51
|
Montgomery MK, De Nardo W, Watt MJ. Impact of Lipotoxicity on Tissue "Cross Talk" and Metabolic Regulation. Physiology (Bethesda) 2019; 34:134-149. [PMID: 30724128 DOI: 10.1152/physiol.00037.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Obesity-associated comorbidities include non-alcoholic fatty liver disease, Type 2 diabetes, and cardiovascular disease. These diseases are associated with accumulation of lipids in non-adipose tissues, which can impact many intracellular cellular signaling pathways and functions that have been broadly defined as "lipotoxic." This review moves beyond understanding intracellular lipotoxic outcomes and outlines the consequences of lipotoxicity on protein secretion and inter-tissue "cross talk," and the impact this exerts on systemic metabolism.
Collapse
Affiliation(s)
| | - William De Nardo
- Department of Physiology, The University of Melbourne , Melbourne, Victoria , Australia
| | - Matthew J Watt
- Department of Physiology, The University of Melbourne , Melbourne, Victoria , Australia
| |
Collapse
|
52
|
Crespo-Facorro B, Prieto C, Sainz J. Altered gene expression in antipsychotic-induced weight gain. NPJ SCHIZOPHRENIA 2019; 5:7. [PMID: 30971689 PMCID: PMC6458173 DOI: 10.1038/s41537-019-0075-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 03/08/2019] [Indexed: 12/17/2022]
Abstract
Antipsychotic drugs are one of the largest types of prescribed drugs. However, antipsychotic-induced weight gain (AIWG) is a major problem for the patients. AIWG increases cardiovascular and cerebrovascular morbidity and mortality, and reduces quality of life and drug compliance. To characterize changes in gene expression related to AIWG, we sequenced total messenger RNA from the blood samples of two groups of schizophrenia patients before and after 3 months of treatment with antipsychotics. The "weight gain" group was defined by an increase of body mass index (BMI) >1.5 points (18 patients; median BMI increase = 2.69) and the "no weight gain" group was defined by a change of BMI between <1.0 and >-1.0 points (18 patients; median BMI increase = 0.26). We found 115 genes with significant differential expression in the weight gain group before and after medication and 156 in the no weight gain group before and after medication. The weight gain group was significantly enriched with genes related to "obesity" and "BMI" (Fisher; p = 0.0002 and 0.01, respectively) according to the Gene Reference into Function (GeneRIF) database. In the no weight gain group, the enrichment was much smaller (Fisher; p = 0.02 and 0.79). This study is a first step toward detecting genetic factors that cause AIWG and to generating prediction tests in future studies with larger data sets.
Collapse
Affiliation(s)
- Benedicto Crespo-Facorro
- Department of Psychiatry, School of Medicine, University Hospital Marqués de Valdecilla, IDIVAL, HU Virgen del Rocio-IBIS-Universidad de Sevilla, University of Cantabria, Santander, Spain. .,CIBERSAM - Centro Investigación Biomédica en Red Salud Mental, Santander, Spain.
| | - Carlos Prieto
- Bioinformatics Service, Nucleus, University of Salamanca (USAL), Salamanca, Spain
| | - Jesus Sainz
- Spanish National Research Council (CSIC), Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC), Santander, Spain.
| |
Collapse
|
53
|
Relationship between the Degrees of Itch and Serum Lipocalin-2 Levels in Patients with Psoriasis. J Immunol Res 2019; 2019:8171373. [PMID: 30805373 PMCID: PMC6360588 DOI: 10.1155/2019/8171373] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 11/29/2018] [Indexed: 11/18/2022] Open
Abstract
Background Lipocalin-2 (LCN2), a protein secreted mainly by activated neutrophils, has been associated with neurodegeneration, obesity, and inflammatory responses. Serum LCN2 concentration has been reported elevated in patients with psoriasis, but lower in patients with atopic dermatitis (AD). Spinal astrocyte-derived LCN2 was found to be involved in enhancement of itch in a mouse model of AD. However, the relationship between LCN2 and itch in patients with psoriasis has not been determined. Objective. This study examined the correlation between serum LCN2 levels and the degrees of itch in patients with psoriasis. Methods Serum LCN2 concentrations were measured by enzyme-linked immunosorbent assays (ELISA) in patients with psoriasis and AD and in healthy controls. The degree of itch was assessed using a visual analog scale (VAS), and disease severity was determined by measuring psoriasis area and severity index (PASI) and scoring atopic dermatitis (SCORAD). Correlations among serum LCN2 level, VAS, PASI, and SCORAD were analyzed statistically. We further examined the serum LCN levels in psoriasis patients before and after biological treatment. Results Serum LCN2 concentrations were significantly higher in patients with psoriasis and AD than those in healthy controls. In patients with psoriasis, serum LCN2 concentrations were significantly correlated with VAS, but not with PASI. In contrast, serum LCN2 concentrations did not correlate with VAS or SCORAD in patients with AD. Serum LCN2 levels in psoriasis patients significantly decreased after the biological treatment along with improvement of VAS. Conclusion Serum LCN2 concentration is associated with the degree of itch in patients with psoriasis, suggesting that serum LCN2 may be a useful clinical marker for itch in psoriasis.
Collapse
|
54
|
Feraco A, Armani A, Urbanet R, Nguyen Dinh Cat A, Marzolla V, Jaisser F, Caprio M. Minor role of mature adipocyte mineralocorticoid receptor in high-fat diet-induced obesity. J Endocrinol 2018; 239:229-240. [PMID: 30121579 DOI: 10.1530/joe-18-0314] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 08/07/2018] [Accepted: 08/15/2018] [Indexed: 01/07/2023]
Abstract
Obesity is a major risk factor that contributes to the development of cardiovascular disease and type 2 diabetes. Mineralocorticoid receptor (MR) expression is increased in the adipose tissue of obese patients and several studies provide evidence that MR pharmacological antagonism improves glucose metabolism in genetic and diet-induced mouse models of obesity. In order to investigate whether the lack of adipocyte MR is sufficient to explain these beneficial metabolic effects, we generated a mouse model with inducible adipocyte-specific deletion of Nr3c2 gene encoding MR (adipo-MRKO). We observed a significant, yet not complete, reduction of Nr3c2 transcript and MR protein expression in subcutaneous and visceral adipose depots of adipo-MRKO mice. Notably, only mature adipocyte fraction lacks MR, whereas the stromal vascular fraction maintains normal MR expression in our mouse model. Adipo-MRKO mice fed a 45% high-fat diet for 14 weeks did not show any significant difference in body weight and fat mass compared to control littermates. Glucose and insulin tolerance tests revealed that mature adipocyte MR deficiency did not improve insulin sensitivity in response to a metabolic homeostatic challenge. Accordingly, no significant changes were observed in gene expression profile of adipogenic and inflammatory markers in adipose tissue of adipo-MRKO mice. Moreover, pharmacological MR antagonism in mature primary murine adipocytes, which differentiated ex vivo from WT mice, did not display any effect on adipokine expression. Taken together, these data demonstrate that the depletion of MR in mature adipocytes displays a minor role in diet-induced obesity and metabolic dysfunctions.
Collapse
Affiliation(s)
- A Feraco
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
- INSERM, UMR_S 1138, Teams 1, Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France
| | - A Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - R Urbanet
- INSERM, UMR_S 1138, Teams 1, Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France
| | - A Nguyen Dinh Cat
- INSERM, UMR_S 1138, Teams 1, Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France
| | - V Marzolla
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - F Jaisser
- INSERM, UMR_S 1138, Teams 1, Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France
| | - M Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy
| |
Collapse
|
55
|
Pancreatic Cancer and Obesity: Molecular Mechanisms of Cell Transformation and Chemoresistance. Int J Mol Sci 2018; 19:ijms19113331. [PMID: 30366466 PMCID: PMC6274743 DOI: 10.3390/ijms19113331] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/27/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022] Open
Abstract
Cancer and obesity are the two major epidemics of the 21st century. Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of death, with a five-year overall survival rate of only 8%. Its incidence and mortality have increased in recent years, and this cancer type is expected to be among the top five leading causes of cancer-related death by 2030 in the United States (US). In the last three decades, the prevalence of overweight people has boosted with a consequent increase in obesity-related diseases. Considerable epidemiologic evidence correlates overweight and obese conditions to an increased risk of several types of cancer, including PDAC. Besides being a risk factor for multiple metabolic disorders, the tumor-promoting effects of obesity occur at the local level via inflammatory mediators that are associated with adipose inflammation and metabolic or hormones mediators and microbiota dysbiosis. Although an excess of body mass index (BMI) represents the second most modifiable risk factor for PDAC with an increased cancer related-death of more than 20–40%, still little is known about the molecular mechanisms that underlie this strong association. In this review, we focused on the role of obesity as a preventable risk factor of PDAC, discussing the molecular mechanisms linking obesity to cancer initiation and progression. Moreover, we highlighted the role of obesity in defining chemoresistance, showing how a high BMI can actually reduce response to chemotherapy.
Collapse
|
56
|
Abstract
At the simplest level, obesity is the manifestation of an imbalance between caloric intake and expenditure; however, the pathophysiological mechanisms that govern the development of obesity and associated complications are enormously complex. Fibrosis within the adipose tissue compartment is one such factor that may influence the development of obesity and/or obesity-related comorbidities. Furthermore, the functional consequences of adipose tissue fibrosis are a matter of considerable debate, with evidence that fibrosis serves both adaptive and maladaptive roles. Tissue fibrosis itself is incompletely understood, and multiple cellular and molecular pathways are involved in the development, maintenance, and resolution of the fibrotic state. Within the context of obesity, fibrosis influences molecular and cellular events that relate to adipocytes, inflammatory cells, inflammatory mediators, and supporting adipose stromal tissue. In this Review, we explore what is known about the interplay between the development of adipose tissue fibrosis and obesity, with a view toward future investigative and therapeutic avenues.
Collapse
Affiliation(s)
| | - Michael J Podolsky
- Cardiovascular Research Institute.,Lung Biology Center, and.,Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Kamran Atabai
- Cardiovascular Research Institute.,Lung Biology Center, and.,Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
57
|
Stephens JM, Bailey JL, Hang H, Rittell V, Dietrich MA, Mynatt RL, Elks CM. Adipose Tissue Dysfunction Occurs Independently of Obesity in Adipocyte-Specific Oncostatin Receptor Knockout Mice. Obesity (Silver Spring) 2018; 26:1439-1447. [PMID: 30226002 PMCID: PMC6146404 DOI: 10.1002/oby.22254] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 06/20/2018] [Accepted: 06/23/2018] [Indexed: 01/21/2023]
Abstract
OBJECTIVE This study examined the phenotypic effects of adipocyte-specific oncostatin M receptor (OSMR) loss in chow-fed mice. METHODS Chow-fed adipocyte-specific OSMR knockout (FKO) mice and littermate OSMRfl/fl controls were studied. Tissue weights, insulin sensitivity, adipokine production, and stromal cell immunophenotypes were assessed in epididymal fat (eWAT); serum adipokine production was also assessed. In vitro, adipocytes were treated with oncostatin M, and adipokine gene expression was assessed. RESULTS Body weights, fasting blood glucose levels, and eWAT weights did not differ between genotypes. However, the eWAT of OSMRFKO mice was modestly less responsive to insulin stimulation than that of OSMRfl/fl mice. Notably, significant increases in adipokines, including C-reactive protein, lipocalin 2, intercellular adhesion molecule-1, and insulinlike growth factor binding protein 6, were observed in the eWAT of OSMRFKO mice. In addition, significant increases in fetuin A and intercellular adhesion molecule-1 were detected in OSMRFKO serum. Flow cytometry revealed a significant increase in leukocyte number and modest, but not statistically significant, increases in B cells and T cells in the eWAT of OSMRFKO mice. CONCLUSIONS The chow-fed OSMRFKO mice exhibited adipose tissue dysfunction and increased proinflammatory adipokine production. These results suggest that intact adipocyte oncostatin M-OSMR signaling is necessary for adipose tissue immune cell homeostasis.
Collapse
Affiliation(s)
- Jacqueline M. Stephens
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808
| | - Jennifer L. Bailey
- Matrix Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808
| | - Hardy Hang
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808
| | - Victoria Rittell
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808
- Matrix Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808
| | - Marilyn A. Dietrich
- Cell Biology and Bioimaging Core, Pennington Biomedical Research Center, Baton Rouge, LA, 70808
| | - Randall L. Mynatt
- Transgenics Core, Pennington Biomedical Research Center, Baton Rouge, LA, 70808
| | - Carrie M. Elks
- Matrix Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808
- Corresponding Author: Carrie M. Elks, PhD, RD, Matrix Biology Laboratory, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA, Phone: (225) 763-3140,
| |
Collapse
|
58
|
Unamuno X, Gómez-Ambrosi J, Rodríguez A, Becerril S, Frühbeck G, Catalán V. Adipokine dysregulation and adipose tissue inflammation in human obesity. Eur J Clin Invest 2018; 48:e12997. [PMID: 29995306 DOI: 10.1111/eci.12997] [Citation(s) in RCA: 367] [Impact Index Per Article: 61.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 07/10/2018] [Indexed: 12/13/2022]
Abstract
Obesity, a worldwide epidemic, confers increased risk for multiple serious conditions, including type 2 diabetes, cardiovascular diseases, nonalcoholic fatty liver disease and cancer. Adipose tissue is considered one of the largest endocrine organs in the body as well as an active tissue for cellular reactions and metabolic homeostasis rather than an inert tissue for energy storage. The functional pleiotropism of adipose tissue relies on its ability to synthesize and release a large number of hormones, cytokines, extracellular matrix proteins and growth and vasoactive factors, collectively termed adipokines that influence a variety of physiological and pathophysiological processes. In the obese state, excessive visceral fat accumulation causes adipose tissue dysfunctionality that strongly contributes to the onset of obesity-related comorbidities. The mechanisms underlying adipose tissue dysfunction include adipocyte hypertrophy and hyperplasia, increased inflammation, impaired extracellular matrix remodelling and fibrosis together with an altered secretion of adipokines. This review describes how adipose tissue becomes inflamed in obesity and summarizes key players and molecular mechanisms involved in adipose inflammation.
Collapse
Affiliation(s)
- Xabier Unamuno
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.,Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
59
|
Amersfoort J, Schaftenaar FH, Douna H, van Santbrink PJ, Kröner MJ, van Puijvelde GHM, Quax PHA, Kuiper J, Bot I. Lipocalin-2 contributes to experimental atherosclerosis in a stage-dependent manner. Atherosclerosis 2018; 275:214-224. [PMID: 29960897 DOI: 10.1016/j.atherosclerosis.2018.06.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 05/15/2018] [Accepted: 06/08/2018] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND AIMS Lipocalin-2 (Lcn2) is a glycoprotein which can be secreted by immune cells. Several studies in humans have suggested Lcn2 can be used as a biomarker for the detection of unstable atherosclerotic lesions, partly as it is known to interact with MMP-9. METHODS In this study, we generated Ldlr-/-Lcn2-/- mice to assess the functional role of Lcn2 in different stages of atherosclerosis. Atherosclerotic lesions were characterized through histological analysis and myeloid cell populations were examined using flow cytometry. RESULTS We show that Ldlr-/-Lcn2-/- mice developed larger atherosclerotic lesions during earlier stages of atherosclerosis and had increased circulating Ly6Chi inflammatory monocytes compared to Ldlr-/- mice. Advanced atherosclerotic lesions from Ldlr-/-Lcn2-/- mice had decreased necrotic core area, suggesting Lcn2 deficiency may affect lesion stability. Furthermore, MMP-9 activity was diminished in plaques from Ldlr-/-Lcn2-/- mice. CONCLUSIONS Altogether, these findings suggest that Lcn2 deficiency promotes lesion growth in earlier stages of the disease while it decreases MMP-9 activity and necrotic core size in advanced atherosclerosis.
Collapse
Affiliation(s)
- J Amersfoort
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands.
| | - F H Schaftenaar
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - H Douna
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - P J van Santbrink
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - M J Kröner
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - G H M van Puijvelde
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - P H A Quax
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333ZA, Leiden, The Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - J Kuiper
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - I Bot
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| |
Collapse
|
60
|
Kuten O, Simon M, Hornyák I, De Luna-Preitschopf A, Nehrer S, Lacza Z. The Effects of Hyperacute Serum on Adipogenesis and Cell Proliferation of Mesenchymal Stromal Cells. Tissue Eng Part A 2018; 24:1011-1021. [PMID: 29265000 DOI: 10.1089/ten.tea.2017.0384] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fat tissue, due to its high concentration of stem cells, has a role in aesthetic medicine and reconstructive surgery. However, poor survival of the transplanted cells still limits the usefulness of this material in regenerative medicine. Several studies indicated that platelet-rich plasma (PRP) may improve adipose tissue viability due to its growth factor content. This study aimed at investigating the effects of PRP and hyperacute serum (HAS) on the adipogenic lineage in vitro. PRP was prepared by using two centrifugation steps in the presence of anticoagulants, and HAS was isolated from activated platelet-rich fibrin within 10 min of blood drawing to prevent the propagation of inflammatory cascades. Metabolic activity and proliferation rate of human bone marrow-derived mesenchymal stem cells (hMSCs) cultivated in media supplemented with three types of serum additives (fetal calf serum [FCS], human PRP, or HAS) was determined by using a tetrazolium assay. Adipogenesis was evaluated in standard and pro-adipogenic media and tested by oil red staining, triglyceride content, and expression of specific genes. Adipogenic regulators in the sera were measured by multiplex ELISA assays. We observed that proliferation of hMSCs was supported by both FCS and HAS in a time-dependent manner, but surprisingly, PRP had a much weaker effect (change in proliferation rate after 5 days relative to metabolic activity on day 0-FCS: 5.4-fold change, HAS: 5.8-fold change, serum free 1.9-fold change, PRP: 3.0-fold change, p < 0.05). Lipogenesis was only observed in groups with adipogenic differentiation medium, with HAS showing a significantly stronger effect than PRP. This was confirmed by intensive accumulation of lysochrome dye in lipid droplets, higher triglyceride concentration, and elevated expression of specific adipogenic genes. Measurement of lipogenic proteins in the sera revealed that both PRP and HAS are abundant in them; however, PRP also contains anti-adipogenic factors, which explains its weaker and less reliable effect. The results of this study suggest that HAS provides more robust support than PRP in hMSCs proliferation as well as lipogenic differentiation, indicating that it may be a better adjuvant in fat grafting procedures.
Collapse
Affiliation(s)
- Olga Kuten
- 1 OrthoSera GmbH , Krems an der Donau, Austria
| | - Melinda Simon
- 2 Institute of Clinical Experimental Research, Semmelweis University , Budapest, Hungary
| | - István Hornyák
- 2 Institute of Clinical Experimental Research, Semmelweis University , Budapest, Hungary
| | | | - Stefan Nehrer
- 3 Centre for Regenerative Medicine and Orthopedics, Danube University Krems , Krems, Austria
| | | |
Collapse
|
61
|
Szczęsny W, Kuligowska-Prusińska M, Dąbrowiecki S, Szmytkowski J, Reśliński A, Słupski M. Activity of metalloproteinases and adiponectin in obese patients-a possible factor of incisional hernias after bariatric procedures. J Zhejiang Univ Sci B 2018; 19:65-70. [PMID: 29308609 DOI: 10.1631/jzus.b1600383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Metalloproteinases are a key component of the pathogenesis of abdominal hernias. Obesity is considered a risk factor in herniogenesis and hernia recurrence. The aim of this study was to evaluate the serum concentrations of metalloproteinase-2 (MMP-2), MMP-9, MMP-13, and adiponectin in morbidly obese and non-overweight controls. MATERIALS AND METHODS The participants were recruited from among patients undergoing bariatric and non-bariatric surgery and divided into two groups: I (body mass index (BMI)≥35 kg/m2, n=40) and II (BMI<25 kg/m2, n=30). Serum concentrations of MMP-2, MMP-9, MMP-13, and adiponectin were measured using enzyme-linked immunosorbent assay (ELISA). RESULTS A statistically significant difference between groups was observed for MMP-2 concentration. The median MMP-9 concentration was higher in the obese group, but the difference was not statistically significant. Median MMP-13 concentrations did not differ between groups. Serum adiponectin concentration was insignificantly higher in the non-obese group. CONCLUSIONS The elevated serum MMP-2 and MMP-9 concentrations in obese individuals may be related to the higher incidence of incisional hernias in this population.
Collapse
Affiliation(s)
- Wojciech Szczęsny
- Ludwik Rydygier College of Medicine in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | | | - Stanisław Dąbrowiecki
- Ludwik Rydygier College of Medicine in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Jakub Szmytkowski
- Ludwik Rydygier College of Medicine in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Adrian Reśliński
- Ludwik Rydygier College of Medicine in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Maciej Słupski
- Ludwik Rydygier College of Medicine in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| |
Collapse
|
62
|
Dong Z, Gong H, Chen Y, Wu H, Wu J, Deng Y, Song X. LH-21, A Peripheral Cannabinoid Receptor 1 Antagonist, Exerts Favorable Metabolic Modulation Including Antihypertensive Effect in KKAy Mice by Regulating Inflammatory Cytokines and Adipokines on Adipose Tissue. Front Endocrinol (Lausanne) 2018; 9:167. [PMID: 29731737 PMCID: PMC5920035 DOI: 10.3389/fendo.2018.00167] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 03/28/2018] [Indexed: 12/21/2022] Open
Abstract
Patients with obesity are susceptible to hypertension and diabetes. Over-activation of cannabinoid receptor 1 (CB1R) in adipose tissue is proposed in the pathophysiology of metabolic disorders, which led to the metabolic dysfunction of adipose tissue and deregulated production and secretion of adipokines. In the current study, we determined the impact of LH-21, a representative peripheral CB1R antagonist, on the obesity-accompanied hypertension and explored the modulatory action of LH-21 on the adipose tissue in genetically obese and diabetic KKAy mice. 3-week LH-21 treatment significantly decreased blood pressure with a concomitant reduction in body weight, white adipose tissue (WAT) mass, and a slight loss on food intake in KKAy mice. Meanwhile, glucose handling and dyslipidemia were also markedly ameliorated after treatment. Gene expression of pro-inflammatory cytokines in WAT and the aortae were both attenuated apparently by LH-21, as well the mRNA expression of adipokines (lipocalin-2, leptin) in WAT. Concomitant amelioration on the accumulation of lipocalin-2 was observed in both WAT and aortae. In corresponding with this, serum inflammatory related cytokines (tumor necrosis factor α, IL-6, and CXCL1), and lipocalin-2 and leptin were lowered notably. Thus according to current results, it can be concluded that the peripheral CB1R antagonist LH-21 is effective in managing the obesity-accompanied hypertension in KKAy mice. These metabolic benefits are closely associated with the regulation on the production and secretion of inflammatory cytokines and adipokines in the WAT, particularly alleviated circulating lipocalin-2 and its accumulation in aortae.
Collapse
Affiliation(s)
- Ziqi Dong
- Department of Cardiology, Jinshan Hospital of FuDan University, Shanghai, China
| | - Hui Gong
- Department of Cardiology, Jinshan Hospital of FuDan University, Shanghai, China
- *Correspondence: Hui Gong, ; Xinmao Song,
| | - Yadan Chen
- Department of Pharmacy, The Second Hospital of Jilin University, Changchun, China
| | - Hong Wu
- Department of Cardiology, Jinshan Hospital of FuDan University, Shanghai, China
| | - Jun Wu
- Department of Cardiology, Jinshan Hospital of FuDan University, Shanghai, China
| | - Yinghong Deng
- Department of Cardiology, Jinshan Hospital of FuDan University, Shanghai, China
| | - Xinmao Song
- Department of Radiation Oncology, Eye, Ear, Nose & Throat, Hospital of FuDan University, Shanghai, China
- *Correspondence: Hui Gong, ; Xinmao Song,
| |
Collapse
|
63
|
Roth-Walter F, Pacios LF, Bianchini R, Jensen-Jarolim E. Linking iron-deficiency with allergy: role of molecular allergens and the microbiome. Metallomics 2017; 9:1676-1692. [PMID: 29120476 DOI: 10.1039/c7mt00241f] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Atopic individuals tend to develop a Th2 dominant immune response, resulting in hyperresponsiveness to harmless antigens, termed allergens. In the last decade, epidemiological studies have emerged that connected allergy with a deficient iron-status. Immune activation under iron-deficient conditions results in the expansion of Th2-, but not Th1 cells, can induce class-switching in B-cells and hampers the proper activation of M2, but not M1 macrophages. Moreover, many allergens, in particular with the lipocalin and lipocalin-like folds, seem to be capable of binding iron indirectly via siderophores harboring catechol moieties. The resulting locally restricted iron-deficiency may then lead during immune activation to the generation of Th2-cells and thus prepare for allergic sensitization. Moreover, iron-chelators seem to also influence clinical reactivity: mast cells accumulate iron before degranulation and seem to respond differently depending on the type of the encountered siderophore. Whereas deferoxamine triggers degranulation of connective tissue-type mast cells, catechol-based siderophores reduce activation and degranulation and improve clinical symptoms. Considering the complex interplay of iron, siderophores and immune molecules, it remains to be determined whether iron-deficiencies are the cause or the result of allergy.
Collapse
Affiliation(s)
- Franziska Roth-Walter
- Department of Comparative Medicine, at the Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University of Vienna and University of Vienna, Vienna, Austria.
| | - Luis F Pacios
- Centro de Biotecnología y Genómica de Plantas (CBGP, UPM-INIA), Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Campus de Montegancedo-UPM, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Rodolfo Bianchini
- Department of Comparative Medicine, at the Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University of Vienna and University of Vienna, Vienna, Austria.
| | - Erika Jensen-Jarolim
- Department of Comparative Medicine, at the Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University of Vienna and University of Vienna, Vienna, Austria. and Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
64
|
Sun B, Guo W, Hu S, Yao F, Yu K, Xing J, Wang R, Song H, Liao Y, Wang T, Jiang P, Han B, Deng J. Gprc5a-knockout mouse lung epithelial cells predicts ceruloplasmin, lipocalin 2 and periostin as potential biomarkers at early stages of lung tumorigenesis. Oncotarget 2017; 8:13532-13544. [PMID: 28088789 PMCID: PMC5355118 DOI: 10.18632/oncotarget.14589] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 12/31/2016] [Indexed: 02/07/2023] Open
Abstract
Lung cancer is the leading cause of cancer death. As most of lung cancer patients were diagnosed with the advanced stage, early detection is considered as the most effective strategy to reduce high mortality. Thus, it is desirable to identify specific biomarkers at early stages of lung tumorigenesis. GPRC5A is a lung tumor suppressor gene. GPRC5A deficiency is linked to lung cancer development. We hyposthesized that, dysregulated gene expression that results from Gprc5a deficiency may provide potential biomarkers at early stages of lung tumorigenesis. By analysis of top 20 upregulated genes in mouse tracheal epithelial cells (MTEC) of Gprc5a knockout (KO) vs wild-type (WT), we found that ceruloplasmin, lipocalin-2, and periostin are not only upregulated in lung epithelial cells of Gprc5a-ko mice, but also expressed at high levels in lung tumor tissues of Gprc5a-ko mice. This suggests that increased expression of these genes is associated with lung tumorigenesis. Importantly, expression of ceruloplasmin, lipocalin-2, and periostin has also been found to be significantly increased, both at mRNA and protein levels, in the lung tissues from NSCLC patients, which is correlated with repressed GPRC5A. Thus, dysregulated ceruloplasmin, lipocalin-2, and periostin may be used as potential biomarkers at early stages of lung tumorigenesis.
Collapse
Affiliation(s)
- Beibei Sun
- Translation Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wenzheng Guo
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Song Hu
- Translation Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Yao
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Keke Yu
- Department of Biobank, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Xing
- Department of Biobank, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ronghua Wang
- Translation Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hongyong Song
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yueling Liao
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tong Wang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengfei Jiang
- Department of Laboratory Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Baohui Han
- Translation Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.,Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jiong Deng
- Translation Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.,Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
65
|
Lu G, He Q, Shen Y, Cao F. Potential biomarkers for predicting hemorrhagic transformation of ischemic stroke. Int J Neurosci 2017; 128:79-89. [PMID: 28726570 DOI: 10.1080/00207454.2017.1349766] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Reperfusion therapy contributes to better clinical outcomes in patients with acute ischemic stroke but carries a more significant risk of hemorrhagic transformation (HT) compared to supportive care. Once HT occurs, the outcome is usually poor and this causes a dilemma in the treatment of ischemic stroke. Consequently, early prediction of HT would be extremely helpful for guiding precise treatment of ischemic stroke. In this review, we focus on summarizing biomarkers of HT and elucidating possible mechanisms so as to identify potential biomarkers for predicting HT.
Collapse
Affiliation(s)
- Guanfeng Lu
- a Department of Neurology, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China
| | - Quanwei He
- a Department of Neurology, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China
| | - Yan Shen
- a Department of Neurology, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China
| | - Fei Cao
- a Department of Neurology, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China
| |
Collapse
|
66
|
Acinetobacter baumannii Lipopolysaccharide Influences Adipokine Expression in 3T3-L1 Adipocytes. Mediators Inflamm 2017; 2017:9039302. [PMID: 28757686 PMCID: PMC5516741 DOI: 10.1155/2017/9039302] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 05/21/2017] [Accepted: 06/06/2017] [Indexed: 12/11/2022] Open
Abstract
Acinetobacter baumannii is one of the most important nosocomial opportunistic pathogen worldwide. In addition, obesity has been associated with an increased risk of nosocomial infection, suggesting that there may be an association between A. baumannii and white adipose tissue. However, the effects of A. baumannii on adipocytes have not been well studied at the molecular level. Here, we investigated the potential role of A. baumannii-derived lipopolysaccharides (LPS) as signaling molecules that affect adipocyte functionality. We tested the effect of increasing concentrations of A. baumannii-derived LPS (10, 100, or 1000 ng/mL) on the 3T3-L1 adipocyte cell line. Exposure to LPS was found to increase the expression of several adipokines (e.g., MIP-2, MCP-1, TNF-α, IL-6, lipocalin-2, and FABP4) in 3T3-L1 adipocytes and significantly reduced the expression of leptin and adiponectin. The effects of A. baumannii-derived LPS on MIP-2 expression were similar in comparison with that of LPS prepared from Pseudomonas aeruginosa and Escherichia coli in our cell culture-based system. This study suggests that A. baumannii-derived LPS functions as a signaling molecule that impacts the inflammatory function of white adipose tissue on the level of gene expression.
Collapse
|
67
|
Abstract
Lipocalin 2 (Lcn2), an innate immune protein, has emerged as a critical iron regulatory protein during physiological and inflammatory conditions. As a bacteriostatic factor, Lcn2 obstructs the siderophore iron-acquiring strategy of bacteria and thus inhibits bacterial growth. As part of host nutritional immunity, Lcn2 facilitates systemic, cellular, and mucosal hypoferremia during inflammation, in addition to stabilizing the siderophore-bound labile iron pool. In this review, we summarize recent advances in understanding the interaction between Lcn2 and iron, and its effects in various inflammatory diseases. Lcn2 exerts mostly a protective role in infectious and inflammatory bowel diseases, whereas both beneficial and detrimental functions have been documented in neurodegenerative diseases, metabolic syndrome, renal disorders, skin disorders, and cancer. Further animal and clinical studies are necessary to unveil the multifaceted roles of Lcn2 in iron dysregulation during inflammation and to explore its therapeutic potential for treating inflammatory diseases.
Collapse
Affiliation(s)
- Xia Xiao
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802;
| | - Beng San Yeoh
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802;
| | - Matam Vijay-Kumar
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802; .,Department of Medicine, The Pennsylvania State University Medical Center, Hershey, Pennsylvania 17033
| |
Collapse
|
68
|
Gomez-Chou SB, Swidnicka-Siergiejko AK, Badi N, Chavez-Tomar M, Lesinski GB, Bekaii-Saab T, Farren MR, Mace TA, Schmidt C, Liu Y, Deng D, Hwang RF, Zhou L, Moore T, Chatterjee D, Wang H, Leng X, Arlinghaus RB, Logsdon CD, Cruz-Monserrate Z. Lipocalin-2 Promotes Pancreatic Ductal Adenocarcinoma by Regulating Inflammation in the Tumor Microenvironment. Cancer Res 2017; 77:2647-2660. [PMID: 28249896 PMCID: PMC5441230 DOI: 10.1158/0008-5472.can-16-1986] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 09/16/2016] [Accepted: 01/10/2017] [Indexed: 12/19/2022]
Abstract
Lipocalin-2 (LCN2) promotes malignant development in many cancer types. LCN2 is upregulated in patients with pancreatic ductal adenocarcinoma (PDAC) and in obese individuals, but whether it contributes to PDAC development is unclear. In this study, we investigated the effects of Lcn2 depletion on diet-induced obesity, inflammation, and PDAC development. Mice with acinar cell-specific expression of KrasG12D were crossed with Lcn2-depleted animals and fed isocaloric diets with varying amounts of fat content. Pancreas were collected and analyzed for inflammation, pancreatic intraepithelial neoplasia (PanIN), and PDAC. We also used a syngeneic orthotopic PDAC mouse model to study tumor growth in the presence or absence of Lcn2 expression. In addition, to understand the mechanistic role of how LCN2 could be mediating PDAC, we studied LCN2 and its specific receptor solute carrier family 22 member 17 (SLC22A17) in human pancreatic cancer stellate cells (PSC), key mediators of the PDAC stroma. Depletion of Lcn2 diminished extracellular matrix deposition, immune cell infiltration, PanIN formation, and tumor growth. Notably, it also increased survival in both obesity-driven and syngeneic orthotopic PDAC mouse models. LCN2 modulated the secretion of proinflammatory cytokines in PSC of the PDAC tumor microenvironment, whereas downregulation of LCN2-specific receptor SLC22A17 blocked these effects. Our results reveal how LCN2 acts in the tumor microenvironment links obesity, inflammation, and PDAC development. Cancer Res; 77(10); 2647-60. ©2017 AACR.
Collapse
Affiliation(s)
- Sobeyda B Gomez-Chou
- Department of Cancer Biology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Agnieszka Katarzyna Swidnicka-Siergiejko
- Department of Cancer Biology, University of Texas, MD Anderson Cancer Center, Houston, Texas
- Department of Gastroenterology and Internal Medicine, University of Bialystok, Bialystok, Poland
| | - Niharika Badi
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Myrriah Chavez-Tomar
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Gregory B Lesinski
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Tanios Bekaii-Saab
- Department of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Matthew R Farren
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Thomas A Mace
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Carl Schmidt
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Yan Liu
- Department of Cancer Biology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Defeng Deng
- Department of Cancer Biology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Rosa F Hwang
- Department of Surgical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Liran Zhou
- Department of Surgical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Todd Moore
- Department of Surgical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Deyali Chatterjee
- Department of Pathology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Huamin Wang
- Department of Pathology, University of Texas, MD Anderson Cancer Center, Houston, Texas
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Xiaohong Leng
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Ralph B Arlinghaus
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Craig D Logsdon
- Department of Cancer Biology, University of Texas, MD Anderson Cancer Center, Houston, Texas.
- Department of Gastrointestinal Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Zobeida Cruz-Monserrate
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio.
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
69
|
Singh RG, Pendharkar SA, Plank LD, Petrov MS. Role of human lipocalin proteins in abdominal obesity after acute pancreatitis. Peptides 2017; 91:1-7. [PMID: 28279688 DOI: 10.1016/j.peptides.2017.03.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 03/01/2017] [Accepted: 03/06/2017] [Indexed: 02/07/2023]
Abstract
Lipocalin proteins are small regulatory peptides implicated in metabolism, inflammation, and immunity. Although lipocalin proteins have been linked to various clinical conditions, their role in the acute inflammatory setting, such as acute pancreatitis (AP), has only been sparsely investigated. Two members of the lipocalin family, lipocalin-2 (LCN-2) and retinol binding protein -4 (RBP-4), play an important role in obesity and insulin resistance. In this study, we analysed circulating levels of LCN-2 and RBP-4 in 92 individuals after AP, of whom 41 individuals had abdominal obesity and 51 did not. Binary logistic regression analyses were performed to determine whether abdominal obesity was associated with the two lipocalin proteins. Lipocalin-2 was significantly associated with abdominal obesity in the unadjusted model (Odds ratio (OR)=1.014 [95% confidence interval (CI): 1.000, 1.028], P=0.05) and after adjusting for patient related (age, ethnicity, and diabetes mellitus) and pancreatitis related (aetiology, severity, recurrence, and duration of AP) characteristics (OR=1.018 [95% CI: 1.001, 1.036], p=0.04). Further, the association of LCN-2 with waist circumference was significant in individuals with alcohol aetiology of AP (β=1.082 [95% CI: 1.011, 1.158], p=0.02]. The association between RBP-4 and abdominal obesity was not significant in both unadjusted and adjusted models. These findings indicate that circulating levels of LCN-2 in patients after AP may play a role in chronic low grade inflammation associated with abdominal adiposity and that alcohol consumption may further exacerbate adipose tissue dysfunction.
Collapse
Affiliation(s)
- Ruma G Singh
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | | | - Lindsay D Plank
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Maxim S Petrov
- Department of Surgery, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
70
|
Moschen AR, Adolph TE, Gerner RR, Wieser V, Tilg H. Lipocalin-2: A Master Mediator of Intestinal and Metabolic Inflammation. Trends Endocrinol Metab 2017; 28:388-397. [PMID: 28214071 DOI: 10.1016/j.tem.2017.01.003] [Citation(s) in RCA: 229] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/16/2017] [Accepted: 01/18/2017] [Indexed: 02/07/2023]
Abstract
Lipocalin-2 (LCN2), also known as neutrophil gelatinase-associated lipocalin (NGAL), is released by various cell types and is an attractive biomarker of inflammation, ischemia, infection, and kidney damage. Both intestinal and metabolic inflammation, as observed in obesity and related disorders, are associated with increased LCN2 synthesis. While LCN2 in the intestinal tract regulates the composition of the gut microbiota and shows anti-inflammatory activities, it also exhibits proinflammatory activities in other experimental settings. In animal models of metabolic inflammation, type 2 diabetes mellitus (T2DM), or nonalcoholic steatohepatitis (NASH), increased LCN2 expression favors inflammation via the recruitment of inflammatory cells, such as neutrophils, and the induction of proinflammatory cytokines. A better understanding of this crucial marker of innate immunity might pave the way for targeting this pathway in future therapies.
Collapse
Affiliation(s)
- Alexander R Moschen
- Department of Internal Medicine I, Gastroenterology, Hepatology & Endocrinology and Christian Doppler Laboratory for Mucosal Immunology, Medical University Innsbruck, Innsbruck, Austria
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology & Endocrinology and Christian Doppler Laboratory for Mucosal Immunology, Medical University Innsbruck, Innsbruck, Austria
| | - Romana R Gerner
- Department of Internal Medicine I, Gastroenterology, Hepatology & Endocrinology and Christian Doppler Laboratory for Mucosal Immunology, Medical University Innsbruck, Innsbruck, Austria
| | - Verena Wieser
- Department of Internal Medicine I, Gastroenterology, Hepatology & Endocrinology and Christian Doppler Laboratory for Mucosal Immunology, Medical University Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology & Endocrinology and Christian Doppler Laboratory for Mucosal Immunology, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
71
|
Pérez-Matute P, Iñiguez M, Recio-Fernández E, Oteo JA. Deciphering the molecular mechanisms involved in HIV-associated lipoatrophy by transcriptomics: a pilot study. J Physiol Biochem 2017; 73:431-443. [PMID: 28074419 DOI: 10.1007/s13105-016-0547-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 12/26/2016] [Indexed: 01/11/2023]
Abstract
HIV-associated lipoatrophy (LA) has considerable implications for risk of metabolic diseases, quality of life, and adherence to treatments. Although it has decreased in high-income countries, it is still very common in resource-limited countries. Understanding the pathophysiological mechanisms of LA can open the possibility to explore new ways to treat or prevent this condition. To identify new markers for an accurate and quick diagnosis will be also of interest. Thus, we aimed to examine functional classes of genes implicated in LA and to identify potential new markers for an accurate/quick diagnosis of LA and future complications. Eighteen participants were recruited: seven healthy volunteers, five non-LA-HIV patients, and six LA-HIV subjects. Clinical lipoatrophy was considered when changes in fat volume in the cheeks next to the nose, lateral aspect of the face, legs, arms, and buttocks were observed by the physicians. mRNA was isolated from peripheral blood mononuclear cells (PBMCs) to perform a transcriptomic and Gene Ontology analysis. To confirm RNA sequencing results, qPCRs were developed. A total of 55 genes were differentially expressed between LA and non-LA patients. Thirty-seven genes were overexpressed, whereas 18 genes were repressed. Functional analysis showed that overexpressed genes were involved in lymphocyte/neutrophil activation, inflammation, and atherogenesis. Several lymphoma markers and members of the lipocalin and aquaporin families were also found more expressed in LA patients. In contrast, most of the genes found less expressed in LA subjects were involved in angiogenesis and protection against myocardial infarction. Our results demonstrated a distinct transcriptomic signature in PBMCs of LA patients in comparison with non-LA-HIV subjects and, therefore, provided novel insights to the pathogenesis of HIV-associated lipoatrophy. Our study also highlights the potential usage of some of these genes as early markers of future complications.
Collapse
Affiliation(s)
- Patricia Pérez-Matute
- HIV and Associated Metabolic Alterations Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR)-Hospital San Pedro, Piqueras 98, 26006, Logroño, Spain.
| | - María Iñiguez
- Genomics Core Facility, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006, Logroño, Spain
| | - Emma Recio-Fernández
- HIV and Associated Metabolic Alterations Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR)-Hospital San Pedro, Piqueras 98, 26006, Logroño, Spain
| | - José-Antonio Oteo
- HIV and Associated Metabolic Alterations Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR)-Hospital San Pedro, Piqueras 98, 26006, Logroño, Spain
| |
Collapse
|
72
|
Testosterone Deficiency Induces Changes of the Transcriptomes of Visceral Adipose Tissue in Miniature Pigs Fed a High-Fat and High-Cholesterol Diet. Int J Mol Sci 2016; 17:ijms17122125. [PMID: 27999286 PMCID: PMC5187925 DOI: 10.3390/ijms17122125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/08/2016] [Accepted: 12/12/2016] [Indexed: 12/14/2022] Open
Abstract
Testosterone deficiency causes fat deposition, particularly in visceral fat, and its replacement might reverse fat accumulation, however, the underlying mechanisms of such processes under diet-induced adiposity are largely unknown. To gain insights into the genome-wide role of androgen on visceral adipose tissue (VAT), RNA-Seq was used to investigate testosterone deficiency induced changes of VAT in miniature pigs fed a high-fat and high-cholesterol (HFC) diet among intact male pigs (IM), castrated male pigs (CM), and castrated male pigs with testosterone replacement (CMT) treatments. The results showed that testosterone deficiency significantly increased VAT deposition and serum leptin concentrations. Moreover, a total of 1732 differentially expressed genes (DEGs) were identified between any two groups. Compared with gene expression profiles in IM and CMT pigs, upregulated genes in CM pigs, i.e., LOC100520753 (CD68), LCN2, EMR1, S100A9, NCF1 (p47phox), and LEP, were mainly involved in inflammatory response, oxidation-reduction process, and lipid metabolic process, while downregulated genes in CM pigs, i.e., ABHD5, SPP1, and GAS6, were focused on cell differentiation and cell adhesion. Taken together, our study demonstrates that testosterone deficiency alters the expression of numerous genes involved in key biological processes of VAT accumulation under HFC diet and provides a novel genome-wide view on the role of androgen on VAT deposition under HFC diet, thus improving our understanding of the molecular mechanisms involved in VAT changes induced by testosterone deficiency.
Collapse
|
73
|
Ritter AMV, de Faria AP, Barbaro N, Sabbatini AR, Corrêa NB, Brunelli V, Amorim R, Modolo R, Moreno H. Crosstalk between obesity and MMP-9 in cardiac remodelling –a cross-sectional study in apparent treatment-resistant hypertension. Blood Press 2016; 26:122-129. [DOI: 10.1080/08037051.2016.1249336] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Alessandra Mileni Versuti Ritter
- Laboratory of Pharmacology Cardiovascular, Faculty of Medical Sciences, Department of Pharmacology, University of Campinas, Campinas, SP, Brazil
| | - Ana Paula de Faria
- Laboratory of Pharmacology Cardiovascular, Faculty of Medical Sciences, Department of Pharmacology, University of Campinas, Campinas, SP, Brazil
| | - Natália Barbaro
- Laboratory of Pharmacology Cardiovascular, Faculty of Medical Sciences, Department of Pharmacology, University of Campinas, Campinas, SP, Brazil
| | - Andréa Rodrigues Sabbatini
- Laboratory of Pharmacology Cardiovascular, Faculty of Medical Sciences, Department of Pharmacology, University of Campinas, Campinas, SP, Brazil
| | - Nathália Batista Corrêa
- Laboratory of Pharmacology Cardiovascular, Faculty of Medical Sciences, Department of Pharmacology, University of Campinas, Campinas, SP, Brazil
| | - Veridiana Brunelli
- Laboratory of Pharmacology Cardiovascular, Faculty of Medical Sciences, Department of Pharmacology, University of Campinas, Campinas, SP, Brazil
| | - Rivadavio Amorim
- Laboratory of Neuromodulation & Center for Clinical Research Learning, Department of Physical Medicine and Rehabilitation (PM&R), Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA
| | - Rodrigo Modolo
- Laboratory of Pharmacology Cardiovascular, Faculty of Medical Sciences, Department of Pharmacology, University of Campinas, Campinas, SP, Brazil
- Laboratory of Pharmacology Cardiovascular, Faculty of Medical Sciences, Department of Internal Medicine, University of Campinas, SP, Brazil
| | - Heitor Moreno
- Laboratory of Pharmacology Cardiovascular, Faculty of Medical Sciences, Department of Internal Medicine, University of Campinas, SP, Brazil
| |
Collapse
|
74
|
Prist IH, Salles AG, de Lima TM, Modolin MLA, Gemperli R, Souza HP. Extracellular matrix remodeling derangement in ex-obese patients. Mol Cell Biochem 2016; 425:1-7. [DOI: 10.1007/s11010-016-2857-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 10/22/2016] [Indexed: 12/26/2022]
|
75
|
Hart PA, Bellin MD, Andersen DK, Bradley D, Cruz-Monserrate Z, Forsmark CE, Goodarzi MO, Habtezion A, Korc M, Kudva YC, Pandol SJ, Yadav D, Chari ST. Type 3c (pancreatogenic) diabetes mellitus secondary to chronic pancreatitis and pancreatic cancer. Lancet Gastroenterol Hepatol 2016; 1:226-237. [PMID: 28404095 DOI: 10.1016/s2468-1253(16)30106-6] [Citation(s) in RCA: 281] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 07/25/2016] [Accepted: 07/26/2016] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus is a group of diseases defined by persistent hyperglycaemia. Type 2 diabetes, the most prevalent form, is characterised initially by impaired insulin sensitivity and subsequently by an inadequate compensatory insulin response. Diabetes can also develop as a direct consequence of other diseases, including diseases of the exocrine pancreas. Historically, diabetes due to diseases of the exocrine pancreas was described as pancreatogenic or pancreatogenous diabetes mellitus, but recent literature refers to it as type 3c diabetes. It is important to note that type 3c diabetes is not a single entity; it occurs because of a variety of exocrine pancreatic diseases with varying mechanisms of hyperglycaemia. The most commonly identified causes of type 3c diabetes are chronic pancreatitis, pancreatic ductal adenocarcinoma, haemochromatosis, cystic fibrosis, and previous pancreatic surgery. In this Review, we discuss the epidemiology, pathogenesis, and clinical relevance of type 3c diabetes secondary to chronic pancreatitis and pancreatic ductal adenocarcinoma, and highlight several important knowledge gaps.
Collapse
Affiliation(s)
- Phil A Hart
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| | - Melena D Bellin
- Division of Pediatric Endocrinology and Schulze Diabetes Institute, University of Minnesota Medical Center, Minneapolis, MN, USA
| | - Dana K Andersen
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David Bradley
- Division of Endocrinology, Diabetes, and Metabolism, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Christopher E Forsmark
- Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville, FL, USA
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Aida Habtezion
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Murray Korc
- Departments of Medicine, Biochemistry, and Molecular Biology, Indiana University School of Medicine, Indiana University Simon Cancer Center, Indianapolis, IN, USA; Pancreatic Cancer Signature Center, Indiana University Simon Cancer Center, Indianapolis, IN, USA
| | - Yogish C Kudva
- Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Stephen J Pandol
- Department of Veterans Affairs, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dhiraj Yadav
- Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh and UPMC Medical Center, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh and UPMC Medical Center, Pittsburgh, PA, USA
| | - Suresh T Chari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
76
|
Ponnusamy S, Tran QT, Harvey I, Smallwood HS, Thiyagarajan T, Banerjee S, Johnson DL, Dalton JT, Sullivan RD, Miller DD, Bridges D, Narayanan R. Pharmacologic activation of estrogen receptor β increases mitochondrial function, energy expenditure, and brown adipose tissue. FASEB J 2016; 31:266-281. [PMID: 27733447 DOI: 10.1096/fj.201600787rr] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/22/2016] [Indexed: 01/03/2023]
Abstract
Most satiety-inducing obesity therapeutics, despite modest efficacy, have safety concerns that underscore the need for effective peripherally acting drugs. An attractive therapeutic approach for obesity is to optimize/maximize energy expenditure by increasing energy-utilizing thermogenic brown adipose tissue. We used in vivo and in vitro models to determine the role of estrogen receptor β (ER-β) and its ligands on adipose biology. RNA sequencing and metabolomics were used to determine the mechanism of action of ER-β and its ligands. Estrogen receptor β (ER-β) and its selective ligand reprogrammed preadipocytes and precursor stem cells into brown adipose tissue and increased mitochondrial respiration. An ER-β-selective ligand increased markers of tricarboxylic acid-dependent and -independent energy biogenesis and oxygen consumption in mice without a concomitant increase in physical activity or food consumption, all culminating in significantly reduced weight gain and adiposity. The antiobesity effects of ER-β ligand were not observed in ER-β-knockout mice. Serum metabolite profiles of adult lean and juvenile mice were comparable, while that of adult obese mice was distinct, indicating a possible impact of obesity on age-dependent metabolism. This phenotype was partially reversed by ER-β-selective ligand. These data highlight a new role for ER-β in adipose biology and its potential to be a safer alternative peripheral therapeutic target for obesity.-Ponnusamy, S., Tran, Q. T., Harvey, I., Smallwood, H. S., Thiyagarajan, T., Banerjee, S., Johnson, D. L., Dalton, J. T., Sullivan, R. D., Miller, D. D., Bridges, D., Narayanan, R. Pharmacologic activation of estrogen receptor β increases mitochondrial function, energy expenditure, and brown adipose tissue.
Collapse
Affiliation(s)
- Suriyan Ponnusamy
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Quynh T Tran
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Innocence Harvey
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Heather S Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Thirumagal Thiyagarajan
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Souvik Banerjee
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Daniel L Johnson
- Molecular Informatics Core, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - James T Dalton
- Preclinical Research and Development, GTx, Incorporated, Memphis, Tennessee, USA
| | - Ryan D Sullivan
- Department of Comparative Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA; and
| | - Duane D Miller
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Dave Bridges
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Ramesh Narayanan
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA; .,West Cancer Center, Memphis, Tennessee, USA
| |
Collapse
|
77
|
Lauhio A, Färkkilä E, Pietiläinen KH, Åström P, Winkelmann A, Tervahartiala T, Pirilä E, Rissanen A, Kaprio J, Sorsa TA, Salo T. Association of MMP-8 with obesity, smoking and insulin resistance. Eur J Clin Invest 2016; 46:757-65. [PMID: 27296149 DOI: 10.1111/eci.12649] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 06/13/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND Obesity has been recognized as a state of subclinical inflammation resulting in a loss of insulin receptors and decreased insulin sensitivity. We here studied in vivo the role of circulating matrix metalloproteinase-8 (MMP-8) among young healthy twin adults. Also, in vitro analysis of the cleavage of human insulin receptor (INSR) by MMP-8 was investigated as well its inhibition by doxycycline and other MMP-8 inhibitor, Ilomastat/GM6001, which are broad-spectrum MMP inhibitors. MATERIALS AND METHODS We analysed serum MMP-8 levels by a time-resolved immunofluorometric assay in obese (n = 34), overweight (n = 76) and normal weight (n = 130) twin individuals. The effect of MMP-8 on INSR and the effects of synthetic MMP-8 inhibitors, doxycycline and Ilomastat/GM6001, were studied by SDS-PAGE. RESULTS We found that in obese individuals relative to normal weight individuals, the serum MMP-8 levels and MMP-8/TIMP-1 ratio were significantly increased (P = 0·0031 and P = 0·031, respectively). Among normal weight and obese individuals, also smoking significantly increases serum MMP-8 and MMP-8/TIMP-1 ratio. In vitro, we found that INSR was degraded by MMP-8 and this was inhibited by doxycycline and Ilomastat/GM6001. CONCLUSIONS Obesity associated with elevated circulating MMP-8 found among young adults may contribute to progression of insulin resistance by cleaving INSR. This INSR cleavage by MMP-8 can be inhibited by synthetic MMP-8 inhibitors such as doxycycline. In addition to obesity, also smoking independently explained increased MMP-8 levels. Our results suggest that MMP-8 is an essential mediator in systemic subclinical inflammatory response in obesity, and a potential drug target.
Collapse
Affiliation(s)
- Anneli Lauhio
- Department of Infectious Diseases, Helsinki University Central Hospital, Helsinki, Finland.,Clinicum, University of Helsinki, Helsinki, Finland
| | - Esa Färkkilä
- Department of Infectious Diseases, Helsinki University Central Hospital, Helsinki, Finland.,Clinicum, University of Helsinki, Helsinki, Finland.,Department of Oral and Maxillofacial Diseases, Helsinki University Central Hospital, Helsinki, Finland
| | - Kirsi H Pietiläinen
- Clinicum, University of Helsinki, Helsinki, Finland.,Obesity Research Unit, Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland.,FIMM, Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland.,Endocrinology, Abdominal Center, Helsinki University Central Hospital, Helsinki, Finland
| | - Pirjo Åström
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Alina Winkelmann
- Department of Periodontology, Institute of Dentistry, University of Helsinki, Helsinki, Finland
| | - Taina Tervahartiala
- Department of Periodontology, Institute of Dentistry, University of Helsinki, Helsinki, Finland
| | - Emma Pirilä
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Aila Rissanen
- Obesity Research Unit, Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland.,Department of Psychiatry, Helsinki University Central Hospital, Helsinki, Finland
| | - Jaakko Kaprio
- FIMM, Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland.,Department of Public Health, University of Helsinki, Helsinki, Finland.,Department of Health, National Institute for Health and Welfare, Helsinki, Finland
| | - Timo A Sorsa
- Department of Oral and Maxillofacial Diseases, Helsinki University Central Hospital, Helsinki, Finland.,Department of Periodontology, Institute of Dentistry, University of Helsinki, Helsinki, Finland.,Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Tuula Salo
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland.,Department of Oral Pathology, Institute of Dentistry, University of Helsinki, Helsinki, Finland.,Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
78
|
Ham JR, Lee HI, Choi RY, Sim MO, Choi MS, Kwon EY, Yun KW, Kim MJ, Lee MK. Anti-obesity and anti-hepatosteatosis effects of dietary scopoletin in high-fat diet fed mice. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.06.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
79
|
Luo Y, Ma X, Pan X, Xu Y, Xiong Q, Xiao Y, Bao Y, Jia W. Serum lipocalin-2 levels are positively associated with not only total body fat but also visceral fat area in Chinese men. Medicine (Baltimore) 2016; 95:e4039. [PMID: 27472678 PMCID: PMC5265815 DOI: 10.1097/md.0000000000004039] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Serum lipocalin-2 (LCN2) plays an important role in the regulation of the obesity-associated dysmetabolic state and cardiovascular disease. However, relatively little is known about the relationship between serum LCN2 levels and body fat content and distribution. We examined the associations of total body fat content and abdominal fat distribution with serum LCN2 levels in Chinese men.The study was based on a cross-sectional analysis of data for 1203 Chinese men aged 22 to 78 years from the Shanghai Obesity Study. Body fat percentage (fat%) was assessed by bioelectrical impedance analysis, and magnetic resonance imaging was adopted to quantify the visceral fat area (VFA) and subcutaneous fat area (SFA). Serum levels of LCN2 were measured with a standard enzyme-linked immunosorbent assay method.Subjects with a high fat% had higher serum LCN2 levels than those with a normal fat% regardless of their body mass index category (<25 and ≥25 kg/m). The frequency of isolated high VFA was increased with increasing quintiles of serum LCN2 levels (P < 0.001), but the frequency of isolated high SFA did not differ between quintiles of serum LCN2 levels. A trend of increasing VFA was observed with increasing serum LCN2 levels (P < 0.001). Multiple stepwise regression analysis showed that VFA was positively associated with serum LCN2 levels, independent of overall obesity and other confounding factors (standardized β = 0.082, P = 0.008).Serum LCN2 levels are positively correlated with body fat content and independently associated with VFA in Chinese men.
Collapse
Affiliation(s)
- Yuqi Luo
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| | - Xiaojing Ma
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| | - Xiaoping Pan
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| | - Yiting Xu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| | - Qin Xiong
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| | - Yunfeng Xiao
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yuqian Bao
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
- Correspondence: Prof Yuqian Bao, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China (e-mail: )
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| |
Collapse
|
80
|
Coimbra S, Catarino C, Santos-Silva A. The triad psoriasis-obesity-adipokine profile. J Eur Acad Dermatol Venereol 2016; 30:1876-1885. [PMID: 27230733 DOI: 10.1111/jdv.13701] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 03/25/2016] [Indexed: 12/17/2022]
Abstract
Psoriasis is a chronic inflammatory skin disease, often associated with overweight/obesity. The adipose tissue is a complex organ that secretes several adipokines, involved in the regulation of some metabolic processes, such as lipid metabolism, glucose homeostasis, angiogenesis, blood pressure and inflammation. In obesity, the distribution and function of adipose tissue, and the adipokine profile are altered. The unbalanced production of pro- and anti-inflammatory adipokines in obesity, contributes to the development of a chronic low-grade inflammation state, which seems to favour worsening of psoriasis lesion and a poorer response to treatment. In this review, we will debate published data concerning the current knowledge about the triad psoriasis-obesity-adipokine profile.
Collapse
Affiliation(s)
- S Coimbra
- UCIBIO\REQUIMTE, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia, Universidade do Porto (FFUP), Porto, Portugal. .,CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, GRD-Paredes, Portugal.
| | - C Catarino
- UCIBIO\REQUIMTE, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia, Universidade do Porto (FFUP), Porto, Portugal
| | - A Santos-Silva
- UCIBIO\REQUIMTE, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia, Universidade do Porto (FFUP), Porto, Portugal.
| |
Collapse
|
81
|
Di Rosa M, Malaguarnera L. Chitinase 3 Like-1: An Emerging Molecule Involved in Diabetes and Diabetic Complications. Pathobiology 2016; 83:228-42. [DOI: 10.1159/000444855] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/18/2016] [Indexed: 11/19/2022] Open
|
82
|
Kamble PG, Pereira MJ, Sidibeh CO, Amini S, Sundbom M, Börjesson JL, Eriksson JW. Lipocalin 2 produces insulin resistance and can be upregulated by glucocorticoids in human adipose tissue. Mol Cell Endocrinol 2016; 427:124-32. [PMID: 26973291 DOI: 10.1016/j.mce.2016.03.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/11/2016] [Accepted: 03/08/2016] [Indexed: 01/09/2023]
Abstract
The adipokine lipocalin 2 is linked to obesity and metabolic disorders. However, its role in human adipose tissue glucose and lipid metabolism is not explored. Here we show that the synthetic glucocorticoid dexamethasone dose-dependently increased lipocalin 2 gene expression in subcutaneous and omental adipose tissue from pre-menopausal females, while it had no effect in post-menopausal females or in males. Subcutaneous adipose tissue from both genders treated with recombinant human lipocalin 2 showed a reduction in protein levels of GLUT1 and GLUT4 and in glucose uptake in isolated adipocytes. In subcutaneous adipose tissue, lipocalin 2 increased IL-6 gene expression whereas expression of PPARγ and adiponectin was reduced. Our findings suggest that lipocalin 2 can contribute to insulin resistance in human adipose tissue. In pre-menopausal females, it may partly mediate adverse metabolic effects exerted by glucocorticoid excess.
Collapse
Affiliation(s)
- Prasad G Kamble
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Maria J Pereira
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Cherno O Sidibeh
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Sam Amini
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Magnus Sundbom
- Department of Surgery, Uppsala University, Uppsala, Sweden
| | | | - Jan W Eriksson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
83
|
Lin CW, Yang WE, Lee WJ, Hua KT, Hsieh FK, Hsiao M, Chen CC, Chow JM, Chen MK, Yang SF, Chien MH. Lipocalin 2 prevents oral cancer metastasis through carbonic anhydrase IX inhibition and is associated with favourable prognosis. Carcinogenesis 2016; 37:712-722. [PMID: 27207653 DOI: 10.1093/carcin/bgw050] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 04/12/2016] [Indexed: 12/11/2022] Open
Abstract
Lipocalin 2 (LCN2), a secreted glycoprotein, is up- or downregulated in different human cancers. At present, the functional role of LCN2 in the progression of oral squamous cell carcinoma (OSCC), which accounts for most head and neck cancers, remains poorly understood, particularly with respect to its involvement in invasion and metastasis. In this study, we observed that LCN2 expression decreased in patients with OSCC and lymph node metastasis compared with that in patients without metastasis. A higher LCN2 expression correlated with the survival of patients with OSCC. Furthermore, LCN2 overexpression in OSCC cells reduced in vitro migration and invasion and in vivo metastasis, whereas its silencing induced an increase in cell motility. Mechanistically, LCN2 inhibited the cell motility of OSCC cells through hypoxia-inducible factor (HIF)-1α-dependent transcriptional inhibition of the carbonic anhydrase IX (CAIX). CAIX overexpression relieved the migration inhibition imposed by LCN2 overexpression in OSCC cells. Moreover, a microRNA (miR) analysis revealed that LCN2 can suppress CAIX expression and cell migration through miR-4505 induction. Examination of tumour tissues from patients with OSCC and OSCC-transplanted mice revealed an inverse correlation between LCN2 and CAIX expression. Furthermore, patients with LCN2(strong)/CAIX(weak) revealed the lowest frequency of lymph node metastasis and the longest survival. Our findings suggest that LCN2 suppresses tumour metastasis by targeting the transcriptional and post-transcriptional regulation of CAIX in OSCC cells. LCN2 overexpression may be a novel OSCC treatment strategy and a useful biomarker for predicting OSCC progression.
Collapse
Affiliation(s)
- Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung 40201, Taiwan.,Department of Dentistry, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Wei-En Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Wei-Jiunn Lee
- Department of Medical Research, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
| | - Kuo-Tai Hua
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Feng-Koo Hsieh
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians University, 80539 Munich, Germany
| | - Michael Hsiao
- The Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Chia-Cheng Chen
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Shin-Kong Memorial Hospital, Taipei 111, Taiwan
| | - Jyh-Ming Chow
- Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
| | - Mu-Kuan Chen
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.,Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, Changhua 505, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan and
| | - Ming-Hsien Chien
- Department of Medical Research, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan.,Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
84
|
Cabia B, Andrade S, Carreira MC, Casanueva FF, Crujeiras AB. A role for novel adipose tissue-secreted factors in obesity-related carcinogenesis. Obes Rev 2016; 17:361-76. [PMID: 26914773 DOI: 10.1111/obr.12377] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 12/15/2015] [Indexed: 12/12/2022]
Abstract
Obesity, a pandemic disease, is caused by an excessive accumulation of fat that can have detrimental effects on health. Adipose tissue plays a very important endocrine role, secreting different molecules that affect body physiology. In obesity, this function is altered, leading to a dysfunctional production of several factors, known as adipocytokines. This process has been linked to various comorbidities associated with obesity, such as carcinogenesis. In fact, several classical adipocytokines with increased levels in obesity have been demonstrated to exert a pro-carcinogenic role, including leptin, TNF-α, IL-6 and resistin, whereas others like adiponectin, with decreased levels in obesity, might have an anti-carcinogenic function. In this expanding field, new proteomic techniques and approaches have allowed the identification of novel adipocytokines, a number of which exhibit an altered production in obesity and type 2 diabetes and thus are related to adiposity. Many of these novel adipocytokines have also been identified in various tumour types, such as that of the breast, liver or endometrium, thereby increasing the list of potential contributors to carcinogenesis. This review is focused on the regulation of these novel adipocytokines by obesity, including apelin, endotrophin, FABP4, lipocalin 2, omentin-1, visfatin, chemerin, ANGPTL2 or osteopontin, emphasizing its involvement in tumorigenesis.
Collapse
Affiliation(s)
- B Cabia
- Laboratory of Molecular and Cellular Endocrinology, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Madrid, Spain
| | - S Andrade
- Laboratory of Molecular and Cellular Endocrinology, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Madrid, Spain
| | - M C Carreira
- Laboratory of Molecular and Cellular Endocrinology, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Madrid, Spain
| | - F F Casanueva
- Laboratory of Molecular and Cellular Endocrinology, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Madrid, Spain
| | - A B Crujeiras
- Laboratory of Molecular and Cellular Endocrinology, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Madrid, Spain
| |
Collapse
|
85
|
Yamada Y, Miyamoto T, Kashima H, Kobara H, Asaka R, Ando H, Higuchi S, Ida K, Shiozawa T. Lipocalin 2 attenuates iron-related oxidative stress and prolongs the survival of ovarian clear cell carcinoma cells by up-regulating the CD44 variant. Free Radic Res 2016; 50:414-25. [PMID: 26729415 DOI: 10.3109/10715762.2015.1134795] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Ovarian clear cell carcinoma (CCC) arises from ovarian endometriosis. Intra-cystic fluid contains abundant amounts of free iron, which causes persistent oxidative stress, a factor that has been suggested to induce malignant transformation. However, the mechanisms linking oxidative stress and carcinogenesis in CCC currently remain unclear. Lipocalin 2 (LCN2), a multifunctional secretory protein, functions as an iron transporter as well as an antioxidant. Therefore, we herein examined the roles of LCN2 in the regulation of intracellular iron concentrations, oxidative stress, DNA damage, and antioxidative functions using LCN2-overexpressing (ES2), and LCN2-silenced (RMG-1) CCC cell lines. The results of calcein staining indicated that the up-regulated expression of LCN2 correlated with increases in intracellular iron concentrations. However, a DCFH-DA assay and 8OHdG staining revealed that LCN2 reduced intracellular levels of reactive oxygen species and DNA damage. Furthermore, the expression of LCN2 suppressed hydrogen peroxide-induced apoptosis and prolonged cell survival, suggesting an antioxidative role for LCN2. The expression of mRNAs and proteins for various oxidative stress-catalyzing enzymes, such as heme oxygenase (HO), superoxide dismutase (SOD), and glutathione peroxidase, was not affected by LCN2, whereas the intracellular concentration of the potent antioxidant, glutathione (GSH), was increased by LCN2. Furthermore, the expression of xCT, a cystine transporter protein, and CD44 variant 8-10 (CD44v), a stem cell marker, was up-regulated by LCN2. Although LCN2 increased intracellular iron concentrations, LCN2-induced GSH may catalyze and override oxidative stress via CD44v and xCT, and subsequently enhance the survival of CCC cells in oxidative stress-rich endometriosis.
Collapse
Affiliation(s)
- Yasushi Yamada
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Tsutomu Miyamoto
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Hiroyasu Kashima
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Hisanori Kobara
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Ryoichi Asaka
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Hirofumi Ando
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Shotaro Higuchi
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Koichi Ida
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Tanri Shiozawa
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| |
Collapse
|
86
|
Hau CS, Kanda N, Tada Y, Shibata S, Uozaki H, Fukusato T, Sato S, Watanabe S. Lipocalin-2 exacerbates psoriasiform skin inflammation by augmenting T-helper 17 response. J Dermatol 2015; 43:785-94. [DOI: 10.1111/1346-8138.13227] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 10/15/2015] [Indexed: 12/01/2022]
Affiliation(s)
- Carren S. Hau
- Department of Dermatology; Teikyo University School of Medicine; Tokyo Japan
| | - Naoko Kanda
- Department of Dermatology; Teikyo University School of Medicine; Tokyo Japan
| | - Yayoi Tada
- Department of Dermatology; Teikyo University School of Medicine; Tokyo Japan
- Department of Dermatology; University of Tokyo Faculty of Medicine; Tokyo Japan
| | - Sayaka Shibata
- Department of Dermatology; University of Tokyo Faculty of Medicine; Tokyo Japan
| | - Hiroshi Uozaki
- Department of Pathology; Teikyo University School of Medicine; Tokyo Japan
| | - Toshio Fukusato
- Department of Pathology; Teikyo University School of Medicine; Tokyo Japan
| | - Shinichi Sato
- Department of Dermatology; University of Tokyo Faculty of Medicine; Tokyo Japan
| | - Shinichi Watanabe
- Department of Dermatology; Teikyo University School of Medicine; Tokyo Japan
| |
Collapse
|
87
|
Cabedo Martinez AI, Weinhäupl K, Lee WK, Wolff NA, Storch B, Żerko S, Konrat R, Koźmiński W, Breuker K, Thévenod F, Coudevylle N. Biochemical and Structural Characterization of the Interaction between the Siderocalin NGAL/LCN2 (Neutrophil Gelatinase-associated Lipocalin/Lipocalin 2) and the N-terminal Domain of Its Endocytic Receptor SLC22A17. J Biol Chem 2015; 291:2917-30. [PMID: 26635366 PMCID: PMC4742754 DOI: 10.1074/jbc.m115.685644] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Indexed: 11/22/2022] Open
Abstract
The neutrophil gelatinase-associated lipocalin (NGAL, also known as LCN2) and its cellular receptor (LCN2-R, SLC22A17) are involved in many physiological and pathological processes such as cell differentiation, apoptosis, and inflammation. These pleiotropic functions mainly rely on NGAL's siderophore-mediated iron transport properties. However, the molecular determinants underlying the interaction between NGAL and its cellular receptor remain largely unknown. Here, using solution-state biomolecular NMR in conjunction with other biophysical methods, we show that the N-terminal domain of LCN2-R is a soluble extracellular domain that is intrinsically disordered and interacts with NGAL preferentially in its apo state to form a fuzzy complex. The relatively weak affinity (≈10 μm) between human LCN2-R-NTD and apoNGAL suggests that the N terminus on its own cannot account for the internalization of NGAL by LCN2-R. However, human LCN2-R-NTD could be involved in the fine-tuning of the interaction between NGAL and its cellular receptor or in a biochemical mechanism allowing the receptor to discriminate between apo- and holo-NGAL.
Collapse
Affiliation(s)
- Ana-Isabel Cabedo Martinez
- From the Department of Computational and Structural Biology, Max F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030 Vienna, Austria
| | - Katharina Weinhäupl
- From the Department of Computational and Structural Biology, Max F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030 Vienna, Austria
| | - Wing-Kee Lee
- Chair of Physiology, Pathophysiology, and Toxicology and ZBAF, Faculty of Health, School of Medicine, Witten/Herdecke University, Stockumer Strasse 12, 58453 Witten, Germany
| | - Natascha A Wolff
- Chair of Physiology, Pathophysiology, and Toxicology and ZBAF, Faculty of Health, School of Medicine, Witten/Herdecke University, Stockumer Strasse 12, 58453 Witten, Germany
| | - Barbara Storch
- Institute of Organic Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, CCB, Innrain 80/82, 6020 Innsbruck, Austria, and
| | - Szymon Żerko
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-089 Warsaw, Poland
| | - Robert Konrat
- From the Department of Computational and Structural Biology, Max F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030 Vienna, Austria
| | - Wiktor Koźmiński
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-089 Warsaw, Poland
| | - Kathrin Breuker
- Institute of Organic Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, CCB, Innrain 80/82, 6020 Innsbruck, Austria, and
| | - Frank Thévenod
- Chair of Physiology, Pathophysiology, and Toxicology and ZBAF, Faculty of Health, School of Medicine, Witten/Herdecke University, Stockumer Strasse 12, 58453 Witten, Germany
| | - Nicolas Coudevylle
- From the Department of Computational and Structural Biology, Max F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030 Vienna, Austria,
| |
Collapse
|
88
|
Rodríguez A, Ezquerro S, Méndez-Giménez L, Becerril S, Frühbeck G. Revisiting the adipocyte: a model for integration of cytokine signaling in the regulation of energy metabolism. Am J Physiol Endocrinol Metab 2015; 309:E691-714. [PMID: 26330344 DOI: 10.1152/ajpendo.00297.2015] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/24/2015] [Indexed: 02/08/2023]
Abstract
Adipose tissue constitutes an extremely active endocrine organ with a network of signaling pathways enabling the organism to adapt to a wide range of different metabolic challenges, such as starvation, stress, infection, and short periods of gross energy excess. The functional pleiotropism of adipose tissue relies on its ability to synthesize and release a huge variety of hormones, cytokines, complement and growth factors, extracellular matrix proteins, and vasoactive factors, collectively termed adipokines. Obesity is associated with adipose tissue dysfunction leading to the onset of several pathologies including type 2 diabetes, dyslipidemia, nonalcoholic fatty liver, or hypertension, among others. The mechanisms underlying the development of obesity and its associated comorbidities include the hypertrophy and/or hyperplasia of adipocytes, adipose tissue inflammation, impaired extracellular matrix remodeling, and fibrosis together with an altered secretion of adipokines. Recently, the potential role of brown and beige adipose tissue in the protection against obesity has been also recognized. In contrast to white adipocytes, which store energy in the form of fat, brown and beige fat cells display energy-dissipating capacity through the promotion of triacylglycerol clearance, glucose disposal, and generation of heat for thermogenesis. Identification of the morphological and molecular changes in white, beige, and brown adipose tissue during weight gain is of utmost relevance for the identification of pharmacological targets for the treatment of obesity and its associated metabolic diseases.
Collapse
Affiliation(s)
- Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; and Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Silvia Ezquerro
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | - Leire Méndez-Giménez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; and Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; and Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; and Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| |
Collapse
|
89
|
Abstract
Objective The objective of this study was to assess the emerging biochemical markers of arterial remodeling in patients with morbid obesity before and after surgical treatment and to compare the results to a control group. Material and Methods The prospective study included 40 patients with BMI 47.73 ± 6.18 kg/m2, qualified for elective bariatric surgery and re-examined 6 months after the surgery. The control group consisted of non obese, age and sex matched 15 subjects. Following laboratory examinations were performed in all patients: basic laboratory examinations, MMP-2, MMP-9, adiponectin, PAI-1, CD40L, E-selectin. Results Examination of patients 6 m after bariatric surgery revealed a 34.57 ± 9.71 reduction in excess body weight. Comparison of the study group at two time points revealed differences in adiponectin, MMP-2 and MMP-9 levels. Hypoadiponectinemia was observed in 35 % patients 6 months after bariatric surgery compared to 90 % patients before the surgery. In addition, a strong correlation was observed between body fat mass and adiponectin levels (r = −0.504, p = 0.055). Moderate correlations were demonstrated between E-selectin levels and BMI (r = 0.361; p = 0.022), and metalloproteinase-9 levels (r = 0.326; p = 0.040). In addition, strong relationship was demonstrated between MMP-2 and MMP-9 (r = 0.502; p = 0.001), and moderate between MMP-2 and adiponectin levels (r = 0.449; p = 0.003). MMP-9 levels were moderately correlated with HDL-cholesterol levels (r = 0.316; p = 0.046). Conclusions Assessment of laboratory markers of arterial remodeling and metabolism suggest their adverse changes in patients with morbid obesity. However, body mass reduction due to bariatric surgery decreases inflammatory status, improves biochemical markers of arterial remodeling as well as to beneficial changes in the metabolism.
Collapse
|
90
|
Lee YJ, Heo YS, Park HS, Lee SH, Lee SK, Jang YJ. Serum SPARC and matrix metalloproteinase-2 and metalloproteinase-9 concentrations after bariatric surgery in obese adults. Obes Surg 2015; 24:604-10. [PMID: 24234777 DOI: 10.1007/s11695-013-1111-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Remodeling of the extracellular matrix (ECM) of adipose tissue is regarded as part of the pathophysiology of obesity. Secreted protein acidic and rich in cysteine (SPARC) was the first ECM protein described in adipose tissue. Matrix metalloproteinases (MMPs) also play a role in ECM remodeling, and MMP-2 and MMP-9 may be associated with abnormal ECM metabolism. Here, we investigated changes in serum SPARC, MMP-2, and MMP-9 concentrations after bariatric surgery in obese adults. METHODS We recruited 34 obese patients who were scheduled to undergo bariatric surgery for weight loss. We analyzed changes in serum SPARC, MMP-2, and MMP-9 concentrations before and 9 months after bariatric surgery and any associations between changes in SPARC, MMP-2, and MMP-9 concentrations and obesity-related parameters. RESULTS Serum leptin levels significantly decreased, and the serum adiponectin level significantly increased after bariatric surgery. The serum SPARC concentration decreased significantly from 165.0 ± 18.2 to 68.7 ± 6.7 ng/mL (p < 0.001), and the MMP-2 concentration also decreased significantly from 262.2 ± 15.2 to 235.9 ± 10.5 ng/mL (p < 0.001). Changes in the serum SPARC concentration were significantly correlated with HOMA-IR changes, and changes in the serum MMP-9 concentration were found to inversely correlate with serum adiponectin changes. CONCLUSION These findings show that significant decreases in serum SPARC and MMP-2 concentrations occur after bariatric surgery. Our results thus suggest that weight loss via bariatric surgery could alter the ECM environment, and that these changes are related to certain metabolic changes.
Collapse
|
91
|
Ferreira AC, Dá Mesquita S, Sousa JC, Correia-Neves M, Sousa N, Palha JA, Marques F. From the periphery to the brain: Lipocalin-2, a friend or foe? Prog Neurobiol 2015; 131:120-36. [PMID: 26159707 DOI: 10.1016/j.pneurobio.2015.06.005] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 06/23/2015] [Accepted: 06/28/2015] [Indexed: 01/08/2023]
Abstract
Lipocalin-2 (LCN2) is an acute-phase protein that, by binding to iron-loaded siderophores, acts as a potent bacteriostatic agent in the iron-depletion strategy of the immune system to control pathogens. The recent identification of a mammalian siderophore also suggests a physiological role for LCN2 in iron homeostasis, specifically in iron delivery to cells via a transferrin-independent mechanism. LCN2 participates, as well, in a variety of cellular processes, including cell proliferation, cell differentiation and apoptosis, and has been mostly found up-regulated in various tissues and under inflammatory states, being its expression regulated by several inducers. In the central nervous system less is known about the processes involving LCN2, namely by which cells it is produced/secreted, and its impact on cell proliferation and death, or in neuronal plasticity and behaviour. Importantly, LCN2 recently emerged as a potential clinical biomarker in multiple sclerosis and in ageing-related cognitive decline. Still, there are conflicting views on the role of LCN2 in pathophysiological processes, with some studies pointing to its neurodeleterious effects, while others indicate neuroprotection. Herein, these various perspectives are reviewed and a comprehensive and cohesive view of the general function of LCN2, particularly in the brain, is provided.
Collapse
Affiliation(s)
- Ana C Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sandro Dá Mesquita
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João C Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana A Palha
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
92
|
Tarjus A, Martínez-Martínez E, Amador C, Latouche C, El Moghrabi S, Berger T, Mak TW, Fay R, Farman N, Rossignol P, Zannad F, López-Andrés N, Jaisser F. Neutrophil Gelatinase-Associated Lipocalin, a Novel Mineralocorticoid Biotarget, Mediates Vascular Profibrotic Effects of Mineralocorticoids. Hypertension 2015; 66:158-66. [PMID: 25987661 DOI: 10.1161/hypertensionaha.115.05431] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 03/18/2015] [Indexed: 01/19/2023]
Abstract
Activation of the mineralocorticoid receptor has been shown to be deleterious in cardiovascular diseases (CVDs). We have recently shown that lipocalin 2 (Lcn2), or neutrophil gelatinase-associated lipocalin (NGAL), is a primary target of aldosterone/mineralocorticoid receptor in the cardiovascular system. Lcn2 is a circulating protein, which binds matrix metalloproteinase 9 and modulates its stability. We hypothesized that Lcn2 could be a mediator of aldosterone/mineralocorticoid receptor profibrotic effects in the cardiovascular system. Correlations between aldosterone and profibrotic markers, such as procollagen type I N-terminal peptide, were investigated in healthy subjects and subjects with abdominal obesity. The implication of Lcn2 in the mineralocorticoid pathway was studied using Lcn2 knockout mice subjected to a nephrectomy/aldosterone/salt (NAS) challenge for 4 weeks. In human subjects, NGAL/matrix metalloproteinase 9 was positively correlated with plasma aldosterone and fibrosis biomarkers. In mice, loss of Lcn2 prevented the NAS-induced increase of plasma procollagen type I N-terminal peptide, as well as the increase of collagen fibers deposition and collagen I expression in the coronary vessels and the aorta. The lack of Lcn2 also blunted the NAS-induced increase in systolic blood pressure. Ex vivo, treatment of human fibroblasts with recombinant Lcn2 induced the expression of collagen I and the profibrotic galectin-3 and cardiotrophin-1 molecules. Our results showed that Lcn2 plays a key role in aldosterone/mineralocorticoid receptor-mediated vascular fibrosis. The clinical data indicate that this may translate in human patients. Lcn2 is, therefore, a new biotarget in cardiovascular fibrosis induced by mineralocorticoid activation.
Collapse
Affiliation(s)
- Antoine Tarjus
- From the INSERM UMR 1138 Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (A.T., C.A., C.L., S.E.M., N.F., F.J.); NAVARRABIOMED-FUNDACIÓN MIGUEL SERVET, Pamplona, Spain (E.M.-M., N.L.-A.); The Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, Canada (T.B., T.W.M.); INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); Université de Lorraine, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); and INI-CRCT F-CRIN, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.)
| | - Ernesto Martínez-Martínez
- From the INSERM UMR 1138 Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (A.T., C.A., C.L., S.E.M., N.F., F.J.); NAVARRABIOMED-FUNDACIÓN MIGUEL SERVET, Pamplona, Spain (E.M.-M., N.L.-A.); The Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, Canada (T.B., T.W.M.); INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); Université de Lorraine, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); and INI-CRCT F-CRIN, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.)
| | - Cristian Amador
- From the INSERM UMR 1138 Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (A.T., C.A., C.L., S.E.M., N.F., F.J.); NAVARRABIOMED-FUNDACIÓN MIGUEL SERVET, Pamplona, Spain (E.M.-M., N.L.-A.); The Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, Canada (T.B., T.W.M.); INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); Université de Lorraine, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); and INI-CRCT F-CRIN, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.)
| | - Céline Latouche
- From the INSERM UMR 1138 Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (A.T., C.A., C.L., S.E.M., N.F., F.J.); NAVARRABIOMED-FUNDACIÓN MIGUEL SERVET, Pamplona, Spain (E.M.-M., N.L.-A.); The Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, Canada (T.B., T.W.M.); INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); Université de Lorraine, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); and INI-CRCT F-CRIN, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.)
| | - Soumaya El Moghrabi
- From the INSERM UMR 1138 Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (A.T., C.A., C.L., S.E.M., N.F., F.J.); NAVARRABIOMED-FUNDACIÓN MIGUEL SERVET, Pamplona, Spain (E.M.-M., N.L.-A.); The Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, Canada (T.B., T.W.M.); INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); Université de Lorraine, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); and INI-CRCT F-CRIN, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.)
| | - Thorsten Berger
- From the INSERM UMR 1138 Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (A.T., C.A., C.L., S.E.M., N.F., F.J.); NAVARRABIOMED-FUNDACIÓN MIGUEL SERVET, Pamplona, Spain (E.M.-M., N.L.-A.); The Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, Canada (T.B., T.W.M.); INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); Université de Lorraine, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); and INI-CRCT F-CRIN, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.)
| | - Tak W Mak
- From the INSERM UMR 1138 Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (A.T., C.A., C.L., S.E.M., N.F., F.J.); NAVARRABIOMED-FUNDACIÓN MIGUEL SERVET, Pamplona, Spain (E.M.-M., N.L.-A.); The Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, Canada (T.B., T.W.M.); INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); Université de Lorraine, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); and INI-CRCT F-CRIN, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.)
| | - Renaud Fay
- From the INSERM UMR 1138 Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (A.T., C.A., C.L., S.E.M., N.F., F.J.); NAVARRABIOMED-FUNDACIÓN MIGUEL SERVET, Pamplona, Spain (E.M.-M., N.L.-A.); The Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, Canada (T.B., T.W.M.); INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); Université de Lorraine, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); and INI-CRCT F-CRIN, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.)
| | - Nicolette Farman
- From the INSERM UMR 1138 Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (A.T., C.A., C.L., S.E.M., N.F., F.J.); NAVARRABIOMED-FUNDACIÓN MIGUEL SERVET, Pamplona, Spain (E.M.-M., N.L.-A.); The Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, Canada (T.B., T.W.M.); INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); Université de Lorraine, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); and INI-CRCT F-CRIN, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.)
| | - Patrick Rossignol
- From the INSERM UMR 1138 Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (A.T., C.A., C.L., S.E.M., N.F., F.J.); NAVARRABIOMED-FUNDACIÓN MIGUEL SERVET, Pamplona, Spain (E.M.-M., N.L.-A.); The Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, Canada (T.B., T.W.M.); INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); Université de Lorraine, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); and INI-CRCT F-CRIN, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.)
| | - Faiez Zannad
- From the INSERM UMR 1138 Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (A.T., C.A., C.L., S.E.M., N.F., F.J.); NAVARRABIOMED-FUNDACIÓN MIGUEL SERVET, Pamplona, Spain (E.M.-M., N.L.-A.); The Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, Canada (T.B., T.W.M.); INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); Université de Lorraine, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); and INI-CRCT F-CRIN, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.)
| | - Natalia López-Andrés
- From the INSERM UMR 1138 Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (A.T., C.A., C.L., S.E.M., N.F., F.J.); NAVARRABIOMED-FUNDACIÓN MIGUEL SERVET, Pamplona, Spain (E.M.-M., N.L.-A.); The Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, Canada (T.B., T.W.M.); INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); Université de Lorraine, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); and INI-CRCT F-CRIN, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.)
| | - Frédéric Jaisser
- From the INSERM UMR 1138 Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (A.T., C.A., C.L., S.E.M., N.F., F.J.); NAVARRABIOMED-FUNDACIÓN MIGUEL SERVET, Pamplona, Spain (E.M.-M., N.L.-A.); The Campbell Family Institute for Cancer Research, University Health Network, Toronto, ON, Canada (T.B., T.W.M.); INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); Université de Lorraine, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.); and INI-CRCT F-CRIN, Nancy, France (R.F., P.R., F.Z., N.L.-A., F.J.).
| |
Collapse
|
93
|
Catalán V, Gómez-Ambrosi J, Rodríguez A, Ramírez B, Valentí V, Moncada R, Silva C, Salvador J, Frühbeck G. Peripheral mononuclear blood cells contribute to the obesity-associated inflammatory state independently of glycemic status: involvement of the novel proinflammatory adipokines chemerin, chitinase-3-like protein 1, lipocalin-2 and osteopontin. GENES AND NUTRITION 2015; 10:460. [PMID: 25869413 DOI: 10.1007/s12263-015-0460-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 03/28/2015] [Indexed: 12/17/2022]
Abstract
Inflammation is a critical contributor to the pathogenesis of metabolic disorders with adipose tissue being crucial in the inflammatory response by releasing multiple adipokines with either pro- or anti-inflammatory activities with potential functions as metabolic regulators. Peripheral blood mononuclear cells (PBMC) have been proposed as representative of the inflammatory status in obesity. The aim of the present study was to evaluate the contribution of PBMC to the obesity-associated chronic inflammation analyzing the expression of novel adipokines. Samples obtained from 69 subjects were used in the study. Real-time PCR determinations were performed to quantify gene expression levels in PBMC of novel adipokines including chemerin, chitinase-3-like protein 1 (YKL-40), lipocalin-2 (LCN-2) and osteopontin (OPN), and their circulating concentrations were also determined by ELISA. We show, for the first time, that PBMC gene expression levels of chemerin (P < 0.0001), chitinase-3-like protein 1 (P = 0.010), lipocalin-2 (P < 0.0001) and osteopontin (P < 0.0001) were strongly upregulated in obesity independently of the glycemic state. Circulating concentrations of these adipokines followed the same trend being significantly higher (P < 0.05) in obese normoglycemic and type 2 diabetic patients compared to lean volunteers and also associated (P < 0.05) with their corresponding mRNA levels in PBMC. These results provide evidence that alterations in inflammation-related adipokines are manifest in PBMC, which might contribute to the low-grade chronic inflammation that characterizes obesity.
Collapse
Affiliation(s)
- Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,
| | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Kennedy A, Spiers JP, Crowley V, Williams E, Lithander FE. Postprandial adiponectin and gelatinase response to a high-fat versus an isoenergetic low-fat meal in lean, healthy men. Nutrition 2015; 31:863-70. [PMID: 25933495 DOI: 10.1016/j.nut.2015.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 01/21/2015] [Accepted: 01/21/2015] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Evidence suggests that an acute systemic inflammatory response is invoked after consumption of a high-energy meal. Postprandial regulation of adiponectin, an adipose tissue-derived, anti-inflammatory hormone, and the gelatinases, matrix metalloproteinase (MMP)-2 and MMP-9, endopeptidases implicated in a diverse range of inflammatory processes, remain inconclusive. The aim of this study was to assess the postprandial effect of a high-energy (1212 kcal) meal on plasma adiponectin, MMP-2 and MMP-9 activity, glucose, insulin, triacylglycerols, total cholesterol, high-density lipoprotein cholesterol, and the differential effects on these parameters depending on whether the test meal was high fat (HF; 46 g fat, 1210 kcal) or isoenergetic and low fat (LF; 15 g fat, 1214 kcal energy). METHODS Test meals were consumed by 17 lean, healthy men on two separate occasions with blood samples collected by venipuncture at baseline (0 h) and 1 and 3 h after consumption of each test meal. RESULTS At baseline, no significant difference was seen in the parameters between the two groups, except for MMP-2, MMP-9, and total cholesterol. Over the 3-h postprandial period, no significant differential effect of the HF versus the LF test meal was observed on adiponectin, MMP-2, MMP-9, or on metabolic markers other than triacylglycerol, which increased significantly in response to the HF test meal (time × treatment, P = 0.002). When analyzed independent of time, MMP-2 (treatment, P = 0.006), MMP-9 (treatment, P = 0.022), and glucose (treatment, P = 0.026) were lower after consumption of the HF meal compared with the LF test meal. When analyzed independent of treatment, adiponectin increased over the 3-h postprandial period (time, P = 0.031), but there was no change in MMP-2 or MMP-9 (time, P = 0.503 and P = 0.525, respectively). Over the 3-h postprandial period, insulin (time, P < 0.001) and total cholesterol (time, P = 0.002) increased, whereas glucose (time, P < 0.001) and high-density lipoprotein cholesterol (time, P < 0.001) decreased. CONCLUSION No differential effects of a HF versus a LF isoenergetic meal were seen on postprandial adiponectin or the gelatinases. Adiponectin increased in response to a high-energy meal independent of treatment, and the gelatinases were lower in response to the HF versus the LF isoenergetic meal, independent of time point. Given the considerable amount of time that humans spend in the postprandial state, additional research is necessary to further understand inflammatory changes in this state.
Collapse
Affiliation(s)
- Alan Kennedy
- Department of Pharmacology and Therapeutics, Trinity College Dublin, Dublin, Ireland.
| | - J Paul Spiers
- Department of Pharmacology and Therapeutics, Trinity College Dublin, Dublin, Ireland
| | - Vivion Crowley
- Department of Biochemistry, St James' Hospital, Dublin, Ireland
| | - Emlyn Williams
- Statistical Consulting Unit, The Australian National University, Canberra, Australia
| | - Fiona E Lithander
- Nutrition and Dietetics, Faculty of Health, University of Canberra, Canberra, Australia
| |
Collapse
|
95
|
Janas RM, Ochocińska A, Snitko R, Dudka D, Kierkuś J, Teisseyre M, Najberg E. Neutrophil gelatinase-associated lipocalin in blood in children with inflammatory bowel disease. J Gastroenterol Hepatol 2014; 29:1883-9. [PMID: 24720485 DOI: 10.1111/jgh.12597] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/27/2014] [Indexed: 12/09/2022]
Abstract
BACKGROUND Neutrophil gelatinase-associated lipocalin (NGAL) is a 25 kDa glycoprotein present in the bodily fluids and tissues. It is secreted by neutrophils, epithelial cells, hepatocytes and adipocytes, and its expression is highly increased in response to cellular stress. The role of NGAL in the pathophysiology of inflammatory bowel disease including Crohn's disease and ulcerative colitis in children has thus far not been studied. METHODS The following groups of children were included: (i) inflammatory bowel disease group, n = 36, aged from 1 to 18 years with Crohn's disease (n = 19) and ulcerative colitis (n = 17); (ii) control group, n = 126; and (iii) disease control group, n = 27, without inflammatory bowel disease, with a food and/or inhalant allergy. RESULTS Healthy children aged from 1 to 8 years exhibited lower NGAL level than those of 9 to 18 years old (39.0; 18.1-83.7 ng/mL vs 57.6; 28.7-107 ng/mL, P = 0.001). In the younger, but not in the older children, the serum NGAL level correlated with their age, r = 0.334, P = 0.001. In children with inflammatory bowel disease, serum NGAL level was higher (108; 37.3-245 ng/mL) than in healthy (42.0; 18.1-107 ng/mL) and allergic, noninflammatory bowel disease children (49.3; 19.3-107 ng/mL), P = 0.001. Serum NGAL levels in Crohn's disease and ulcerative colitis children did not correlate with age, gender, disease activity, and indices of the inflammation. CONCLUSION Serum NAGL levels are highly elevated in Crohn's disease and ulcerative colitis in children compared to the healthy control group. Systematic studies are needed to explain the role of this protein in the inflammatory bowel disease.
Collapse
Affiliation(s)
- Roman M Janas
- Department of Biochemistry, Radioimmunology and Experimental Medicine, The Children's Memorial Health Institute, Warsaw, Poland
| | | | | | | | | | | | | |
Collapse
|
96
|
Gómez-Ambrosi J, Catalán V, Rodríguez A, Andrada P, Ramírez B, Ibáñez P, Vila N, Romero S, Margall MA, Gil MJ, Moncada R, Valentí V, Silva C, Salvador J, Frühbeck G. Increased cardiometabolic risk factors and inflammation in adipose tissue in obese subjects classified as metabolically healthy. Diabetes Care 2014; 37:2813-21. [PMID: 25011950 DOI: 10.2337/dc14-0937] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE It has been suggested that individuals with the condition known as metabolically healthy obesity (MHO) may not have the same increased risk for the development of metabolic abnormalities as their non-metabolically healthy counterparts. However, the validity of this concept has recently been challenged, since it may not translate into lower morbidity and mortality. The aim of the current study was to compare the cardiometabolic/inflammatory profile and the prevalence of impaired glucose tolerance (IGT) and type 2 diabetes (T2D) in patients categorized as having MHO or metabolically abnormal obesity (MAO). RESEARCH DESIGN AND METHODS We performed a cross-sectional analysis to compare the cardiometabolic/inflammatory profile of 222 MHO and 222 MAO patients (62% women) matched by age, including 255 lean subjects as reference (cohort 1). In a second cohort, we analyzed the adipokine profile and the expression of genes involved in inflammation and extracellular matrix remodeling in visceral adipose tissue (VAT; n = 82) and liver (n = 55). RESULTS The cardiometabolic and inflammatory profiles (CRP, fibrinogen, uric acid, leukocyte count, and hepatic enzymes) were similarly increased in MHO and MAO in both cohorts. Moreover, above 30%of patients classified as MHO according to fasting plasma glucose exhibited IGT or T2D [corrected]. The profile of classic (leptin, adiponectin, resistin) as well as novel (serum amyloid A and matrix metallopeptidase 9) adipokines was almost identical in MHO and MAO groups in cohort 2. Expression of genes involved in inflammation and tissue remodeling in VAT and liver showed a similar alteration pattern in MHO and MAO individuals. CONCLUSIONS The current study provides evidence for the existence of a comparable adverse cardiometabolic profile in MHO and MAO patients; thus the MHO concept should be applied with caution. A better identification of the obesity phenotypes and a more precise diagnosis are needed for improving the management of obese individuals.
Collapse
Affiliation(s)
- Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Pamplona, Spain
| | - Patricia Andrada
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Pamplona, Spain
| | - Patricia Ibáñez
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Pamplona, Spain Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Neus Vila
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Sonia Romero
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - María A Margall
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - María J Gil
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Pamplona, Spain Department of Biochemistry, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- Department of Anesthesiology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Víctor Valentí
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Pamplona, Spain Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Camilo Silva
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Pamplona, Spain Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Salvador
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Pamplona, Spain Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Pamplona, Spain Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
97
|
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
|
98
|
Lancha A, Rodríguez A, Catalán V, Becerril S, Sáinz N, Ramírez B, Burrell MA, Salvador J, Frühbeck G, Gómez-Ambrosi J. Osteopontin deletion prevents the development of obesity and hepatic steatosis via impaired adipose tissue matrix remodeling and reduced inflammation and fibrosis in adipose tissue and liver in mice. PLoS One 2014; 9:e98398. [PMID: 24871103 PMCID: PMC4037189 DOI: 10.1371/journal.pone.0098398] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 05/02/2014] [Indexed: 02/06/2023] Open
Abstract
Osteopontin (OPN) is a multifunctional extracellular matrix (ECM) protein involved in multiple physiological processes. OPN expression is dramatically increased in visceral adipose tissue in obesity and the lack of OPN protects against the development of insulin resistance and inflammation in mice. We sought to unravel the potential mechanisms involved in the beneficial effects of the absence of OPN. We analyzed the effect of the lack of OPN in the development of obesity and hepatic steatosis induced by a high-fat diet (HFD) using OPN-KO mice. OPN expression was upregulated in epididymal white adipose tissue (EWAT) and liver in wild type (WT) mice with HFD. OPN-KO mice had higher insulin sensitivity, lower body weight and fat mass with reduced adipose tissue ECM remodeling and reduced adipocyte size than WT mice under a HFD. Reduced MMP2 and MMP9 activity was involved in the decreased ECM remodeling. Crown-like structure number in EWAT as well as F4/80-positive cells and Emr1 expression in EWAT and liver increased with HFD, while OPN-deficiency blunted the increase. Moreover, our data show for the first time that OPN-KO under a HFD mice display reduced fibrosis in adipose tissue and liver, as well as reduced oxidative stress in adipose tissue. Gene expression of collagens Col1a1, Col6a1 and Col6a3 in EWAT and liver, as well as the profibrotic cytokine Tgfb1 in EWAT were increased with HFD, while OPN-deficiency prevented this increase. OPN deficiency prevented hepatic steatosis via reduction in the expression of molecules involved in the onset of fat accumulation such as Pparg, Srebf1, Fasn, Mogat1, Dgat2 and Cidec. Furthermore, OPN-KO mice exhibited higher body temperature and improved BAT function. The present data reveal novel mechanisms of OPN in the development of obesity, pointing out the inhibition of OPN as a promising target for the treatment of obesity and fatty liver.
Collapse
Affiliation(s)
- Andoni Lancha
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Neira Sáinz
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - María A. Burrell
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Histology and Pathology, University of Navarra, Pamplona, Spain
| | - Javier Salvador
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- * E-mail:
| |
Collapse
|
99
|
Lou Y, Wu C, Wu M, Xie C, Ren L. The changes of neutrophil gelatinase-associated lipocalin in plasma and its expression in adipose tissue in pregnant women with gestational diabetes. Diabetes Res Clin Pract 2014; 104:136-42. [PMID: 24530115 DOI: 10.1016/j.diabres.2014.01.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 12/20/2013] [Accepted: 01/13/2014] [Indexed: 11/26/2022]
Abstract
AIMS To investigate plasma levels and the expression of neutrophil gelatinase-associated lipocalin (NGAL) in subcutaneous adipose tissue (SAT) in patients with gestational diabetes mellitus (GDM). METHODS The study recruited 260 Chinese women divided into three groups: 96 were healthy pregnant women with pre-pregnancy body mass index (pre-pregnancy BMI) below 25kg/m(2) (GROUP 1), 84 were women with GDM with pre-pregnancy BMI below 25kg/m(2) (GROUP 2) and 80 were women with GDM with pre-pregnancy BMI over 25kg/m(2) (GROUP 3). Laboratory and anthropometric measurements were recorded and NGAL plasma levels were determined by ELISA for subjects in all groups. Real-time RT-PCR and Western blotting were used to assess the relative mRNA and protein expression of NGAL and tumor necrosis factor-α (TNF-α) in SAT (30 cases in each group). RESULTS Our results demonstrated statistically significant elevation in plasma NGAL concentrations in GROUP 2 and GROUP 3 compared with GROUP 1 (p<0.001 for both group comparisons). Moreover, SAT NGAL mRNA (p<0.001 and p<0.001, respectively) and protein (p<0.001 and p<0.001, respectively) expression levels were higher in GROUP 3 than in both GROUP 1 and GROUP 2. Correlations were noted between the plasma NGAL concentration and various parameters of insulin resistance. CONCLUSIONS Plasma NGAL may play a role in the development of insulin resistance in GDM, and the high levels of NGAL expression in SAT in overweight women with GDM suggests that NGAL in SAT is associated with obesity in women with GDM.
Collapse
Affiliation(s)
- Yanqin Lou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chaoying Wu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Min Wu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Cui Xie
- Department of Obstetrics and Gynecology, Shenzhen Baoan Maternal and Child Health Hospital, Shenzhen, China
| | - Lirong Ren
- Department of Obstetrics and Gynecology, Shenzhen Baoan Maternal and Child Health Hospital, Shenzhen, China.
| |
Collapse
|
100
|
Zhao P, Elks CM, Stephens JM. The induction of lipocalin-2 protein expression in vivo and in vitro. J Biol Chem 2014; 289:5960-9. [PMID: 24391115 DOI: 10.1074/jbc.m113.532234] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Lipocalin-2 (LCN2) is secreted from adipocytes, and its expression is up-regulated in obese and diabetic mice and humans. LCN2 expression and secretion have been shown to be induced by two proinflammatory cytokines, IFNγ and TNFα, in cultured murine and human adipocytes. In these studies, we demonstrated that IFNγ and TNFα induced LCN2 expression and secretion in vivo. Although we observed a strong induction of LCN2 expression and secretion from white adipose tissue (WAT) depots, the induction of LCN2 varied among different insulin-sensitive tissues. Knockdown experiments also demonstrated that STAT1 is required for IFNγ-induced lipocalin-2 expression in murine adipocytes. Similarly, knockdown of p65 in adipocytes demonstrated the necessity of the NF-κB signaling pathway for TNFα-mediated effects on LCN2. Activation of ERKs by IFNγ and TNFα also affected STAT1 and NF-κB signaling through modulation of serine phosphorylation. ERK activation-induced serine phosphorylation of both STAT1 and p65 mediated the additive effects of IFNγ and TNFα on LCN2 expression. Our results suggest that these same mechanisms occur in humans as we observed STAT1 and NF-κB binding to the human LCN2 promoter in chromatin immunoprecipitation assays performed in human fat cells. These studies substantially increase our knowledge regarding the requirements and mechanisms used by proinflammatory cytokines to induce LCN2 expression.
Collapse
Affiliation(s)
- Peng Zhao
- From the Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana 70803 and Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808
| | | | | |
Collapse
|