1
|
Nascimento Júnior JXD, Gomes JDC, Imbroisi Filho R, Valença HDM, Branco JR, Araújo AB, Moreira ADOE, Crepaldi LD, Paixão LP, Ochioni AC, Demaria TM, Leandro JGB, Casanova LM, Sola-Penna M, Zancan P. Dietary caloric input and tumor growth accelerate senescence and modulate liver and adipose tissue crosstalk. Commun Biol 2025; 8:18. [PMID: 39775048 PMCID: PMC11707351 DOI: 10.1038/s42003-025-07451-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025] Open
Abstract
Metabolic alterations are related to tumorigenesis and other age-related diseases that are accelerated by "Westernized" diets. In fact, hypercaloric nutrition is associated with an increased incidence of cancers and faster aging. Conversely, lifespan-extending strategies, such as caloric restriction, impose beneficial effects on both processes. Here, we investigated the metabolic consequences of hypercaloric-induced aging on tumor growth in female mice. Our findings indicate that a high-fat high-sucrose diet increases tumor growth mainly due to the boosted oxidation of glucose and fatty acids. Consequently, through an increased expression of lactate, IGFBP3, and PTHLH, tumors modulate liver and white adipose tissue metabolism. In the liver, the induced tumor increases fibrosis and accelerates the senescence process, despite the lower systemic pro-inflammatory state. Importantly, the induced tumor induces the wasting and browning of white adipose tissue, thereby reversing diet-induced insulin resistance. Finally, we suggest that tumor growth alters liver-adipose tissue crosstalk that upregulates Fgf21, induces senescence, and negatively modulates lipids and carbohydrates metabolism even in caloric-restricted-fed mice.
Collapse
Affiliation(s)
- José Xavier do Nascimento Júnior
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Júlia da Conceição Gomes
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo Imbroisi Filho
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Helber de Maia Valença
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jéssica Ristow Branco
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amanda Bandeira Araújo
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amanda de Oliveira Esteves Moreira
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Letícia Diniz Crepaldi
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Larissa Pereira Paixão
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alan C Ochioni
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thainá M Demaria
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - João Gabriel Bernardo Leandro
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Livia Marques Casanova
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mauro Sola-Penna
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Zancan
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
2
|
Chirivi M, Contreras GA. Endotoxin-induced alterations of adipose tissue function: a pathway to bovine metabolic stress. J Anim Sci Biotechnol 2024; 15:53. [PMID: 38581064 PMCID: PMC10998405 DOI: 10.1186/s40104-024-01013-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/14/2024] [Indexed: 04/07/2024] Open
Abstract
During the periparturient period, dairy cows exhibit negative energy balance due to limited appetite and increased energy requirements for lactogenesis. The delicate equilibrium between energy availability and expenditure puts cows in a state of metabolic stress characterized by excessive lipolysis in white adipose tissues (AT), increased production of reactive oxygen species, and immune cell dysfunction. Metabolic stress, especially in AT, increases the risk for metabolic and inflammatory diseases. Around parturition, cows are also susceptible to endotoxemia. Bacterial-derived toxins cause endotoxemia by promoting inflammatory processes and immune cell infiltration in different organs and systems while impacting metabolic function by altering lipolysis, mitochondrial activity, and insulin sensitivity. In dairy cows, endotoxins enter the bloodstream after overcoming the defense mechanisms of the epithelial barriers, particularly during common periparturient conditions such as mastitis, metritis, and pneumonia, or after abrupt changes in the gut microbiome. In the bovine AT, endotoxins induce a pro-inflammatory response and stimulate lipolysis in AT, leading to the release of free fatty acids into the bloodstream. When excessive and protracted, endotoxin-induced lipolysis can impair adipocyte's insulin signaling pathways and lipid synthesis. Endotoxin exposure can also induce oxidative stress in AT through the production of reactive oxygen species by inflammatory cells and other cellular components. This review provides insights into endotoxins' impact on AT function, highlighting the gaps in our knowledge of the mechanisms underlying AT dysfunction, its connection with periparturient cows' disease risk, and the need to develop effective interventions to prevent and treat endotoxemia-related inflammatory conditions in dairy cattle.
Collapse
Affiliation(s)
- Miguel Chirivi
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - G Andres Contreras
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
3
|
Cimas FJ, De la Cruz-Morcillo MÁ, Cifuentes C, Moratalla-López N, Alonso GL, Nava E, Llorens S. Effect of Crocetin on Basal Lipolysis in 3T3-L1 Adipocytes. Antioxidants (Basel) 2023; 12:1254. [PMID: 37371984 DOI: 10.3390/antiox12061254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Crocetin (CCT) is a natural saffron-derived apocarotenoid that possesses healthy properties such as anti-adipogenic, anti-inflammatory, and antioxidant activities. Lipolysis is enhanced in obesity and correlates with a pro-inflammatory, pro-oxidant state. In this context, we aimed to investigate whether CCT affects lipolysis. To evaluate CCT's possible lipolytic effect, 3T3-L1 adipocytes were treated with CCT10μM at day 5 post-differentiation. Glycerol content and antioxidant activity were assessed using colorimetric assays. Gene expression was measured using qRT-PCR to evaluate the effect of CCT on key lipolytic enzymes and on nitric oxide synthase (NOS) expression. Total lipid accumulation was assessed using Oil Red O staining. CCT10μM decreased glycerol release from 3T3-L1 adipocytes and downregulated adipose tissue triglyceride lipase (ATGL) and perilipin-1, but not hormone-sensitive lipase (HSL), suggesting an anti-lipolytic effect. CCT increased catalase (CAT) and superoxide dismutase (SOD) activity, thus showing an antioxidant effect. In addition, CCT exhibited an anti-inflammatory profile, i.e., diminished inducible NOS (NOS2) and resistin expression, while enhanced the expression of adiponectin. CCT10μM also decreased intracellular fat and C/EBPα expression (a transcription factor involved in adipogenesis), thus revealing an anti-adipogenic effect. These findings point to CCT as a promising biocompound for improving lipid mobilisation in obesity.
Collapse
Affiliation(s)
- Francisco J Cimas
- Mecenazgo COVID-19, Regional Center for Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), 02008 Albacete, Spain
| | - Miguel Ángel De la Cruz-Morcillo
- Food Quality Research Group, Institute for Regional Development (IDR), Campus Universitario s/n, University of Castilla-La Mancha (UCLM), 02071 Albacete, Spain
| | - Carmen Cifuentes
- Regional Center for Biomedical Research (CRIB), Department of Medical Sciences, Faculty of Medicine of Albacete, University of Castilla-La Mancha (UCLM), 02008 Albacete, Spain
| | - Natalia Moratalla-López
- Cátedra de Química Agrícola, Higher Technical School of Agronomic and Forestry Engineering and Biotechnology (ETSIAMB), University of Castilla-La Mancha (UCLM), Campus Universitario, 02006 Albacete, Spain
| | - Gonzalo L Alonso
- Cátedra de Química Agrícola, Higher Technical School of Agronomic and Forestry Engineering and Biotechnology (ETSIAMB), University of Castilla-La Mancha (UCLM), Campus Universitario, 02006 Albacete, Spain
| | - Eduardo Nava
- Regional Center for Biomedical Research (CRIB), Department of Medical Sciences, Faculty of Medicine of Albacete, University of Castilla-La Mancha (UCLM), 02008 Albacete, Spain
| | - Sílvia Llorens
- Regional Center for Biomedical Research (CRIB), Department of Medical Sciences, Faculty of Medicine of Albacete, University of Castilla-La Mancha (UCLM), 02008 Albacete, Spain
| |
Collapse
|
4
|
Pappas G, Wilkinson ML, Gow AJ. Nitric oxide regulation of cellular metabolism: Adaptive tuning of cellular energy. Nitric Oxide 2023; 131:8-17. [PMID: 36470373 PMCID: PMC9839556 DOI: 10.1016/j.niox.2022.11.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/24/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Nitric oxide can interact with a wide range of proteins including many that are involved in metabolism. In this review we have summarized the effects of NO on glycolysis, fatty acid metabolism, the TCA cycle, and oxidative phosphorylation with reference to skeletal muscle. Low to moderate NO concentrations upregulate glucose and fatty acid oxidation, while higher NO concentrations shift cellular reliance toward a fully glycolytic phenotype. Moderate NO production directly inhibits pyruvate dehydrogenase activity, reducing glucose-derived carbon entry into the TCA cycle and subsequently increasing anaploretic reactions. NO directly inhibits aconitase activity, increasing reliance on glutamine for continued energy production. At higher or prolonged NO exposure, citrate accumulation can inhibit multiple ATP-producing pathways. Reduced TCA flux slows NADH/FADH entry into the ETC. NO can also inhibit the ETC directly, further limiting oxidative phosphorylation. Moderate NO production improves mitochondrial efficiency while improving O2 utilization increasing whole-body energy production. Long-term bioenergetic capacity may be increased because of NO-derived ROS, which participate in adaptive cellular redox signaling through AMPK, PCG1-α, HIF-1, and NF-κB. However, prolonged exposure or high concentrations of NO can result in membrane depolarization and opening of the MPT. In this way NO may serve as a biochemical rheostat matching energy supply with demand for optimal respiratory function.
Collapse
Affiliation(s)
- Gregory Pappas
- Department of Kinesiology & Applied Physiology, Rutgers the State University of New Jersey, NJ, 08854, USA.
| | - Melissa L Wilkinson
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers the State University of New Jersey, NJ, 08854, USA.
| | - Andrew J Gow
- Department of Kinesiology & Applied Physiology, Rutgers the State University of New Jersey, NJ, 08854, USA; Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers the State University of New Jersey, NJ, 08854, USA.
| |
Collapse
|
5
|
Caputa G, Matsushita M, Sanin DE, Kabat AM, Edwards-Hicks J, Grzes KM, Pohlmeyer R, Stanczak MA, Castoldi A, Cupovic J, Forde AJ, Apostolova P, Seidl M, van Teijlingen Bakker N, Villa M, Baixauli F, Quintana A, Hackl A, Flachsmann L, Hässler F, Curtis JD, Patterson AE, Henneke P, Pearce EL, Pearce EJ. Intracellular infection and immune system cues rewire adipocytes to acquire immune function. Cell Metab 2022; 34:747-760.e6. [PMID: 35508110 DOI: 10.1016/j.cmet.2022.04.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 01/24/2022] [Accepted: 04/13/2022] [Indexed: 12/11/2022]
Abstract
Adipose tissue (AT) plays a central role in systemic metabolic homeostasis, but its function during bacterial infection remains unclear. Following subcutaneous bacterial infection, adipocytes surrounding draining lymph nodes initiated a transcriptional response indicative of stimulation with IFN-γ and a shift away from lipid metabolism toward an immunologic function. Natural killer (NK) and invariant NK T (iNKT) cells were identified as sources of infection-induced IFN-γ in perinodal AT (PAT). IFN-γ induced Nos2 expression in adipocytes through a process dependent on nuclear-binding oligomerization domain 1 (NOD1) sensing of live intracellular bacteria. iNOS expression was coupled to metabolic rewiring, inducing increased diversion of extracellular L-arginine through the arginosuccinate shunt and urea cycle to produce nitric oxide (NO), directly mediating bacterial clearance. In vivo, control of infection in adipocytes was dependent on adipocyte-intrinsic sensing of IFN-γ and expression of iNOS. Thus, adipocytes are licensed by innate lymphocytes to acquire anti-bacterial functions during infection.
Collapse
Affiliation(s)
- George Caputa
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Mai Matsushita
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - David E Sanin
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany; Bloomberg Kimmel Institute, and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Agnieszka M Kabat
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Joy Edwards-Hicks
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Katarzyna M Grzes
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Roland Pohlmeyer
- Imaging Facility, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Michal A Stanczak
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Angela Castoldi
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Jovana Cupovic
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Aaron J Forde
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; Center for Chronic Immune Deficiency, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Petya Apostolova
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Maximilian Seidl
- Center for Chronic Immune Deficiency, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Institute of Surgical Pathology, Faculty of Medicine, Medical Center, University of Freiburg, 79104 Freiburg, Germany; Institute of Pathology, Heinrich Heine University and University Hospital of Duesseldorf, 40225 Duesseldorf, Germany
| | - Nikki van Teijlingen Bakker
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Matteo Villa
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Francesc Baixauli
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Andrea Quintana
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Alexandra Hackl
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Lea Flachsmann
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Fabian Hässler
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Jonathan D Curtis
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Annette E Patterson
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Philipp Henneke
- Center for Chronic Immune Deficiency, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Erika L Pearce
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany; Bloomberg Kimmel Institute, and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA.
| | - Edward J Pearce
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; Bloomberg Kimmel Institute, and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA.
| |
Collapse
|
6
|
Vilela VR, Samson N, Nachbar R, Perazza LR, Lachance G, Rokatoarivelo V, Centano-Baez C, Zancan P, Sola-Penna M, Bellmann K, Di Marzo V, Laplante M, Marette A. Adipocyte-specific Nos2 deletion improves insulin resistance and dyslipidemia through brown fat activation in diet-induced obese mice. Mol Metab 2022; 57:101437. [PMID: 35033724 PMCID: PMC8802131 DOI: 10.1016/j.molmet.2022.101437] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 12/22/2021] [Accepted: 01/03/2022] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Inducible nitric oxide (NO) synthase (NOS2) is a well documented inflammatory mediator of insulin resistance in obesity. NOS2 expression is induced in both adipocytes and macrophages within adipose tissue during high-fat (HF)-induced obesity. METHODS Eight week old male mice with adipocyte selective deletion of the Nos2 gene (Nos2AD-KO) and their wildtype littermates (Nos2fl/fl) were subjected to chow or high-fat high-sucrose (HFHS) diet for 10 weeks followed by metabolic phenotyping and determination of brown adipose tissue (BAT) thermogenesis. The direct impact of NO on BAT mitochondrial respiration was also assessed in brown adipocytes. RESULTS Here, we show that HFHS-fed Nos2AD-KO mice had improved insulin sensitivity as compared to Nos2fl/fl littermates. Nos2AD-KO mice were also protected from HF-induced dyslipidemia and exhibited increased energy expenditure compared to Nos2fl/fl mice. This was linked to activation of BAT in HFHS-fed Nos2AD-KO mice as shown by increased Ucp1 and Ucp2 gene expression and augmented respiratory capacity of BAT mitochondria. Furthermore, mitochondrial respiration was inhibited by NO, or upon cytokine-induced NOS2 activation, but improved by NOS2 inhibition in brown adipocytes. CONCLUSIONS These results demonstrate a key role for adipocyte NOS2 in the development of obesity-linked insulin resistance and dyslipidemia, partly through NO dependent inhibition of BAT mitochondrial bioenergetics.
Collapse
Affiliation(s)
| | - Nolwenn Samson
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Renato Nachbar
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Lia Rossi Perazza
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Gabriel Lachance
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Volatiana Rokatoarivelo
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Carolina Centano-Baez
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Patricia Zancan
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Mauro Sola-Penna
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Kerstin Bellmann
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - Vincenzo Di Marzo
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada; Institute of Nutrition and Functional Foods, Centre NUTRISS, Université Laval, 2440 Boulevard Hochelaga Suite 1710, Québec, QC, G1V 0A6, Canada; Canada Excellence Research Chair Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND)
| | - Mathieu Laplante
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada
| | - André Marette
- Quebec Heart & Lung Institute, Université Laval, 2725 Ch Ste-Foy, Québec, QC, G1V 4G5, Canada; Institute of Nutrition and Functional Foods, Centre NUTRISS, Université Laval, 2440 Boulevard Hochelaga Suite 1710, Québec, QC, G1V 0A6, Canada.
| |
Collapse
|
7
|
Qin A, Chen S, Wang P, Huang X, Zhang Y, Liang L, Du LR, Lai DH, Ding L, Yu X, Xiang AP. Knockout of NOS2 Promotes Adipogenic Differentiation of Rat MSCs by Enhancing Activation of JAK/STAT3 Signaling. Front Cell Dev Biol 2021; 9:638518. [PMID: 33816486 PMCID: PMC8017136 DOI: 10.3389/fcell.2021.638518] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are a heterogeneous population of cells that possess multilineage differentiation potential and extensive immunomodulatory properties. In mice and rats, MSCs produce nitric oxide (NO), as immunomodulatory effector molecule that exerts an antiproliferative effect on T cells, while the role of NO in differentiation was less clear. Here, we investigated the role of NO synthase 2 (NOS2) on adipogenic and osteogenic differentiation of rat MSCs. MSCs isolated from NOS2-null (NOS2–/–) and wild type (WT) Sprague–Dawley (SD) rats exhibited homogenous fibroblast-like morphology and characteristic phenotypes. However, after induction, adipogenic differentiation was found significantly promoted in NOS2–/– MSCs compared to WT MSCs, but not in osteogenic differentiation. Accordingly, qRT-PCR revealed that the adipogenesis-related genes PPAR-γ, C/EBP-α, LPL and FABP4 were markedly upregulated in NOS2–/– MSCs, but not for osteogenic transcription factors or marker genes. Further investigations revealed that the significant enhancement of adipogenic differentiation in NOS2–/– MSCs was due to overactivation of the STAT3 signaling pathway. Both AG490 and S3I-201, small molecule inhibitors that selectively inhibit STAT3 activation, reversed this adipogenic effect. Furthermore, after high-fat diet (HFD) feeding, knockout of NOS2 in rat MSCs resulted in significant obesity. In summary, NOS2 is involved in the regulation of rat MSC adipogenic differentiation via the STAT3 signaling pathway.
Collapse
Affiliation(s)
- Aiping Qin
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Sheng Chen
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ping Wang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xiaotao Huang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yu Zhang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Lu Liang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ling-Ran Du
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - De-Hua Lai
- Center for Parasitic Organisms, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Li Ding
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiyong Yu
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
8
|
Boudreau A, Burke S, Collier J, Richard AJ, Ribnicky DM, Stephens JM. Mechanisms of Artemisia scoparia's Anti-Inflammatory Activity in Cultured Adipocytes, Macrophages, and Pancreatic β-Cells. Obesity (Silver Spring) 2020; 28:1726-1735. [PMID: 32741148 PMCID: PMC7483878 DOI: 10.1002/oby.22912] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/22/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVE An ethanolic extract of Artemisia scoparia (SCO) improves adipose tissue function and reduces negative metabolic consequences of high-fat feeding. A. scoparia has a long history of medicinal use across Asia and has anti-inflammatory effects in various cell types and disease models. The objective of the current study was to investigate SCO's effects on inflammation in cells relevant to metabolic health. METHODS Inflammatory responses were assayed in cultured adipocytes, macrophages, and insulinoma cells by quantitative polymerase chain reaction, immunoblotting, and NF-κB reporter assays. RESULTS In tumor necrosis factor α-treated adipocytes, SCO mitigated ERK and NF-κB signaling as well as transcriptional responses but had no effect on fatty acid-binding protein 4 secretion. SCO also reduced levels of deleted in breast cancer 1 protein in adipocytes and inhibited inflammatory gene expression in stimulated macrophages. Finally, in pancreatic β-cells, SCO decreased NF-κB-responsive promoter activity induced by IL-1β treatment. CONCLUSIONS SCO's ability to promote adipocyte development and function is thought to mediate its insulin-sensitizing actions in vivo. Our findings that SCO inhibits inflammatory responses through at least two distinct signaling pathways (ERK and NF-κB) in three cell types known to contribute to metabolic disease reveal that SCO may act more broadly than previously thought to improve metabolic health.
Collapse
Affiliation(s)
- Anik Boudreau
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | - Susan Burke
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | - Jason Collier
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | | | - David M. Ribnicky
- Department of Plant Biology and Pathology, Rutgers University, New Brunswick, NJ
| | - Jacqueline M. Stephens
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803
- To whom correspondence should be addressed: Jacqueline Stephens, Louisiana State University, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, Phone (225) 763-2648, FAX (225) 578-2597,
| |
Collapse
|
9
|
Systemic Insulin Resistance and Metabolic Perturbations in Chow Fed Inducible Nitric Oxide Synthase Knockout Male Mice: Partial Reversal by Nitrite Supplementation. Antioxidants (Basel) 2020; 9:antiox9080736. [PMID: 32806494 PMCID: PMC7465804 DOI: 10.3390/antiox9080736] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/06/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023] Open
Abstract
iNOS, an important mediator of inflammation, has emerged as an important metabolic regulator. There are conflicting observations on the incidence of insulin resistance (IR) due to hyperglycemia/dyslipidemia in iNOS−/− mice. There are reports that high fat diet (HFD) fed mice exhibited no change, protection, or enhanced susceptibility to IR. Similar observations were also reported for low fat diet (LFD) fed KO mice. In the present study chow fed iNOS−/− mice were examined for the incidence of IR, and metabolic perturbations, and also for the effect of sodium nitrite supplementation (50 mg/L). In IR-iNOS−/− mice, we observed significantly higher body weight, BMI, adiposity, blood glucose, HOMA-IR, serum/tissue lipids, glucose intolerance, enhanced gluconeogenesis, and disrupted insulin signaling. Expression of genes involved in hepatic and adipose tissue lipid uptake, synthesis, oxidation, and gluconeogenesis was upregulated with concomitant downregulation of genes for hepatic lipid excretion. Nitrite supplementation restored NO levels, significantly improved systemic IR, glucose tolerance, and also reduced lipid accumulation by rescuing hepatic insulin sensitivity, glucose, and lipid homeostasis. Obesity, gluconeogenesis, and adipose tissue insulin signaling were only partially reversed in nitrite supplemented iNOS−/− mice. Our results thus demonstrate that nitrite supplementation to iNOS−/− mice improves insulin sensitivity and metabolic homeostasis, thus further highlighting the metabolic role of iNOS.
Collapse
|
10
|
Zeinali Khosroshahi M, Asbaghi O, Moradi S, Rezaei kelishadi M, Kaviani M, Mardani M, Jalili C. The effects of supplementation with L-arginine on anthropometric indices and body composition in overweight or obese subjects: A systematic review and meta-analysis. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
11
|
Becerril S, Rodríguez A, Catalán V, Ramírez B, Unamuno X, Portincasa P, Gómez-Ambrosi J, Frühbeck G. Functional Relationship between Leptin and Nitric Oxide in Metabolism. Nutrients 2019; 11:2129. [PMID: 31500090 PMCID: PMC6769456 DOI: 10.3390/nu11092129] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/23/2019] [Accepted: 09/02/2019] [Indexed: 12/28/2022] Open
Abstract
Leptin, the product of the ob gene, was originally described as a satiety factor, playing a crucial role in the control of body weight. Nevertheless, the wide distribution of leptin receptors in peripheral tissues supports that leptin exerts pleiotropic biological effects, consisting of the modulation of numerous processes including thermogenesis, reproduction, angiogenesis, hematopoiesis, osteogenesis, neuroendocrine, and immune functions as well as arterial pressure control. Nitric oxide (NO) is a free radical synthesized from L-arginine by the action of the NO synthase (NOS) enzyme. Three NOS isoforms have been identified: the neuronal NOS (nNOS) and endothelial NOS (eNOS) constitutive isoforms, and the inducible NOS (iNOS). NO mediates multiple biological effects in a variety of physiological systems such as energy balance, blood pressure, reproduction, immune response, or reproduction. Leptin and NO on their own participate in multiple common physiological processes, with a functional relationship between both factors having been identified. The present review describes the functional relationship between leptin and NO in different physiological processes.
Collapse
Affiliation(s)
- Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Xabier Unamuno
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Medical Engineering Laboratory, University of Navarra, 31008 Pamplona, Spain.
| | - Piero Portincasa
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Policlinico Hospital, 70124 Bari, Italy.
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
| |
Collapse
|
12
|
Foroumandi E, Alizadeh M, Kheirouri S, Asghari Jafarabadi M. Exploring the role of body mass index in relationship of serum nitric oxide and advanced glycation end products in apparently healthy subjects. PLoS One 2019; 14:e0213307. [PMID: 30856212 PMCID: PMC6411143 DOI: 10.1371/journal.pone.0213307] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/18/2019] [Indexed: 11/19/2022] Open
Abstract
This study aimed to identify any association of serum nitric oxide (NO) and advanced glycation end products (AGEs) with body mass index (BMI) in apparently healthy subjects. In this cross-sectional study, participants were 90 apparently healthy subjects, categorized into three BMI groups as follows: BMI≤19.5 (n = 21), 19.6≤BMI≤24.9 (n = 35), and BMI≥25 (n = 34). Serum levels of NO were measured by griess reaction method. Determination of serum pentosidine and carboxymethyllysine (CML) was done using ELISA. Median (95% confidence interval [CI]: lower- upper) of serum NO in subjects with BMI≥25 were 68.94 (CI: 55.01–70.56) μmol/L, which was higher compared with 19.6≤BMI≤24.9 and BMI≤19.5 groups (22.65 (CI: 19.29–28.17) μmol/L and 8.00 (CI: 9.12–29.58) μmol/L, respectively). Serum NO positively correlated with BMI in total subjects (r = 0.585, p<0.001), which this correlation was significant in both male and female groups (r = 0.735, p<0.001 and r = 0.476, p = 0.001, respectively). Serum pentosidine and CML were significantly lower in subjects with higher BMI. Further, BMI showed negative correlations with pentosidine and CML (r = -0.363, p<0.001 and r = -0.484, p<0.001, respectively). There were not any significant differences in serum NO, pentosidine, and CML levels between sex groups. After adjusting the effects of confounders (BMI, sex, age, and waist to hip ratio), serum NO significantly correlated with serum pentosidine and CML (r = -0.319, p = 0.003 and r = -0.433, p<0.001, respectively). It is concluded that higher BMI is accompanied by increased serum NO and suppressed pentosidine and CML.
Collapse
Affiliation(s)
- Elaheh Foroumandi
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Students’ Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Alizadeh
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- * E-mail:
| | - Sorayya Kheirouri
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
13
|
Issa N, Lachance G, Bellmann K, Laplante M, Stadler K, Marette A. Cytokines promote lipolysis in 3T3-L1 adipocytes through induction of NADPH oxidase 3 expression and superoxide production. J Lipid Res 2018; 59:2321-2328. [PMID: 30317185 DOI: 10.1194/jlr.m086504] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 10/12/2018] [Indexed: 01/14/2023] Open
Abstract
NADPH oxidase (NOX) enzymes are one of the major superoxide-generating systems in cells. NOX-generated superoxide has been suggested to promote insulin resistance in the liver. However, the role of NOX enzymes in mediating metabolic dysfunction in other insulin target tissues remains unclear. Here, we show that NOX3 expression is induced in differentiated 3T3-L1 adipocytes upon treatment with proinflammatory cytokines. Superoxide production increased concurrently with NOX3 protein expression in cytokine-treated adipocytes, which was inhibited by the NOX inhibitor diphenyleneiodonium (DPI). Treatment of adipocytes with cytokines increased lipolysis and decreased PPARγ activity. Interestingly, treatment with DPI blunted lipolysis activation by cytokines but failed to restore PPARγ activity. siRNA-mediated NOX3 downregulation also prevented cytokine-induced superoxide generation and lipolysis. In line with increasing lipolysis, cytokines increased the phosphorylation of hormone-sensitive lipase (HSL), which was reversed by treatment with DPI and silencing of NOX3 expression. We conclude that NOX3 is a cytokine-inducible superoxide-generating enzyme in adipocytes, which promotes lipolysis through increasing phosphorylation of HSL. This suggests a key role for NOX3-mediated superoxide production in the increased adipocyte lipolysis in inflammatory settings.
Collapse
Affiliation(s)
- Nahla Issa
- Department of Medicine, Cardiology Axis of the Quebec Heart and Lung Research Institute, Laval University, Quebec, QC, Canada
| | - Gabriel Lachance
- Department of Medicine, Cardiology Axis of the Quebec Heart and Lung Research Institute, Laval University, Quebec, QC, Canada
| | - Kerstin Bellmann
- Department of Medicine, Cardiology Axis of the Quebec Heart and Lung Research Institute, Laval University, Quebec, QC, Canada
| | - Mathieu Laplante
- Department of Medicine, Cardiology Axis of the Quebec Heart and Lung Research Institute, Laval University, Quebec, QC, Canada.,Cancer Research Center (CRC), Laval University, Quebec, QC, Canada
| | - Krisztian Stadler
- Oxidative Stress and Disease Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - André Marette
- Department of Medicine, Cardiology Axis of the Quebec Heart and Lung Research Institute, Laval University, Quebec, QC, Canada
| |
Collapse
|
14
|
Allerton TD, Proctor DN, Stephens JM, Dugas TR, Spielmann G, Irving BA. l-Citrulline Supplementation: Impact on Cardiometabolic Health. Nutrients 2018; 10:nu10070921. [PMID: 30029482 PMCID: PMC6073798 DOI: 10.3390/nu10070921] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 07/09/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022] Open
Abstract
Diminished bioavailability of nitric oxide (NO), the gaseous signaling molecule involved in the regulation of numerous vital biological functions, contributes to the development and progression of multiple age- and lifestyle-related diseases. While l-arginine is the precursor for the synthesis of NO by endothelial-nitric oxide synthase (eNOS), oral l-arginine supplementation is largely ineffective at increasing NO synthesis and/or bioavailability for a variety of reasons. l-citrulline, found in high concentrations in watermelon, is a neutral alpha-amino acid formed by enzymes in the mitochondria that also serves as a substrate for recycling l-arginine. Unlike l-arginine, l-citrulline is not quantitatively extracted from the gastrointestinal tract (i.e., enterocytes) or liver and its supplementation is therefore more effective at increasing l-arginine levels and NO synthesis. Supplementation with l-citrulline has shown promise as a blood pressure lowering intervention (both resting and stress-induced) in adults with pre-/hypertension, with pre-clinical (animal) evidence for atherogenic-endothelial protection. Preliminary evidence is also available for l-citrulline-induced benefits to muscle and metabolic health (via vascular and non-vascular pathways) in susceptible/older populations. In this review, we examine the impact of supplementing this important urea cycle intermediate on cardiovascular and metabolic health outcomes and identify future directions for investigating its therapeutic impact on cardiometabolic health.
Collapse
Affiliation(s)
| | - David N Proctor
- Department of Kinesiology, Pennsylvania State University, University Park, PA 16802, USA.
| | | | - Tammy R Dugas
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Guillaume Spielmann
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
- Department of Kinesiology, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Brian A Irving
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
- Department of Kinesiology, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
15
|
Serum nitric oxide metabolites are associated with the risk of hypertriglyceridemic-waist phenotype in women: Tehran Lipid and Glucose Study. Nitric Oxide 2015; 50:52-57. [PMID: 26284308 DOI: 10.1016/j.niox.2015.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 08/12/2015] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIM There are some controversial issues regarding the association of nitric oxide and obesity-related states. This study was conducted to investigate whether serum nitric oxide metabolites (NOx) could predict the occurrence of visceral lipid accumulation, defined as hypertriglyceridemic-waist (HTW) phenotype. METHODS We used a prospective approach for this study conducted on participants of the Tehran Lipid and Glucose Study, 2243 adult men and women were followed for a median of 6.3 years. Serum NOx concentrations were measured at baseline (2006-2008), and demographics, anthropometrics and biochemical variables were evaluated at baseline and again after a 3-year (2009-2011) and a 6-year follow-up (2012-2014). The occurrence of HTW phenotype, defined as waist circumference ≥90 cm in men and ≥85 cm in women, along with serum triglyceride levels ≥177 mg/dL, were assessed across serum NOx tertiles. RESULTS Mean age of participants was 41.5 ± 14.5 years at baseline and 39.4% were male. The cumulative incidence of HTW phenotype was 37.6% (33.2% in men, 40.5% in women). There was no significant association between serum NOx and the occurrence of HTW phenotype in men. After adjustment of confounding variables, risk of HTW phenotype in women, in the highest compared to the lowest tertile of serum NOx (≥30.9 vs. <19.9 μmol/L), increased by 39% (OR = 1.39, 95% CI = 1.05-1.93, P for trend = 0.053). CONCLUSION Serum NOx level was an independent predictor of HTW phenotype in women.
Collapse
|
16
|
Sarı E, Yeşilkaya E, Bolat A, Topal T, Altan B, Fidancı K, Saldır M, Erdem G, Gülgün M, Gülcan Kurt Y, Güven A. Metabolic and Histopathological Effects of Fructose Intake During Pregestation, Gestation and Lactation in Rats and their Offspring. J Clin Res Pediatr Endocrinol 2015; 7:19-26. [PMID: 25800472 PMCID: PMC4439888 DOI: 10.4274/jcrpe.1776] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
OBJECTIVE Studies have demonstrated a significant relationship between maternal fructose intake and metabolic outcome in their offspring. However, there is a paucity of data about the long-term effects of fructose intake on the offspring of fructose-fed dams. Therefore, we planned a study to evaluate the long-term effects of fructose intake on the offspring of dam rats fed a high-fructose diet. METHODS Sixteen virgin female Sprague-Dawley rats were divided into two groups. Group 1 received a regular diet and Group 2 a high-fructose diet. Both groups received their experimental diets for 8 weeks before conception. They were mated and continued to feed with their experimental diet during mating and during their pregnancy and lactation periods. After weaning, the offspring from each group were divided into two groups. Group 1A received a regular diet, Group 1B - a fructose diet, Group 2A - a regular diet and Group 2B received a fructose diet. After weaning, the offspring were anesthetized and blood samples were collected for biochemical analysis. Liver, kidney and retroperitoneal adipose tissue were harvested for histopathological examination. Primary antibodies against inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) were determined as early inflammation markers. RESULTS After weaning, while daily water consumption was found to be significantly higher in Groups 2B and 1B (p<0.01), daily laboratory chow consumption was significantly lower in Groups 1A and 2A (p<0.01). Body weight was significantly higher in Groups 1B and 2B (p<0.01). Serum glucose, triglyceride, low-density lipoprotein cholesterol and very low-density lipoprotein cholesterol levels were found to be increased and high-density lipoprotein cholesterol levels decreased in Group 2B (p<0.05). The intensities of iNOS staining in the retroperitoneal adipose tissue, COX-2 staining in the liver and both iNOS and COX-2 staining in the kidney were higher in Group 2B (p<0.05). CONCLUSION Based on our findings, we believe that the offspring of dams which received a high fructose intake during their pregestation, gestation and lactation periods are at risk of developing metabolic syndrome in their later life only if they continue to receive a high intake of fructose. We therefore propose that the risk of developing metabolic syndrome can probably be reduced by modifying the diet of the offspring after weaning.
Collapse
Affiliation(s)
- Erkan Sarı
- Gülhane Military Medical Academy, Department of Pediatric Endocrinology, Ankara, Turkey. E-mail:
| | - Ediz Yeşilkaya
- Gülhane Military Medical Academy, Department of Pediatric Endocrinology, Ankara, Turkey
,* Address for Correspondence: Gülhane Military Medical Academy, Department of Pediatric Endocrinology, Ankara, Turkey Phone: +90 312 304 18 98 E-mail:
| | - Ahmet Bolat
- Gülhane Military Medical Academy, Department of Pediatric Endocrinology, Ankara, Turkey
| | - Turgut Topal
- Gülhane Military Medical Academy, Department of Physiology, Ankara, Turkey
| | - Bilal Altan
- Gülhane Military Medical Academy, Department of Pediatric Surgery, Ankara, Turkey
| | - Kürşat Fidancı
- Gülhane Military Medical Academy, Department of Pediatric Endocrinology, Ankara, Turkey
| | - Mehmet Saldır
- Gülhane Military Medical Academy, Department of Pediatric Endocrinology, Ankara, Turkey
| | - Galip Erdem
- Gülhane Military Medical Academy, Department of Pediatric Endocrinology, Ankara, Turkey
| | - Mustafa Gülgün
- Gülhane Military Medical Academy, Department of Pediatric Endocrinology, Ankara, Turkey
| | - Yasemin Gülcan Kurt
- Gulhane Military Medical Academy, Department of Biochemistry, Ankara, Turkey
| | - Ahmet Güven
- Gülhane Military Medical Academy, Department of Pediatric Surgery, Ankara, Turkey
| |
Collapse
|
17
|
Wen JJ, Nagajyothi F, Machado FS, Weiss LM, Scherer PE, Tanowitz HB, Garg NJ. Markers of oxidative stress in adipose tissue during Trypanosoma cruzi infection. Parasitol Res 2014; 113:3159-65. [PMID: 24948102 DOI: 10.1007/s00436-014-3977-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Accepted: 05/27/2014] [Indexed: 12/31/2022]
Abstract
The protozoan parasite Trypanosoma cruzi causes Chagas disease. Cardiac and adipose tissues are among the early targets of infection and are sites of persistent infection. In the heart and adipose tissue, T. cruzi infection results in an upregulation of pro-inflammatory mediators. In the heart, infection is associated with an increase in the markers of oxidative stress. To date, markers of oxidative stress have not been evaluated in adipose tissue in this infection. Brown and white adipose tissues were obtained from CD-1 mice infected with the Brazil strain of T. cruzi for 15, 30, and 130 days post infection. Protein carbonylation and lipid peroxidation assays were performed on these samples. There was an upregulation of these markers of oxidative stress at all time-points in both white and brown adipose tissue. Determinants of anti-oxidative stress were downregulated at similar time-points. This increase in oxidative stress during T. cruzi infection most likely has a deleterious effect on host metabolism and on the heart.
Collapse
Affiliation(s)
- Jian-Jun Wen
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
In adipocytes the hydrolysis of TAG to produce fatty acids and glycerol under fasting conditions or times of elevated energy demands is tightly regulated by neuroendocrine signals, resulting in the activation of lipolytic enzymes. Among the classic regulators of lipolysis, adrenergic stimulation and the insulin-mediated control of lipid mobilisation are the best known. Initially, hormone-sensitive lipase (HSL) was thought to be the rate-limiting enzyme of the first lipolytic step, while we now know that adipocyte TAG lipase is the key enzyme for lipolysis initiation. Pivotal, previously unsuspected components have also been identified at the protective interface of the lipid droplet surface and in the signalling pathways that control lipolysis. Perilipin, comparative gene identification-58 (CGI-58) and other proteins of the lipid droplet surface are currently known to be key regulators of the lipolytic machinery, protecting or exposing the TAG core of the droplet to lipases. The neuroendocrine control of lipolysis is prototypically exerted by catecholaminergic stimulation and insulin-induced suppression, both of which affect cyclic AMP levels and hence the protein kinase A-mediated phosphorylation of HSL and perilipin. Interestingly, in recent decades adipose tissue has been shown to secrete a large number of adipokines, which exert direct effects on lipolysis, while adipocytes reportedly express a wide range of receptors for signals involved in lipid mobilisation. Recently recognised mediators of lipolysis include some adipokines, structural membrane proteins, atrial natriuretic peptides, AMP-activated protein kinase and mitogen-activated protein kinase. Lipolysis needs to be reanalysed from the broader perspective of its specific physiological or pathological context since basal or stimulated lipolytic rates occur under diverse conditions and by different mechanisms.
Collapse
|
19
|
Birsoy K, Festuccia WT, Laplante M. A comparative perspective on lipid storage in animals. J Cell Sci 2013; 126:1541-52. [PMID: 23658371 DOI: 10.1242/jcs.104992] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Lipid storage is an evolutionary conserved process that exists in all organisms from simple prokaryotes to humans. In Metazoa, long-term lipid accumulation is restricted to specialized cell types, while a dedicated tissue for lipid storage (adipose tissue) exists only in vertebrates. Excessive lipid accumulation is associated with serious health complications including insulin resistance, type 2 diabetes, cardiovascular diseases and cancer. Thus, significant advances have been made over the last decades to dissect out the molecular and cellular mechanisms involved in adipose tissue formation and maintenance. Our current understanding of adipose tissue development comes from in vitro cell culture and mouse models, as well as recent approaches to study lipid storage in genetically tractable lower organisms. This Commentary gives a comparative insight into lipid storage in uni- and multi-cellular organisms with a particular emphasis on vertebrate adipose tissue. We also highlight the molecular mechanisms and nutritional signals that regulate the formation of mammalian adipose tissue.
Collapse
Affiliation(s)
- Kivanç Birsoy
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142, USA.
| | | | | |
Collapse
|
20
|
Hydroxytyrosol stimulates lipolysis via A-kinase and extracellular signal-regulated kinase activation in 3T3-L1 adipocytes. Eur J Nutr 2013; 53:743-50. [DOI: 10.1007/s00394-013-0578-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 08/21/2013] [Indexed: 10/26/2022]
|
21
|
Yu Q, Zhang Y, Xia Y, Yang X, Li N, Ye L, Mao X. Analysis of endothelial nitric oxide synthase (eNOS) G894T polymorphism and semen parameters in a Chinese Han population. Andrologia 2013; 46:541-6. [PMID: 23710540 DOI: 10.1111/and.12113] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2013] [Indexed: 11/30/2022] Open
Abstract
Previous studies have shown that endothelial nitric oxide synthase (eNOS) gene may be involved in abnormal semen parameters. However, the relationship between eNOS G894T polymorphism and semen parameters remains controversial. The purpose of this study was to investigate the association of eNOS G894T polymorphism and semen parameters. The genotype frequency of eNOS G894T was determined in 270 idiopathic asthenozoospermia patients and 248 ethnically matched healthy volunteers using iPLEX genotyping assays on a MassARRAY(®) (Sequenom, San Diego, CA, USA) platform. The statistical analysis performed with Fisher's exact test showed no significant difference in frequencies of genotypes between both groups. The logistic regression showed that genotypes GT, TT and allele T were nonassociated with increased risk of asthenozoospermia in the patient group with ≤5% or >5% sperm with normal forms. The dependence on genotypes of semen parameters was further investigated in both patients and control group. There was no significant difference as compared to control group (P > 0.05). Our study indicated that eNOS gene G894T polymorphism may not have an adverse effect on semen parameters in a Chinese Han population.
Collapse
Affiliation(s)
- Q Yu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | |
Collapse
|
22
|
Hemmrich K, Paul NE, Pallua N. The nitric oxide system--cure for shortcomings in adipose tissue engineering? J Tissue Eng Regen Med 2012; 6 Suppl 3:S1-3. [PMID: 22888023 DOI: 10.1002/term.1541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Revised: 03/04/2012] [Accepted: 04/18/2012] [Indexed: 11/07/2022]
Abstract
Adipose tissue engineering aims to grow fat tissue for soft tissue reconstruction after tumour resection or trauma. However, insufficient progenitor cell differentiation and poor vascularization compromise the generation of clinically applicable adipose tissue. The desired process of neo-adipogenesis seems to be difficult to mimic, even though it takes place in all of us, inevitably and rapidly, as soon as we start consuming high-caloric diets. It has previously been proposed that inflammation and its key regulator, nitric oxide (NO), may play a relevant part in neo-adipogenesis. We here discuss how a controlled activation of the nitric oxide system on various levels may represent a cure for several current shortcomings in adipose tissue engineering.
Collapse
Affiliation(s)
- Karsten Hemmrich
- Department of Plastic Surgery and Hand Surgery, Burn Centre, University Hospital of RWTH Aachen University of Technology, Aachen, Germany.
| | | | | |
Collapse
|
23
|
Joffin N, Niang F, Forest C, Jaubert AM. Is there NO help for leptin? Biochimie 2012; 94:2104-10. [PMID: 22750650 DOI: 10.1016/j.biochi.2012.06.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 06/15/2012] [Indexed: 01/14/2023]
Abstract
Since the initial identification of leptin as the product of the ob gene in 1994, the signaling pathways by which this hormone alters cell physiology have been the subject of extensive investigations. The fact that leptin can induce nitric oxide (NO) production was first demonstrated in studies of the pituitary gland and pancreatic islets. A large number of additional studies further showed that this adipokine stimulates NO synthesis in multiple tissues. This review article discusses the role of leptin in NO production and its pathophysiological consequences. The role of this gaseous messenger in cell physiology depends on the cell type, the concentration of NO and the duration of exposure. It can be either a potent oxidant or a protector of cell integrity against the formation of reactive oxygen species. Leptin plays two opposing roles on arterial pressure. It exerts a hypertensive effect due to sympathetic activation and a vasorelaxant effect due to NO production. This adipokine acts via NO to produce pro-inflammatory factors in cartilage pathology, potentially contributing to an increased risk for osteoarthritis. Another well-documented role of leptin-induced NO, acting either directly or via the hypothalamus, concerns lipid metabolism in muscle and adipose tissue. In adipocytes, the direct and rapid action of leptin is to activate the nitric oxide synthase III, which favors lipolysis. In contrast, in the long-term, leptin reduces lipolysis. However, both in the short-term and in the long-term, glyceroneogenesis and its key enzyme, the cytosolic phosphoenolpyruvatecarboxykinase (PEPCK-C), are down-regulated by the adipokine, thus favoring fatty acid release. Hence, leptin-induced NO production plays a crucial role in fatty acid metabolism in adipose tissue. The resulting effects are to prevent lipid storage and to improve energy expenditure, with possible improvements of the obese state and its associated diseases.
Collapse
Affiliation(s)
- Nolwenn Joffin
- Institut National de la Santé et de la Recherche Médicale UMR-S 747, Université Paris Descartes, Pharmacologie Toxicologie et Signalisation Cellulaire, 45 rue des Saints Pères, 75006 Paris, France
| | | | | | | |
Collapse
|
24
|
Garcia-Diaz DF, Campion J, Quintero P, Milagro FI, Moreno-Aliaga MJ, Martinez JA. Vitamin C modulates the interaction between adipocytes and macrophages. Mol Nutr Food Res 2011; 55 Suppl 2:S257-63. [PMID: 21796779 DOI: 10.1002/mnfr.201100296] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 05/02/2011] [Accepted: 05/15/2011] [Indexed: 02/05/2023]
Abstract
SCOPE Increased adiposity is related with monocyte infiltration into the adipose tissue that accentuates inflammation. Antioxidant treatments emerge as approaches to counteract this phenomenon. METHODS AND RESULTS Cocultures of differentiated 3T3-L1 adipocytes and RAW264.7 macrophages were incubated for 24-72 h with/without 100 nM insulin and/or 200 μM vitamin C (VC). Nitric oxide (NO) secretion (24 h) was measured. Also, expression (24 h) and secretion (72 h) of MCP-1, leptin and apelin were analyzed. NO secretion was significantly inhibited by insulin and VC only in cocultures. MCP-1 expression/secretion was enhanced in cocultures. Insulin incubation reduced MCP-1 expression in both cultures and VC only in controls. Both treatments inhibited MCP-1 secretion in cocultures. Apelin gene expression was induced in cocultures. Insulin induced apelin mRNA expression, but VC inhibited its expression in cocultures under insulin treatment. Apelin secretion was notably induced by insulin and inhibited by VC in cocultures. Leptin expression was decreased in coculture, while presented no effects by VC. CONCLUSION VC importantly modulates the established pro-inflammatory state in the interaction between adipocytes and macrophages.
Collapse
Affiliation(s)
- Diego F Garcia-Diaz
- Department of Nutrition and Food Sciences, Physiology and Toxicology, University of Navarra, Pamplona, Spain
| | | | | | | | | | | |
Collapse
|
25
|
Lu M, Li P, Pferdekamper J, Fan W, Saberi M, Schenk S, Olefsky JM. Inducible nitric oxide synthase deficiency in myeloid cells does not prevent diet-induced insulin resistance. Mol Endocrinol 2010; 24:1413-22. [PMID: 20444886 DOI: 10.1210/me.2009-0462] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent findings denote an important contribution of macrophage inflammatory pathways in causing obesity-related insulin resistance. Inducible nitric oxide synthase (iNOS) is activated in proinflammatory macrophages and modestly elevated in insulin-responsive tissues. Although the benefits of systemic iNOS inhibition in insulin-resistant models have been demonstrated, the role of macrophage iNOS in metabolic disorders is not clear. In the current work, we used bone marrow transplantation (BMT) to generate mice with myeloid iNOS deficiency [iNOS BMT knockout (KO)]. Interestingly, disruption of iNOS in myeloid cells did not protect mice from high-fat diet-induced obesity and insulin resistance. When mice were treated with the iNOS inhibitor, N6-(1-Iminoethyl)-L-lysine hydrochloride (L-NIL), we observed a significant and comparable improvement of glucose homeostasis and insulin sensitivity in both wild-type and iNOS BMT KO mice. We further demonstrated that absence of iNOS in primary macrophages did not affect acute TLR4 signaling pathways and had only a modest and mixed effect on inflammatory gene expression. With respect to TNFalpha treatment, iNOS KO macrophages showed, if anything, a greater inflammatory response. In summary, we conclude that iNOS inhibition in tissues other than myeloid cells is responsible for the beneficial effects in obesity/insulin resistance.
Collapse
Affiliation(s)
- Min Lu
- Department of Medicine, University of California, San Diego, California 92093, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Van Linthout S, Foryst-Ludwig A, Spillmann F, Peng J, Feng Y, Meloni M, Van Craeyveld E, Kintscher U, Schultheiss HP, De Geest B, Tschöpe C. Impact of HDL on adipose tissue metabolism and adiponectin expression. Atherosclerosis 2010; 210:438-44. [PMID: 20202635 DOI: 10.1016/j.atherosclerosis.2010.01.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2009] [Revised: 11/26/2009] [Accepted: 01/03/2010] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The objective of the current study was to investigate the hypothesis that high-density lipoprotein (HDL) influences adipocyte metabolism and adiponectin expression. Therefore, HDL was increased in vivo via apolipoprotein (apo) A-I gene transfer and in vitro via supplementation of HDL to partly differentiated adipocytes, in the presence or absence of lipopolysaccharide (LPS), known to decrease HDL cholesterol and adiponectin levels in vivo. METHODS AND RESULTS Apo A-I transfer resulted in a significant increase of HDL cholesterol in control and LPS-injected C57BL/6 mice, which was paralleled by an increase in plasma adiponectin levels and adiponectin expression in abdominal fat. Triglyceride and free fatty acids levels after LPS administration were 2.2-fold (p<0.05) and 1.3-fold (p<0.05) lower, respectively, in Ad.hapoA-I-LPS than in Ad.Null-LPS mice. In parallel, the LPS-induced mRNA expression of hormone sensitive lipase was 3.5-fold (p=0.05) decreased in the Ad.hapoA-I-LPS group. On the other hand, apo A-I transfer abrogated the LPS-mediated reduction in lipin-1 and CD36 mRNA expression by 8.2-fold (p<0.05) and 18-fold (p<0.05), respectively. Concomitantly, the phosphorylation state of Akt was 2.0-fold (p<0.05) increased in the Ad.hapoA-I-LPS compared to the Ad.Null-LPS group. Pre-incubation of partly differentiated adipocytes with HDL (50 microg protein/ml) increased adiponectin expression by 1.5-fold under basal conditions (p<0.05) and could abrogate LPS-induced down-regulation of adiponectin, both in a phosphatidylinositol-3-kinase-dependent manner. CONCLUSIONS HDL affects adipocyte metabolism and adiponectin expression.
Collapse
Affiliation(s)
- Sophie Van Linthout
- Charité, University-Medicine Berlin, Campus Benjamin Franklin, Department of Cardiology, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Lien CC, Au LC, Tsai YL, Ho LT, Juan CC. Short-term regulation of tumor necrosis factor-alpha-induced lipolysis in 3T3-L1 adipocytes is mediated through the inducible nitric oxide synthase/nitric oxide-dependent pathway. Endocrinology 2009; 150:4892-900. [PMID: 19819972 DOI: 10.1210/en.2009-0403] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TNF-alpha has several effects on adipocytes that may be related to the development of type 2 diabetes in obese subjects. Many studies demonstrated that long-term treatment with TNF-alpha increases lipolysis in adipocytes. However, the short-term (<4 h) effects of TNF-alpha on lipolysis have not been well investigated. The aim of this study was to investigate the short-term regulatory mechanism of TNF-alpha-induced lipolysis in 3T3-L1 adipocytes. Well-differentiated 3T3-L1 adipocytes were used. Lipolysis was determined by measuring glycerol release. Expression of inducible nitric oxide (iNOS) and nitric oxide (NO) production were measured, respectively, by Western blots and the Griess reagent. A selective iNOS inhibitor (s-ethylisothiourea . HBr), an adenylyl cyclase inhibitor (SQ22536), and a guanylyl cyclase inhibitor (LY83583) were used to investigate the involvement of iNOS, cAMP, and cGMP in TNF-alpha-induced lipolysis. Transient transfection with iNOS short hairpin RNA was performed to confirm the involvement of iNOS in TNF-alpha-induced lipolysis. Phosphorylation of hormone-sensitive lipase (HSL) was measured by immunoprecipitation and Western blotting. Results showed that short-term TNF-alpha treatment significantly increased lipolysis, iNOS expression, and NO production in a time- and dose-dependent manner. Furthermore, treatment with the NO donor S-nitroso-N-acetylpenicillamine also stimulated lipolysis and HSL phosphorylation in 3T3-L1 adipocytes. Moreover, pretreatment with inhibitors of iNOS and guanylate cyclase, but not an adenylate cyclase inhibitor, abolished TNF-alpha-induced lipolysis and HSL phosphorylation. Suppression of TNF-alpha-induced iNOS expression using short hairpin RNA significantly reduced TNF-alpha-induced lipolysis. In conclusion, short-term TNF-alpha treatment induces lipolysis in 3T3-L1 adipocytes by increasing iNOS expression and NO production, which activates the guanylyl cyclase/cGMP-dependent pathway and induces phosphorylation of HSL.
Collapse
Affiliation(s)
- Chih-Chan Lien
- Department of Physiology, National Yang-Ming University, Number 155, Section 2, Li-Nong Street, Taipei, Taiwan.
| | | | | | | | | |
Collapse
|
28
|
Juan CC, Lien CC, Chang CL, Huang YH, Ho LT. Involvement of iNOS and NO in TNF-alpha-downregulated resistin gene expression in 3T3-L1 adipocytes. Obesity (Silver Spring) 2008; 16:1219-25. [PMID: 18369347 DOI: 10.1038/oby.2008.200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE In order to characterize the regulation of resistin gene expression, we explore the effect of tumornecrosis factor-alpha (TNF-alpha) on resistin mRNA expression and its underlying mechanism in 3T3-L1 adipocytes. METHODS AND PROCEDURES Differentiated 3T3-L1 adipocytes were treated for 24 h with 0-10 ng/ml of TNF-alpha or with 2.5 ng/ml of TNF-alpha for 0-24 h, and then resistin mRNA levels were measured by northern blotting. To further explore the involvement of nitric oxide (NO) in TNF-alpha-regulated resistin expression, the effect of the NO donor, sodium nitroprusside (SNP), on resistin mRNA levels in adipocytes and the effect of the nitric oxide synthase (NOS) inhibitors, N(G)-nitro-L-arginine methyl ester (L-NAME), and S,S'-1,3-phenylene-bis(1,2-ethanediyl)-bis-isothiourea.2HBr (PBITU), on the TNF-alpha effect in adipocytes were examined. The effects of TNF-alpha on inducible NOS (iNOS) protein expression in adipocytes were also measured by western blotting. RESULTS Our results showed that TNF-alpha caused a dose-dependent reduction in resistin mRNA levels. This effect seemed to be associated with the TNF-alpha-induced expression of iNOS. The results showed that TNF-alpha induced iNOS expression and release of NO after 24-h treatment of differentiated 3T3-L1 adipocytes. Pretreatment with L-NAME and PBITU significantly reversed the TNF-alpha-induced downregulation of resistin expression, while treatment with SNP mimicked the inhibitory effect of TNF-alpha on resistin expression. In addition, pretreatment with protein tyrosine kinase (PTK) inhibitors, genistein and AG-1288, prevented TNF-alpha-induced iNOS expression and subsequent resistin downregulation. DISCUSSION Our data suggest that TNF-alpha suppresses resistin expression by inducing iNOS expression, thus causing overproduction of NO, which downregulates resistin gene expression.
Collapse
Affiliation(s)
- Chi-Chang Juan
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| | | | | | | | | |
Collapse
|
29
|
Hanani M, Nissan A, Freund HR. Innervation of submucosal adipocytes in the human colon. Neurosci Lett 2007; 428:7-10. [DOI: 10.1016/j.neulet.2007.09.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2007] [Revised: 09/16/2007] [Accepted: 09/18/2007] [Indexed: 12/23/2022]
|
30
|
Unno Y, Akuta T, Sakamoto YI, Horiuchi S, Akaike T. Nitric oxide-induced downregulation of leptin production by 3T3-L1 adipocytes. Nitric Oxide 2006; 15:125-32. [PMID: 16442319 DOI: 10.1016/j.niox.2005.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2005] [Revised: 11/21/2005] [Accepted: 12/10/2005] [Indexed: 02/06/2023]
Abstract
Leptin secreted mainly by adipocytes plays an important role in insulin sensitivity in metabolic syndrome. Inducible nitric oxide synthase (iNOS) in 3T3-L1 adipocytes is induced by lipopolysaccharide (LPS) and several proinflammatory cytokines such as tumor necrosis factor-alpha and interferon-gamma (IFN-gamma). Because the role of iNOS-derived nitric oxide (NO) in adipocyte function has not been fully clarified, the question that we addressed in the present study was whether iNOS-derived NO is involved in regulation of leptin secretion by adipocytes. Incubation of 3T3-L1 adipocytes for 12h with a mixture of IFN-gamma and LPS caused not only a 55% reduction in leptin secretion and a 52% reduction in leptin mRNA, but also significant induction of iNOS at both protein and mRNA levels. Inhibition of leptin secretion that had been induced by the IFN-gamma-LPS mixture was completely nullified by NOS inhibitors such as Nomega-monomethyl-L-arginine and aminoguanidine. Treatment of adipocytes with NO donors such as an NONOate and S-nitrosoglutathione produced an effect on leptin secretion similar to that of the IFN-gamma-LPS mixture. It is likely therefore that NO mediates downregulation of leptin caused by the IFN-gamma-LPS mixture in 3T3-L1 adipocytes, which suggests an important role for NO in adipocyte functions.
Collapse
Affiliation(s)
- Yuka Unno
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | | | | | | | | |
Collapse
|
31
|
Linscheid P, Seboek D, Zulewski H, Scherberich A, Blau N, Keller U, Müller B. Cytokine-induced metabolic effects in human adipocytes are independent of endogenous nitric oxide. Am J Physiol Endocrinol Metab 2006; 290:E1068-77. [PMID: 16380391 DOI: 10.1152/ajpendo.00374.2005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Nitric oxide (NO) has been recognized as a potential mediator of inflammation-induced metabolic alterations, including insulin resistance. However, expression mechanisms and potential roles of endothelial and inducible NO synthases (eNOS and iNOS, respectively) in human adipocytes are poorly understood. In the present study, we aimed to analyze several aspects of NO-related gene expression and metabolite synthesis in basal and inflammation-activated human adipocyte models. eNOS mRNA was highly expressed in omental and to a lesser extent in human subcutaneous adipose tissue biopsies, but not in purified adipocytes, in mesenchymal stem cell (MSC)- and in preadipocyte-derived adipocytes, respectively. Trace amounts of iNOS mRNA were detected in adipose tissue samples of donors with abdominal infection, as opposed to noninfected subjects. Interferon-gamma, in combination with interleukin-1beta or lipopolysaccharide, evoked a transient (4 h < time < 24 h) iNOS mRNA expression in human MSC and preadipocyte-derived adipocytes, respectively. This induction was preceded by cytokine-specific mRNAs. In addition, it was accompanied by an activation of the tetrahydrobiopterin synthesis pathway and by inhibition of peroxisome proliferator-activated receptor-gamma2. In contrast to murine 3T3-L1-derived adipocytes, iNOS protein and NO oxidation products remained undetectable in iNOS mRNA-positive human adipocytes. Accordingly, coadministration of NOS inhibitors (i.e., Nomega-nitro-L-arginine methyl ester, Nomega-monomethyl-L-arginine, and 1400W) had no effects on insulin-mediated glucose uptake and lipolysis. We conclude that, in human adipocytes, endogenous NO is not involved in metabolic regulation during either basal or cytokine-activated conditions.
Collapse
Affiliation(s)
- Philippe Linscheid
- Dept. of Research, Univ. Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
32
|
Wessells H, Teal TH, Luttrell IP, Sullivan CJ. Effect of endothelial cell-based iNOS gene transfer on cavernosal eNOS expression and mouse erectile responses. Int J Impot Res 2006; 18:438-45. [PMID: 16554854 DOI: 10.1038/sj.ijir.3901464] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Inducible nitric oxide synthase (iNOS) gene transfer is reported to augment erectile responses in rats, although it is also shown to impair vasorelaxation in cerebral arteries. We investigated the effect of endothelial cell-based iNOS gene transfer on endothelial NOS (eNOS) expression and mouse erectile responses. Human coronary artery endothelial cells (EC) transduced with empty vector (control) or iNOS were grown in culture and transplanted into the corpus cavernosum of severe combined immunodeficient mice. Endothelial NOS expression was compared in control and iNOS-transduced cells grown in the presence or absence of a selective iNOS inhibitor, L-N6- (1-iminoethyl) lysine hydrochloride (L-NIL). At 3-5 days after cell transplantation, we recorded intracorporal pressure (ICP) responses to cavernosal nerve stimulation and measured cavernosal total NO and eNOS protein expression. In this study, EC transduced with iNOS produced significantly more NO than controls but exhibited a twofold downregulation of eNOS protein and mRNA. This effect was reversed by L-NIL. In vivo, the cell-based gene transfer of iNOS led to significantly increased ICP responses, compared to mice transplanted with control ECs. Consistent with the in vitro data, cavernosal lysates had significantly reduced eNOS expression. In conclusion, EC gene transfer of iNOS downregulates EC expression of eNOS by an NOS-dependent mechanism. In the cavernosum of mice transplanted with Inos-transduced EC, nerve-stimulated erectile responses were augmented by the short-term gene transfer. However, our findings suggest that iNOS gene transfer may have deleterious effects on endothelial function if used as a treatment for erectile dysfunction.
Collapse
Affiliation(s)
- H Wessells
- Department of Urology, University of Washington School of Medicine and Harborview Medical Center, Seattle, WA, USA.
| | | | | | | |
Collapse
|
33
|
Canová NK, Lincová D, Kmonícková E, Kameníková L, Farghali H. Nitric oxide production from rat adipocytes is modulated by beta3-adrenergic receptor agonists and is involved in a cyclic AMP-dependent lipolysis in adipocytes. Nitric Oxide 2006; 14:200-11. [PMID: 16413212 DOI: 10.1016/j.niox.2005.06.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2005] [Revised: 06/16/2005] [Accepted: 06/23/2005] [Indexed: 11/19/2022]
Abstract
It is established that the modulation of beta(3)-adrenoceptor function could be associated with impairment of lipolysis in white fat and be responsible for disturbed lipid metabolism. Though two isoforms of nitric oxide synthase (NOS) were reported in adipocytes, the role of nitric oxide (NO) in adipose tissue is still ambiguous. The present work was directed to study the interplay between NO production and beta-adrenoceptor/cyclic AMP (cAMP) pathway on lipid mobilization (glycerol and nonesterified fatty acids, NEFA) in cultures of rat adipocytes isolated from epididymal white adipose tissue. beta-Nonselective (isoprenaline) and beta(3)-selective (BRL-37344) agonists and the postadrenoceptor agents such as dibutyryl-cAMP, forskolin, and 3-isobutyl-1-methylxanthine significantly increased nitrite, glycerol, and NEFA levels with BRL-37344 being the most potent. Conversely, addition of beta-nonselective (propranolol) or beta(3)-selective (bupranolol) antagonist or the adenylyl cyclase inhibitor (SQ 22,536) significantly reduced beta-agonist-induced NO production and lipolysis. For beta-adrenoceptor agonists, antagonists, and their pairs, there was a positive correlation between medium nitrite and glycerol or NEFA with r(2) being 0.90 and 0.84, respectively. The possible relationship between NO and lipolysis was revealed after adipocyte treatment with nonspecific (N(omega)-nitro-l-arginine methyl ester, l-NAME) and specific (aminoguanidine) NOS inhibitors. Both l-NAME and aminoguanidine significantly inhibited the lipolytic effect of BRL-37344. Moreover, NO-donor (S-nitroso-N-acetylpenicillamine) at higher concentration increased basal glycerol and NEFA levels. 8-bromo-cyclic GMP had no effect on adipocyte lipolysis. These data suggest that beta-adrenergic lipolysis, specifically beta(3)-adrenoceptor effect, which is realized via the adenylyl cyclase/cAMP/protein kinase A signaling cascade, involves NO production downstream of beta(3)-adrenoceptor/cAMP pathway.
Collapse
Affiliation(s)
- Nikolina Kutinová Canová
- Institute of Pharmacology, 1st Faculty of Medicine, Charles University, Albertov 4, 128 00 Prague 2, Czech Republic
| | | | | | | | | |
Collapse
|