receptor subunit, their final biological effects are completely different. The aim of this review is to summarize our current knowledge about the role of IL-6 in the development of T2D. We will also discuss the importance of specific blockade of IL-6 trans-signalling rather than inhibiting both signalling pathways as a therapeutic strategy for the treatment of T2D and its associated macrovascular complications.
Collapse
|
33
|
Hoffmann C, Weigert C. Skeletal Muscle as an Endocrine Organ: The Role of Myokines in Exercise Adaptations. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a029793. [PMID: 28389517 DOI: 10.1101/cshperspect.a029793] [Citation(s) in RCA: 224] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Exercise stimulates the release of proteins with autocrine, paracrine, or endocrine functions produced in skeletal muscle, termed myokines. Based on the current state of knowledge, the major physiological function of myokines is to protect the functionality and to enhance the exercise capacity of skeletal muscle. Myokines control adaptive processes in skeletal muscle by acting as paracrine regulators of fuel oxidation, hypertrophy, angiogenesis, inflammatory processes, and regulation of the extracellular matrix. Endocrine functions attributed to myokines are involved in body weight regulation, low-grade inflammation, insulin sensitivity, suppression of tumor growth, and improvement of cognitive function. Muscle-derived regulatory RNAs and metabolites, as well as the design of modified myokines, are promising novel directions for treatment of chronic diseases.
Collapse
Affiliation(s)
- Christoph Hoffmann
- Division of Pathobiochemistry and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Cora Weigert
- Division of Pathobiochemistry and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, 72076 Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen, 72076 Tübingen, Germany.,German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
| |
Collapse
|
34
|
Gao S, Durstine JL, Koh HJ, Carver WE, Frizzell N, Carson JA. Acute myotube protein synthesis regulation by IL-6-related cytokines. Am J Physiol Cell Physiol 2017; 313:C487-C500. [PMID: 28768641 DOI: 10.1152/ajpcell.00112.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/20/2017] [Accepted: 07/25/2017] [Indexed: 12/19/2022]
Abstract
IL-6 and leukemia inhibitory factor (LIF), members of the IL-6 family of cytokines, play recognized paradoxical roles in skeletal muscle mass regulation, being associated with both growth and atrophy. Overload or muscle contractions can induce a transient increase in muscle IL-6 and LIF expression, which has a regulatory role in muscle hypertrophy. However, the cellular mechanisms involved in this regulation have not been completely identified. The induction of mammalian target of rapamycin complex 1 (mTORC1)-dependent myofiber protein synthesis is an established regulator of muscle hypertrophy, but the involvement of the IL-6 family of cytokines in this process is poorly understood. Therefore, we investigated the acute effects of IL-6 and LIF administration on mTORC1 signaling and protein synthesis in C2C12 myotubes. The role of glycoprotein 130 (gp130) receptor and downstream signaling pathways, including phosphoinositide 3-kinase (PI3K)-Akt-mTORC1 and signal transducer and activator of transcription 3 (STAT3)-suppressor of cytokine signaling 3 (SOCS3), was investigated by administration of specific siRNA or pharmaceutical inhibitors. Acute administration of IL-6 and LIF induced protein synthesis, which was accompanied by STAT3 activation, Akt-mTORC1 activation, and increased SOCS3 expression. This induction of protein synthesis was blocked by both gp130 siRNA knockdown and Akt inhibition. Interestingly, STAT3 inhibition or Akt downstream mTORC1 signaling inhibition did not fully block the IL-6 or LIF induction of protein synthesis. SOCS3 siRNA knockdown increased basal protein synthesis and extended the duration of the protein synthesis induction by IL-6 and LIF. These results demonstrate that either IL-6 or LIF can activate gp130-Akt signaling axis, which induces protein synthesis via mTORC1-independent mechanisms in cultured myotubes. However, IL-6- or LIF-induced SOCS3 negatively regulates the activation of myotube protein synthesis.
Collapse
Affiliation(s)
- Song Gao
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina
| | - J Larry Durstine
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina
| | - Ho-Jin Koh
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina
| | - Wayne E Carver
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina
| | - Norma Frizzell
- Department of Pharmacology, Physiology, and Neuronscience, School of Medicine, University of South Carolina, Columbia, South Carolina; and
| | - James A Carson
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina; .,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
35
|
Feng XT, Tang SY, Jiang YX, Zhao W. ANTI-DIABETIC EFFECTS OF ZHUODUQING FORMULA, A CHINESE HERBAL DECOCTION, ON A RAT MODEL OF TYPE 2 DIABETES. AFRICAN JOURNAL OF TRADITIONAL, COMPLEMENTARY, AND ALTERNATIVE MEDICINES 2017; 14:42-50. [PMID: 28480415 PMCID: PMC5412237 DOI: 10.21010/ajtcam.v14i3.5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background: Zhuoduqing formula (ZDQ) is a Chinese herbal decoction and used to treat type 2 diabetes in clinical practice, but the potential evidence needs to be provided. Materials and Methods: Type 2 diabetic model rats were induced by feeding high fat diet (HFD) and intraperitoneal injection of streptozotocin (STZ). The model rats were given ZDQ for 4 weeks. Insulin sensitivity was evaluated by homeostasis model assessment of basal insulin resistance (HOMA-IR) and intraperitoneal glucose tolerance test (IPGTT). Blood insulin and tumour necrosis factor-α (TNF-α) levels as well as SOCS-3 levels in skeletal muscles were analyzed by ELISA. Results: ZDQ significantly decreased fasting blood glucose, ameliorated HOMA-IR and IPGTT, and reduced triglyceride and total cholesterol in type 2 diabetic rats. Moreover, ZDQ remarkably lowered blood TNF-α levels and inhibited SOCS-3 levels in skeletal muscles. Conclusion: The results display that ZDQ performs anti-diabetic functions in type 2 diabetic rats induced by feeding HFD and intraperitoneal injection of STZ. Abbreviations: ZDQ, zhuoduqing formula; ROS, rosiglitazone; HOMA-IR, homeostasis model assessment of basal insulin resistance; IPGTT, intraperitoneal glucose tolerance test; HFD, high fat diet; SOCS-3, suppressor of cytokine signaling-3; TNF-α, tumour necrosis factor-α.
Collapse
Affiliation(s)
- Xiao-Tao Feng
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530200, P.R. China.,Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, Nanning 530200, P.R. China
| | - Shu-Yan Tang
- Graduate school, Guangxi University of Chinese Medicine, Nanning 530200, P.R. China
| | - Yun-Xia Jiang
- Graduate school, Guangxi University of Chinese Medicine, Nanning 530200, P.R. China
| | - Wei Zhao
- Department of Endocrinology, The Second Affiliated Hospital, Guiyang University of Chinese Medicine, Guiyang 550003, P.R. China
| |
Collapse
|
36
|
Juretić N, Díaz J, Romero F, González G, Jaimovich E, Riveros N. Interleukin-6 and neuregulin-1 as regulators of utrophin expression via the activation of NRG-1/ErbB signaling pathway in mdx cells. Biochim Biophys Acta Mol Basis Dis 2017; 1863:770-780. [DOI: 10.1016/j.bbadis.2016.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/10/2016] [Accepted: 12/12/2016] [Indexed: 01/16/2023]
|
37
|
Wang J, Yang DL, Chen ZZ, Gou BF. Associations of body mass index with cancer incidence among populations, genders, and menopausal status: A systematic review and meta-analysis. Cancer Epidemiol 2016; 42:1-8. [DOI: 10.1016/j.canep.2016.02.010] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 02/22/2016] [Accepted: 02/25/2016] [Indexed: 02/06/2023]
|
38
|
Belizário JE, Fontes-Oliveira CC, Borges JP, Kashiabara JA, Vannier E. Skeletal muscle wasting and renewal: a pivotal role of myokine IL-6. SPRINGERPLUS 2016; 5:619. [PMID: 27330885 PMCID: PMC4870483 DOI: 10.1186/s40064-016-2197-2] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/20/2016] [Indexed: 12/20/2022]
Abstract
Adult skeletal tissue is composed of heterogeneous population of cells that constantly self-renew by means of a controlled process of activation and proliferation of tissue-resident stem cells named satellite cells. Many growth factors, cytokines and myokines produced by skeletal muscle cells play critical roles in local regulation of the inflammatory process and skeletal muscle regeneration during different pathological conditions. IL-6 is a pleiotropic cytokine released in large amount during infection, autoimmunity and cancer. Low levels of IL-6 can promote activation of satellite cells and myotube regeneration while chronically elevated production promote skeletal muscle wasting. These distinct effects may be explained by a crosstalk of the IL-6/IL-6 receptor and gp130 trans-signaling pathway that oppose to regenerative and anti-inflammatory of the classical IL-6 receptor signaling pathway. Here we discuss on potential therapeutic strategies using monoclonal antibodies to IL-6R for the treatment of skeletal muscle wasting and cachexia. We also highlight on the IL-6/JAK/STAT and FGF/p38αβ MAPK signaling pathways in satellite cell activation and the use of protein kinase inhibitors for tailoring and optimizing satellite cell proliferation during the skeletal muscle renewal. Future investigations on the roles of the IL-6 classical and trans-signaling pathways in both immune and non-immune cells in skeletal muscle tissue will provide new basis for therapeutic approaches to reverse atrophy and degeneration of skeletal muscles in cancer and inflammatory diseases.
Collapse
Affiliation(s)
- José E Belizário
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Avenida Lineu Prestes, 1524, São Paulo, SP 05508-900 Brazil
| | | | - Janaina Padua Borges
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Avenida Lineu Prestes, 1524, São Paulo, SP 05508-900 Brazil
| | - Janete Akemi Kashiabara
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Avenida Lineu Prestes, 1524, São Paulo, SP 05508-900 Brazil
| | - Edouard Vannier
- Division of Geographic Medicine and Infectious Disease, Tufts Medical Center, Boston, MA 02111 USA
| |
Collapse
|
39
|
Zietek T, Rath E. Inflammation Meets Metabolic Disease: Gut Feeling Mediated by GLP-1. Front Immunol 2016; 7:154. [PMID: 27148273 PMCID: PMC4840214 DOI: 10.3389/fimmu.2016.00154] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/08/2016] [Indexed: 12/14/2022] Open
Abstract
Chronic diseases, such as obesity and diabetes, cardiovascular, and inflammatory bowel diseases (IBD) share common features in their pathology. Metabolic disorders exhibit strong inflammatory underpinnings and vice versa, inflammation is associated with metabolic alterations. Next to cytokines and cellular stress pathways, such as the unfolded protein response (UPR), alterations in the enteroendocrine system are intersections of various pathologies. Enteroendocrine cells (EEC) have been studied extensively for their ability to regulate gastrointestinal motility, secretion, and insulin release by release of peptide hormones. In particular, the L-cell-derived incretin hormone glucagon-like peptide 1 (GLP-1) has gained enormous attention due to its insulinotropic action and relevance in the treatment of type 2 diabetes (T2D). Yet, accumulating data indicate a critical role for EEC and in particular for GLP-1 in metabolic adaptation and in orchestrating immune responses beyond blood glucose control. EEC sense the lamina propria and luminal environment, including the microbiota via receptors and transporters. Subsequently, mediating signals by secreting hormones and cytokines, EEC can be considered as integrators of metabolic and inflammatory signaling. This review focuses on L cell and GLP-1 functions in the context of metabolic and inflammatory diseases. The effects of incretin-based therapies on metabolism and immune system are discussed and the interrelation and common features of metabolic and immune-mediated disorders are highlighted. Moreover, it presents data on the impact of inflammation, in particular of IBD on EEC and discusses the potential role of the microbiota as link between nutrients, metabolism, immunity, and disease.
Collapse
Affiliation(s)
- Tamara Zietek
- Department of Nutritional Physiology, Technische Universität München , Freising , Germany
| | - Eva Rath
- Chair of Nutrition and Immunology, Technische Universität München , Freising , Germany
| |
Collapse
|
40
|
Gerst F, Kaiser G, Panse M, Sartorius T, Pujol A, Hennige AM, Machicao F, Lammers R, Bosch F, Häring HU, Ullrich S. Protein kinase Cδ regulates nuclear export of FOXO1 through phosphorylation of the chaperone 14-3-3ζ. Diabetologia 2015; 58:2819-31. [PMID: 26363783 DOI: 10.1007/s00125-015-3744-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/03/2015] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS Forkhead box protein O1 (FOXO1) is a transcription factor essential for beta cell fate. Protein kinase B-dependent phosphorylation of FOXO1 at S256 (P-FOXO1) enables its binding to 14-3-3 dimers and nuclear export. Dephosphorylated FOXO1 enters nuclei and activates pro-apoptotic genes. Since our previous observations suggest that protein kinase C delta (PKCδ) induces nuclear accumulation of FOXO1, the underlying mechanism was examined. METHODS In human islets, genetically modified mice and INS-1E cells apoptosis was assessed by TUNEL staining. Subcellular translocation of proteins was examined by confocal microscopy and signalling pathways were analysed by western blotting and overlay assay. RESULTS In PKCδ-overexpressing (PKCδ-tg) mouse islet cells and INS-1E cells FOXO1 accumulated in nuclei, surprisingly, as P-FOXO1. PKCδ-tg decelerated IGF-1-dependent stimulation of nuclear export, indicating that changes in export caused nuclear retention of P-FOXO1. Nuclear accumulation of P-FOXO1 was accompanied by increased phosphorylation of 14-3-3ζ at S58 and reduced dimerisation of 14-3-3ζ. Palmitic acid further augmented phosphorylation of 14-3-3ζ and triggered nuclear accumulation of FOXO1 in both INS-1E and human islet cells. Furthermore, the overexpression of a phosphomimicking mutant of 14-3-3ζ (S58D) enhanced nuclear FOXO1. In accordance with the nuclear accumulation of P-FOXO1, PKCδ overexpression alone did not increase apoptotic cell death. Additionally, insulin secretion and glucose homeostasis in PKCδ-overexpressing mice remained unaffected. CONCLUSIONS/INTERPRETATION These results suggest that PKCδ-mediated phosphorylation of 14-3-3ζ contributes to the nuclear retention of FOXO1, even when FOXO1 is phosphorylated as under non-stress conditions. P-FOXO1 does not induce pro-apoptotic genes, but may rather exert beneficial effects on beta cells.
Collapse
Affiliation(s)
- Felicia Gerst
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Gabriele Kaiser
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Madhura Panse
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
| | - Tina Sartorius
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Anna Pujol
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Anita M Hennige
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
| | - Fausto Machicao
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Reiner Lammers
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Hans-Ulrich Häring
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Susanne Ullrich
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany.
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany.
| |
Collapse
|
41
|
Perrin L, Loizides-Mangold U, Skarupelova S, Pulimeno P, Chanon S, Robert M, Bouzakri K, Modoux C, Roux-Lombard P, Vidal H, Lefai E, Dibner C. Human skeletal myotubes display a cell-autonomous circadian clock implicated in basal myokine secretion. Mol Metab 2015; 4:834-45. [PMID: 26629407 PMCID: PMC4632112 DOI: 10.1016/j.molmet.2015.07.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 07/27/2015] [Accepted: 07/30/2015] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE Circadian clocks are functional in all light-sensitive organisms, allowing an adaptation to the external world in anticipation of daily environmental changes. In view of the potential role of the skeletal muscle clock in the regulation of glucose metabolism, we aimed to characterize circadian rhythms in primary human skeletal myotubes and investigate their roles in myokine secretion. METHODS We established a system for long-term bioluminescence recording in differentiated human myotubes, employing lentivector gene delivery of the Bmal1-luciferase and Per2-luciferase core clock reporters. Furthermore, we disrupted the circadian clock in skeletal muscle cells by transfecting siRNA targeting CLOCK. Next, we assessed the basal secretion of a large panel of myokines in a circadian manner in the presence or absence of a functional clock. RESULTS Bioluminescence reporter assays revealed that human skeletal myotubes, synchronized in vitro, exhibit a self-sustained circadian rhythm, which was further confirmed by endogenous core clock transcript expression. Moreover, we demonstrate that the basal secretion of IL-6, IL-8 and MCP-1 by synchronized skeletal myotubes has a circadian profile. Importantly, the secretion of IL-6 and several additional myokines was strongly downregulated upon siClock-mediated clock disruption. CONCLUSIONS Our study provides for the first time evidence that primary human skeletal myotubes possess a high-amplitude cell-autonomous circadian clock, which could be attenuated. Furthermore, this oscillator plays an important role in the regulation of basal myokine secretion by skeletal myotubes.
Collapse
Affiliation(s)
- Laurent Perrin
- Division of Endocrinology, Diabetes and Nutrition, Department of Clinical Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ursula Loizides-Mangold
- Division of Endocrinology, Diabetes and Nutrition, Department of Clinical Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Svetlana Skarupelova
- Division of Endocrinology, Diabetes and Nutrition, Department of Clinical Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Pamela Pulimeno
- Division of Endocrinology, Diabetes and Nutrition, Department of Clinical Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Stephanie Chanon
- CarMeN Laboratory, INSERM U1060, INRA 1397, University Lyon 1, Oullins, France
| | - Maud Robert
- Department of Digestive and Bariatric Surgery, Edouard Herriot Hospital, Lyon, France
| | - Karim Bouzakri
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Christine Modoux
- Division of Immunology and Allergy, Department of Medical Specialties, University Hospital and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Pascale Roux-Lombard
- Division of Immunology and Allergy, Department of Medical Specialties, University Hospital and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Hubert Vidal
- CarMeN Laboratory, INSERM U1060, INRA 1397, University Lyon 1, Oullins, France
| | - Etienne Lefai
- CarMeN Laboratory, INSERM U1060, INRA 1397, University Lyon 1, Oullins, France
| | - Charna Dibner
- Division of Endocrinology, Diabetes and Nutrition, Department of Clinical Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
42
|
Park SE, Park CY, Sweeney G. Biomarkers of insulin sensitivity and insulin resistance: Past, present and future. Crit Rev Clin Lab Sci 2015; 52:180-90. [PMID: 26042993 DOI: 10.3109/10408363.2015.1023429] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Insulin resistance in insulin target tissues including liver, skeletal muscle and adipose tissue is an early step in the progression towards type 2 diabetes. Accurate diagnostic parameters reflective of insulin resistance are essential. Longstanding tests for fasting blood glucose and HbA1c are useful and although the hyperinsulinemic euglycemic clamp remains a "gold standard" for accurately determining insulin resistance, it cannot be implemented on a routine basis. The study of adipokines, and more recently myokines and hepatokines, as potential biomarkers for insulin sensitivity is now an attractive and relatively straightforward approach. This review discusses potential biomarkers including adiponectin, RBP4, chemerin, A-FABP, FGF21, fetuin-A, myostatin, IL-6, and irisin, all of which may play significant roles in determining insulin sensitivity. We also review potential future directions of new biological markers for measuring insulin resistance, including metabolomics and gut microbiome. Collectively, these approaches will provide clinicians with the tools for more accurate, and perhaps personalized, diagnosis of insulin resistance.
Collapse
Affiliation(s)
- Se Eun Park
- a Division of Endocrinology and Metabolism, Department of Internal Medicine , Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine , Seoul , South Korea and
| | | | | |
Collapse
|
43
|
Ahmadi-Abhari S, Kaptoge S, Luben RN, Wareham NJ, Khaw KT. Longitudinal association of C-reactive protein and Haemoglobin A1c over 13 years: the European Prospective Investigation into Cancer--Norfolk study. Cardiovasc Diabetol 2015; 14:61. [PMID: 25994228 PMCID: PMC4445808 DOI: 10.1186/s12933-015-0224-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 05/06/2015] [Indexed: 01/21/2023] Open
Abstract
Background Type-2 diabetes is associated with systemic inflammation and higher C-reactive protein (CRP) levels. However, the longitudinal association of CRP and haemoglobin-A1c (HbA1c) has not been described in large prospective studies. Understanding such associations may shed light on the role of inflammation in development of type-2 diabetes and its complications such as cardiovascular diseases. Methods EPIC-Norfolk is a cohort study of men and women aged 40–79 years at time of recruitment (1993–1997). Serum CRP (mg/l) was measured using a high-sensitivity assay at baseline and 13-years follow-up. HbA1c (%) was measured at baseline, 4, and 13 years. Participants were excluded if they were diagnosed with diabetes or were taking diabetes medication. Data on at least one measurement of CRP and HbA1c was available for 14228 participants (55 % of the cohort). Results In the cross-sectional analysis of baseline data, a 1-SD higher loge-CRP (about three-fold higher CRP) was associated with 0.06 (95 % CI 0.04, 0.08) higher HbA1c (%) adjusted for potential confounders. In longitudinal analysis using multivariable linear mixed models, change in CRP over 13 years was to a similar extent positively associated with increase in HbA1c, such that 1-SD higher longitudinal change in loge-CRP was associated with 0.04 (95 % CI 0.02, 0.05) increase in HbA1c. Conclusion In this study we found longitudinal observational evidence suggesting that increase in systemic inflammation is associated with an increase in HbA1c and thus systemic inflammation may have a role in development of type-2 diabetes and its complications.
Collapse
Affiliation(s)
- Sara Ahmadi-Abhari
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK. .,Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge, CB1 8RN, UK.
| | - Stephen Kaptoge
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| | - Robert N Luben
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| | - Nicholas J Wareham
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Sciences, Cambridge, UK.
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| |
Collapse
|
44
|
Buzzetti R, Spoletini M, Zampetti S, Campagna G, Marandola L, Panimolle F, Dotta F, Tiberti C. Tyrosine phosphatase-related islet antigen 2(256-760) autoantibodies, the only marker of islet autoimmunity that increases by increasing the degree of BMI in obese subjects with type 2 diabetes. Diabetes Care 2015; 38:513-20. [PMID: 25567348 DOI: 10.2337/dc14-1638] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Since patients with type 2 diabetes and positive for type 1 diabetes-specific antibodies have wide variations in BMI, this study evaluated whether the frequency and pattern of islet autoantibody positivity is related to BMI. RESEARCH DESIGN AND METHODS Clinical and biochemical characteristics and islet autoantibodies including GAD and protein tyrosine phosphatases islet antigen-2 (IA-2)IC and IA-2(256-760) were evaluated in 1,850 patients with type 2 diabetes from the Non-Insulin Requiring Autoimmune Diabetes study cohort. BMI was evaluated in all patients, who were then subdivided in three groups according to BMI (<25, ≥25 to <30, and ≥30 kg/m(2)). RESULTS Out of 1,850, 120 (6.5%) patients were positive for at least one of the following antibodies: GAD (4.1%), IA-2(256-760) (3.3%), or IA-2IC (1.1%). GAD and IA-2IC antibodies showed decreasing frequencies with increasing BMI (P < 0.0001 and 0.0006, respectively, for trend); in contrast, the frequency of IA-2(256-760) antibodies increased with increasing BMI (P = 0.005 for trend). Patients with type 2 diabetes positive for IA-2(256-760) alone showed a phenotype resembling classical obese type 2 diabetes, with higher BMI, waist circumference, and uric acid (P < 0.005 for all), lower thyroid peroxidase antibodies, and lower progression to insulin requirement than GAD antibody-positive patients (P = 0.04 and P = 0.0005, respectively). CONCLUSIONS The IA-2(256-760) antibody appears to represent an antibody marker that mainly identifies a clinical phenotype very similar to obese type 2 diabetes, suggesting a possible different pathogenetic mechanism.
Collapse
Affiliation(s)
- Raffaella Buzzetti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Simona Zampetti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Campagna
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Lidia Marandola
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesca Panimolle
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Claudio Tiberti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
45
|
Marra AM, Arcopinto M, Bossone E, Ehlken N, Cittadini A, Grünig E. Pulmonary arterial hypertension-related myopathy: an overview of current data and future perspectives. Nutr Metab Cardiovasc Dis 2015; 25:131-139. [PMID: 25455722 DOI: 10.1016/j.numecd.2014.10.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 09/18/2014] [Accepted: 10/13/2014] [Indexed: 01/23/2023]
Abstract
BACKGROUND AND AIM Exercise intolerance is one of the key features of pulmonary arterial hypertension (PAH). The main determinants of exercise impairment include hypoxemia, reduced right ventricular output, perfusion/ventilation mismatch, and weakness of skeletal and breathing muscles. The aim of the current review is to describe the findings in the existing literature about respiratory and muscle dysfunction in PAH. Animal and clinical studies regarding both respiratory and peripheral skeletal muscles and the effect of exercise training on muscle function in PAH patients are analyzed. DATA SYNTHESIS PAH myopathy is characterized by reduced skeletal muscle mass, reduced volitional and non-volitional contractility, reduced generated force, a fiber switch from type I to type II, increased protein degradation through ubiquitin-proteasome system (UPS) activation, reduced mitochondrial functioning, and impaired activation-contractility coupling. Increased inflammatory response, impaired anabolic signaling, hypoxemia, and abnormalities of mitochondrial function are involved in the pathophysiology of this process. Exercise training has been shown to improve exercise capacity, peak oxygen uptake, quality of life, and possibly clinical outcomes of PAH patients. CONCLUSIONS The skeletal muscles of PAH patients show a wide spectrum of cellular abnormalities that finally culminate in muscle atrophy and reduced contractility. Exercise training improves muscle function and bears a positive impact on the clinical outcomes of PAH patients.
Collapse
Affiliation(s)
- A M Marra
- Pulmonary Hypertension Unit, Thoraxclinic, University Hospital Heidelberg, Heidelberg, Germany; Department of Translational Medical Sciences, "Federico II" University School of Medicine, Naples, Italy
| | - M Arcopinto
- Department of Cardiac Surgery, IRCSS Policlinico San Donato, Milan, Italy
| | - E Bossone
- Department of Cardiology and Cardiac Surgery, University Hospital "Scuola Medica Salernitana", Salerno, Italy
| | - N Ehlken
- Pulmonary Hypertension Unit, Thoraxclinic, University Hospital Heidelberg, Heidelberg, Germany
| | - A Cittadini
- Department of Translational Medical Sciences, "Federico II" University School of Medicine, Naples, Italy; Interdisciplinary Research Centre in Biomedical Materials (CRIB), Federico II University, Naples, Italy.
| | - E Grünig
- Pulmonary Hypertension Unit, Thoraxclinic, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
46
|
IL-6 and Akt are involved in muscular pathogenesis in myasthenia gravis. Acta Neuropathol Commun 2015; 3:1. [PMID: 25627031 PMCID: PMC4308930 DOI: 10.1186/s40478-014-0179-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 12/15/2014] [Indexed: 01/11/2023] Open
Abstract
Introduction Anti-acetylcholine receptor (AChR) autoantibodies target muscles in spontaneous human myasthenia gravis (MG) and its induced experimental autoimmune model MG (EAMG). The aim of this study was to identify novel functional mechanisms occurring in the muscle pathology of myasthenia. Results A transcriptome analysis performed on muscle tissue from MG patients (compared with healthy controls) and from EAMG rats (compared with control rats) revealed a deregulation of genes associated with the Interleukin-6 (IL-6) and Insulin-Like Growth Factor 1 (IGF-1) pathways in both humans and rats. The expression of IL-6 and its receptor IL-6R transcripts was found to be altered in muscles of EAMG rats and mice compared with control animals. In muscle biopsies from MG patients, IL-6 protein level was higher than in control muscles. Using cultures of human muscle cells, we evaluated the effects of anti-AChR antibodies on IL-6 production and on the phosphorylation of Protein Kinase B (PKB/Akt). Most MG sera and some monoclonal anti-AChR antibodies induced a significant increase in IL-6 production by human muscle cells. Furthermore, Akt phosphorylation in response to insulin was decreased in the presence of monoclonal anti-AChR antibodies. Conclusions Anti-AChR antibodies alter IL-6 production by muscle cells, suggesting a putative novel functional mechanism of action for the anti-AChR antibodies. IL-6 is a myokine with known effects on signaling pathways such as Akt/mTOR (mammalian Target of Rapamycin). Since Akt plays a key role in multiple cellular processes, the reduced phosphorylation of Akt by the anti-AChR antibodies may have a significant impact on the muscle fatigability observed in MG patients. Electronic supplementary material The online version of this article (doi:10.1186/s40478-014-0179-6) contains supplementary material, which is available to authorized users.
Collapse
|
47
|
Blesson CS, Sathishkumar K, Chinnathambi V, Yallampalli C. Gestational protein restriction impairs insulin-regulated glucose transport mechanisms in gastrocnemius muscles of adult male offspring. Endocrinology 2014; 155:3036-46. [PMID: 24797633 PMCID: PMC4098002 DOI: 10.1210/en.2014-1094] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Type II diabetes originates from various genetic and environmental factors. Recent studies showed that an adverse uterine environment such as that caused by a gestational low-protein (LP) diet can cause insulin resistance in adult offspring. The mechanism of insulin resistance induced by gestational protein restriction is not clearly understood. Our aim was to investigate the role of insulin signaling molecules in gastrocnemius muscles of gestational LP diet-exposed male offspring to understand their role in LP-induced insulin resistance. Pregnant Wistar rats were fed a control (20% protein) or isocaloric LP (6%) diet from gestational day 4 until delivery and a normal diet after weaning. Only male offspring were used in this study. Glucose and insulin responses were assessed after a glucose tolerance test. mRNA and protein levels of molecules involved in insulin signaling were assessed at 4 months in gastrocnemius muscles. Muscles were incubated ex vivo with insulin to evaluate insulin-induced phosphorylation of insulin receptor (IR), Insulin receptor substrate-1, Akt, and AS160. LP diet-fed rats gained less weight than controls during pregnancy. Male pups from LP diet-fed mothers were smaller but exhibited catch-up growth. Plasma glucose and insulin levels were elevated in LP offspring when subjected to a glucose tolerance test; however, fasting levels were comparable. LP offspring showed increased expression of IR and AS160 in gastrocnemius muscles. Ex vivo treatment of muscles with insulin showed increased phosphorylation of IR (Tyr972) in controls, but LP rats showed higher basal phosphorylation. Phosphorylation of Insulin receptor substrate-1 (Tyr608, Tyr895, Ser307, and Ser318) and AS160 (Thr642) were defective in LP offspring. Further, glucose transporter type 4 translocation in LP offspring was also impaired. A gestational LP diet leads to insulin resistance in adult offspring by a mechanism involving inefficient insulin-induced IR, Insulin receptor substrate-1, and AS160 phosphorylation and impaired glucose transporter type 4 translocation.
Collapse
Affiliation(s)
- Chellakkan S Blesson
- Department of Obstetrics and Gynecology (C.S.B., C.Y.), Baylor College of Medicine, Houston, Texas 77030; and Division of Reproductive Endocrinology (K.S., V.C.), Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas 77555
| | | | | | | |
Collapse
|
48
|
Immune modulating capability of two exopolysaccharide-producing Bifidobacterium strains in a Wistar rat model. BIOMED RESEARCH INTERNATIONAL 2014; 2014:106290. [PMID: 24971309 PMCID: PMC4058098 DOI: 10.1155/2014/106290] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 04/28/2014] [Indexed: 12/14/2022]
Abstract
Fermented dairy products are the usual carriers for the delivery of probiotics to humans, Bifidobacterium and Lactobacillus being the most frequently used bacteria. In this work, the strains Bifidobacterium animalis subsp. lactis IPLA R1 and Bifidobacterium longum IPLA E44 were tested for their capability to modulate immune response and the insulin-dependent glucose homeostasis using male Wistar rats fed with a standard diet. Three intervention groups were fed daily for 24 days with 10% skimmed milk, or with 109 cfu of the corresponding strain suspended in the same vehicle. A significant increase of the suppressor-regulatory TGF-β cytokine occurred with both strains in comparison with a control (no intervention) group of rats; the highest levels were reached in rats fed IPLA R1. This strain presented an immune protective profile, as it was able to reduce the production of the proinflammatory IL-6. Moreover, phosphorylated Akt kinase decreased in gastroctemius muscle of rats fed the strain IPLA R1, without affecting the glucose, insulin, and HOMA index in blood, or levels of Glut-4 located in the membrane of muscle and adipose tissue cells. Therefore, the strain B. animalis subsp. lactis IPLA R1 is a probiotic candidate to be tested in mild grade inflammation animal models.
Collapse
|
49
|
Bustamante M, Fernández-Verdejo R, Jaimovich E, Buvinic S. Electrical stimulation induces IL-6 in skeletal muscle through extracellular ATP by activating Ca(2+) signals and an IL-6 autocrine loop. Am J Physiol Endocrinol Metab 2014; 306:E869-82. [PMID: 24518675 PMCID: PMC3989743 DOI: 10.1152/ajpendo.00450.2013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Interleukin-6 (IL-6) is an important myokine that is highly expressed in skeletal muscle cells upon exercise. We assessed IL-6 expression in response to electrical stimulation (ES) or extracellular ATP as a known mediator of the excitation-transcription mechanism in skeletal muscle. We examined whether the canonical signaling cascade downstream of IL-6 (IL-6/JAK2/STAT3) also responds to muscle cell excitation, concluding that IL-6 influences its own expression through a positive loop. Either ES or exogenous ATP (100 μM) increased both IL-6 expression and p-STAT3 levels in rat myotubes, a process inhibited by 100 μM suramin and 2 U/ml apyrase. ATP also evoked IL-6 expression in both isolated skeletal fibers and extracts derived from whole FDB muscles. ATP increased IL-6 release up to 10-fold. STAT3 activation evoked by ATP was abolished by the JAK2 inhibitor HBC. Blockade of secreted IL-6 with a neutralizing antibody or preincubation with the STAT3 inhibitor VIII reduced STAT3 activation evoked by extracellular ATP by 70%. Inhibitor VIII also reduced by 70% IL-6 expression evoked by ATP, suggesting a positive IL-6 loop. In addition, ATP increased up to 60% the protein levels of SOCS3, a negative regulator of the IL-6 signaling pathway. On the other hand, intracellular calcium chelation or blockade of IP3-dependent calcium signals abolished STAT3 phosphorylation evoked by either extracellular ATP or ES. These results suggest that expression of IL-6 in stimulated skeletal muscle cells is mediated by extracellular ATP and nucleotide receptors, involving IP3-dependent calcium signals as an early step that triggers a positive IL-6 autocrine loop.
Collapse
Affiliation(s)
- Mario Bustamante
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; and
| | | | | | | |
Collapse
|
50
|
Pal M, Febbraio MA, Whitham M. From cytokine to myokine: the emerging role of interleukin-6 in metabolic regulation. Immunol Cell Biol 2014; 92:331-9. [PMID: 24751614 DOI: 10.1038/icb.2014.16] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 02/09/2014] [Accepted: 02/10/2014] [Indexed: 12/11/2022]
Abstract
The lack of physical activity and overnutrition in our modern lifestyle culminates in what we now experience as the current obesity and diabetes pandemic. Medical research performed over the past 20 years identified chronic low-grade inflammation as a mediator of these metabolic disorders. Hence, finding therapeutic strategies against this underlying inflammation and identifying molecules implicated in this process is of significant importance. Following the observation of an increased plasma concentration of interleukin-6 (IL-6) in obese patients, this protein, known predominantly as a pro-inflammatory cytokine, came into focus. In an attempt to clarify its importance, several studies implicated IL-6 as a co-inducer of the development of obesity-associated insulin resistance, which precedes the development of type 2 diabetes. However, the identification of IL-6 as a myokine, a protein produced and secreted by skeletal muscle to fulfil paracrine or endocrine roles in the insulin-sensitizing effects following exercise, provides a contrasting and hence paradoxical identity of this protein in the context of metabolism. We review here the literature considering the complex, pleiotropic role of IL-6 in the context of metabolism in health and disease.
Collapse
Affiliation(s)
- Martin Pal
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart and Diabetes Research Institute, Melbourne, Victoria, Australia
| | - Mark A Febbraio
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart and Diabetes Research Institute, Melbourne, Victoria, Australia
| | - Martin Whitham
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart and Diabetes Research Institute, Melbourne, Victoria, Australia
| |
Collapse
|