1
|
Reesor M, Goudiaby Y, Grossett N, Zand N, Chichester R, Echevarria-Javier L, Vysochyn M, Alam A. Effect of Hyperinsulinemia on Leptin and Ghrelin Levels in Polycystic Ovarian Syndrome: A Meta-Analysis. Cureus 2024; 16:e69023. [PMID: 39385878 PMCID: PMC11464008 DOI: 10.7759/cureus.69023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2024] [Indexed: 10/12/2024] Open
Abstract
Polycystic ovarian syndrome (PCOS) is the most common endocrine disorder in women of reproductive age. Leptin and ghrelin are important markers in PCOS due to their correlation with obesity, insulin resistance, and fertility. There is currently a debate in the literature about whether altered leptin and ghrelin levels in women with PCOS are a result of the disease itself or if they are due to factors such as the hyperinsulinemic state characteristic of PCOS. This meta-analysis aims to assess if insulin levels impact leptin and ghrelin levels in PCOS. Eight case-control studies assessing the relationship between insulin and leptin, as well as five case-control studies assessing the relationship between insulin and ghrelin, were identified in PubMed. Pearson's correlation coefficient (PCC) and the sample size were extracted, and two meta-analyses were conducted using a random-effects model. Total heterogeneity (I2) with a confidence interval of 95% was then determined. "Leave-one out" diagnostics were calculated for each case. If a study was identified as being significantly influential, the study was removed from the data set, and the trim and fill procedure was applied. Publication bias was assessed using Egger's regression test and rank correlation test. Our results showed a moderate positive relationship (r=0.56, 95% confidence interval (CI) (0.42, 0.71), with substantial heterogeneity I2=81.35%, 95% CI (25.2799, 88.2451)) between insulin and leptin levels, and a moderate negative relationship (r=-0.33, 95% CI (-0.43, -0.24)), with low heterogeneity (I2=0.00%, 95% CI (0.0000, 80.8159)) between insulin and ghrelin levels. Therefore, there is a significant relationship between insulin and higher leptin and lower ghrelin levels in women with PCOS. Better insulin control may have a positive effect on fertility, appetite, weight, body image, and quality of life in these women. This correlation is likely multifactorial, and further studies are needed to isolate factors influencing these hormones.
Collapse
Affiliation(s)
- Mikyla Reesor
- Clinical Sciences, Saint James School of Medicine, The Valley, AIA
| | - Yvette Goudiaby
- Clinical Sciences, Saint James School of Medicine, The Valley, AIA
| | - Nicole Grossett
- Clinical Sciences, Saint James School of Medicine, The Valley, AIA
| | - Natasha Zand
- Clinical Sciences, Saint James School of Medicine, The Valley, AIA
| | - Royon Chichester
- Clinical Sciences, Saint James School of Medicine, The Valley, AIA
| | | | | | - Amy Alam
- Clinical Sciences, Saint James School of Medicine, The Valley, AIA
| |
Collapse
|
2
|
Hu Y, Liu L, Chen Y, Zhang X, Zhou H, Hu S, Li X, Li M, Li J, Cheng S, Liu Y, Xu Y, Yan W. Cancer-cell-secreted miR-204-5p induces leptin signalling pathway in white adipose tissue to promote cancer-associated cachexia. Nat Commun 2023; 14:5179. [PMID: 37620316 PMCID: PMC10449837 DOI: 10.1038/s41467-023-40571-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023] Open
Abstract
Cancer-associated cachexia is a multi-organ weight loss syndrome, especially with a wasting disorder of adipose tissue and skeletal muscle. Small extracellular vesicles (sEVs) serve as emerging messengers to connect primary tumour and metabolic organs to exert systemic regulation. However, whether and how tumour-derived sEVs regulate white adipose tissue (WAT) browning and fat loss is poorly defined. Here, we report breast cancer cell-secreted exosomal miR-204-5p induces hypoxia-inducible factor 1A (HIF1A) in WAT by targeting von Hippel-Lindau (VHL) gene. Elevated HIF1A protein induces the leptin signalling pathway and thereby enhances lipolysis in WAT. Additionally, exogenous VHL expression blocks the effect of exosomal miR-204-5p on WAT browning. Reduced plasma phosphatidyl ethanolamine level is detected in mice lack of cancer-derived miR-204-5p secretion in vivo. Collectively, our study reveals circulating miR-204-5p induces hypoxia-mediated leptin signalling pathway to promote lipolysis and WAT browning, shedding light on both preventive screenings and early intervention for cancer-associated cachexia.
Collapse
Affiliation(s)
- Yong Hu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430062, China
| | - Liu Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
| | - Yong Chen
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
| | - Xiaohui Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
| | - Haifeng Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
| | - Sheng Hu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
| | - Xu Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
| | - Meixin Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
| | - Juanjuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Siyuan Cheng
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430062, China
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences; TaiKang Center for Life and Medical Sciences; The Institute for Advanced Studies; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, Hubei, 430072, China
| | - Yancheng Xu
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430062, China.
| | - Wei Yan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China.
| |
Collapse
|
3
|
Topete MV, Andrade S, Bernardino RL, Guimarães M, Pereira AM, Oliveira SB, Costa MM, Nora M, Monteiro MP, Pereira SS. Visceral Adipose Tissue Bioenergetics Varies According to Individuals' Obesity Class. Int J Mol Sci 2023; 24:ijms24021679. [PMID: 36675195 PMCID: PMC9863201 DOI: 10.3390/ijms24021679] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Obesity is associated with complex adipose tissue energy metabolism remodeling. Whether AT metabolic reprogramming differs according to body mass index (BMI) and across different obesity classes is unknown. This study’s purpose was to evaluate and compare bioenergetics and energy substrate preference of visceral adipose tissue (VAT) pertaining to individuals with obesity class 2 and class 3. VAT obtained from patients with obesity (n = 15) class 2 (n = 7; BMI 37.53 ± 0.58 kg/m2) or class 3 (n = 8; BMI 47.79 ± 1.52 kg/m2) was used to assess oxygen consumption rate (OCR) bioenergetics and mitochondrial substrate preferences. VAT of patients with obesity class 3 presented significantly higher non-mitochondrial oxygen consumption (p < 0.05). In VAT of patients with obesity class 2, inhibition of pyruvate and glutamine metabolism significantly decreased maximal respiration and spare respiratory capacity (p < 0.05), while pyruvate and fatty acid metabolism inhibition, which renders glutamine the only available substrate, increased the proton leak with a protective role against oxidative stress (p < 0.05). In conclusion, VAT bioenergetics of patients with obesity class 2 depicts a greater dependence on glucose/pyruvate and glutamine metabolism, suggesting that patients within this BMI range are more likely to be responsive to interventions based on energetic substrate modulation for obesity treatment.
Collapse
Affiliation(s)
- Marcelo V. Topete
- UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- ITR-Laboratory of Integrative and Translocation Research in Population Health, Rua das Taipas 135, 4050-600 Porto, Portugal
| | - Sara Andrade
- UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- ITR-Laboratory of Integrative and Translocation Research in Population Health, Rua das Taipas 135, 4050-600 Porto, Portugal
| | - Raquel L. Bernardino
- UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- ITR-Laboratory of Integrative and Translocation Research in Population Health, Rua das Taipas 135, 4050-600 Porto, Portugal
| | - Marta Guimarães
- UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- ITR-Laboratory of Integrative and Translocation Research in Population Health, Rua das Taipas 135, 4050-600 Porto, Portugal
- Department of General Surgery, Hospital São Sebastião, Centro Hospitalar de Entre o Douro e Vouga, Rua Dr. Cândido Pinho, 4050-220 Santa Maia da Feira, Portugal
| | - Ana M. Pereira
- Department of General Surgery, Hospital São Sebastião, Centro Hospitalar de Entre o Douro e Vouga, Rua Dr. Cândido Pinho, 4050-220 Santa Maia da Feira, Portugal
- Hospital da Luz Arrábida, Praceta de Henrique Moreira 150, 4400-346 Vila Nova de Gaia, Portugal
| | - Sofia B. Oliveira
- UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- ITR-Laboratory of Integrative and Translocation Research in Population Health, Rua das Taipas 135, 4050-600 Porto, Portugal
| | - Madalena M. Costa
- UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- ITR-Laboratory of Integrative and Translocation Research in Population Health, Rua das Taipas 135, 4050-600 Porto, Portugal
| | - Mário Nora
- Department of General Surgery, Hospital São Sebastião, Centro Hospitalar de Entre o Douro e Vouga, Rua Dr. Cândido Pinho, 4050-220 Santa Maia da Feira, Portugal
- Hospital da Luz Arrábida, Praceta de Henrique Moreira 150, 4400-346 Vila Nova de Gaia, Portugal
| | - Mariana P. Monteiro
- UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- ITR-Laboratory of Integrative and Translocation Research in Population Health, Rua das Taipas 135, 4050-600 Porto, Portugal
- Hospital da Luz Arrábida, Praceta de Henrique Moreira 150, 4400-346 Vila Nova de Gaia, Portugal
| | - Sofia S. Pereira
- UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- ITR-Laboratory of Integrative and Translocation Research in Population Health, Rua das Taipas 135, 4050-600 Porto, Portugal
- Correspondence:
| |
Collapse
|
4
|
|
5
|
Vilariño-García T, Guadix P, Dorado-Silva M, Sánchez-Martín P, Pérez-Pérez A, Sánchez-Margalet V. Decreased Expression of Sam68 Is Associated with Insulin Resistance in Granulosa Cells from PCOS Patients. Cells 2022; 11:cells11182821. [PMID: 36139396 PMCID: PMC9496917 DOI: 10.3390/cells11182821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Polycystic ovary syndrome (PCOS) is a complex metabolic disorder associated with ovulatory dysfunction, hyperandrogenism, obesity, and insulin resistance, which leads to subfertility. PCOS is the most frequent metabolic disorder in women and the major cause of infertility. Susceptibility to developing PCOS is determined by a complex interaction between environmental and genetic factors. Although different mechanisms have been proposed to explain PCOS manifestations, defects in insulin actions or in the insulin signaling pathways are central in the pathogenesis of the syndrome. However, the mechanisms (molecular players and signaling pathways) underlying its primary origin still remain an unsolved issue. Current research is increasingly focusing on the discovery of novel biomarkers to further elucidate the complex pathophysiology of PCOS. Sam68, an RNA-binding protein, is recruited to insulin signaling, mediating different insulin actions. We aimed to investigate the role of Sam68 in insulin signaling and the possible implications of Sam68 in the insulin resistance in PCOS. MATERIALS AND METHODS Granulosa cells were taken from women with PCOS (n = 25) and healthy donors (n = 25) and, within the age range of 20 to 42 years, from GINEMED, Assisted Reproduction Centre, Seville, Spain. The Sam68 expression level was analyzed both by qPCR and immunoblot. Statistical significance was assessed by one-way ANOVA, followed by a post-hoc test. A p value of < 0.05 was considered statistically significant. RESULTS We found that insulin stimulation increases the phosphorylation and expression level of Sam68 in granulosa cells from normal donors. The downregulation of Sam68 expression resulted in a lower activation of both the MAPK and the PI3K pathways in response to insulin. Moreover, the granulosa cells from the women with PCOS presented a lower expression of Sam68, as well as insulin receptor and insulin receptor substrate-1 (IRS-1). In these cells, the overexpression of Sam68 resulted in an increased activation of both the MAPK and the PI3K pathways in response to insulin. CONCLUSIONS These results suggest the participation of Sam68 in insulin receptor signaling, mediating the insulin effect in granulosa cells, and they suggest the possible role of Sam68 in the insulin resistance of PCOS.
Collapse
Affiliation(s)
- Teresa Vilariño-García
- Department of Medical Biochemistry and Molecular Biology and Immunology, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Pilar Guadix
- Obstetrics and Gynecology Department, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
| | | | | | - Antonio Pérez-Pérez
- Department of Medical Biochemistry and Molecular Biology and Immunology, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
- Correspondence: (A.P.-P.); (V.S.-M.); Tel.: +95-4559-850 (A.P.-P.)
| | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology and Immunology, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
- Correspondence: (A.P.-P.); (V.S.-M.); Tel.: +95-4559-850 (A.P.-P.)
| |
Collapse
|
6
|
Chen P, Jia R, Liu Y, Cao M, Zhou L, Zhao Z. Progress of Adipokines in the Female Reproductive System: A Focus on Polycystic Ovary Syndrome. Front Endocrinol (Lausanne) 2022; 13:881684. [PMID: 35692386 PMCID: PMC9178087 DOI: 10.3389/fendo.2022.881684] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/19/2022] [Indexed: 12/16/2022] Open
Abstract
Adipose tissue, one type of loose connective tissue in the human body, maintains the primary task of energy storage. Adipose tissue is not only an energy reservoir but also plays a vital role as the largest endocrine organ of the whole body via releasing a variety of adipokines, which participate in many pathophysiological processes, such as energy metabolism regulation, glucose and lipid metabolism, and inflammation. Polycystic ovary syndrome (PCOS) is a disorder that mainly involves the female reproductive system, affecting women of childbearing age particularly. Insulin resistance (IR) and hyperandrogenemia (HA) have been implicated as a critical link involving the etiology and outcome of PCOS. A great deal of studies has bridged the gap between adipokines (such as Adiponectin, Chemerin, Metrnl, Apelin, Resistin, Visfatin, Leptin, Vaspin, Lipocalin 2, and Omentin) and reproductive fitness. In this review, we will focus on the adipokines' functions on PCOS and come up with some points of view on the basis of current research.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhiming Zhao
- Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
7
|
Vasilakos LK, Steinbrekera B, Santillan DA, Santillan MK, Brandt DS, Dagle D, Roghair RD. Umbilical Cord Blood Leptin and IL-6 in the Presence of Maternal Diabetes or Chorioamnionitis. Front Endocrinol (Lausanne) 2022; 13:836541. [PMID: 35197933 PMCID: PMC8859102 DOI: 10.3389/fendo.2022.836541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/17/2022] [Indexed: 12/05/2022] Open
Abstract
Diabetes during pregnancy is associated with elevated maternal insulin, leptin and IL-6. Within the placenta, IL-6 can further stimulate leptin production. Despite structural similarities and shared roles in inflammation, leptin and IL-6 have contrasting effects on neurodevelopment, and the relative importance of maternal diabetes or chorioamnionitis on fetal hormone exposure has not been defined. We hypothesized that there would be a positive correlation between IL-6 and leptin with progressively increased levels in pregnancies complicated by maternal diabetes and chorioamnionitis. To test this hypothesis, cord blood samples were obtained from 104 term infants, including 47 exposed to maternal diabetes. Leptin, insulin, and IL-6 were quantified by multiplex assay. Factors independently associated with hormone levels were identified by univariate and multivariate linear regression. Unlike IL-6, leptin and insulin were significantly increased by maternal diabetes. Maternal BMI and birth weight were independent predictors of leptin and insulin with birth weight the strongest predictor of leptin. Clinically diagnosed chorioamnionitis and neonatal sepsis were associated with increased IL-6 but not leptin. Among appropriate for gestational age infants without sepsis, IL-6 and leptin were strongly correlated (R=0.6, P<0.001). In summary, maternal diabetes and birth weight are associated with leptin while chorioamnionitis is associated with IL-6. The constraint of the positive association between leptin and IL-6 to infants without sepsis suggests that the term infant and placenta may have a limited capacity to increase cord blood levels of the neuroprotective hormone leptin in the presence of increased cord blood levels of the potential neurotoxin IL-6.
Collapse
Affiliation(s)
- Lauren K. Vasilakos
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Baiba Steinbrekera
- Department of Pediatrics, University of South Dakota, Sioux Falls, SD, United States
| | - Donna A. Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Mark K. Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Debra S. Brandt
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Daniel Dagle
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Robert D. Roghair
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- *Correspondence: Robert D. Roghair,
| |
Collapse
|
8
|
Rubinstein MM, Brown KA, Iyengar NM. Targeting obesity-related dysfunction in hormonally driven cancers. Br J Cancer 2021; 125:495-509. [PMID: 33911195 PMCID: PMC8368182 DOI: 10.1038/s41416-021-01393-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 03/05/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity is a risk factor for at least 13 different types of cancer, many of which are hormonally driven, and is associated with increased cancer incidence and morbidity. Adult obesity rates are steadily increasing and a subsequent increase in cancer burden is anticipated. Obesity-related dysfunction can contribute to cancer pathogenesis and treatment resistance through various mechanisms, including those mediated by insulin, leptin, adipokine, and aromatase signalling pathways, particularly in women. Furthermore, adiposity-related changes can influence tumour vascularity and inflammation in the tumour microenvironment, which can support tumour development and growth. Trials investigating non-pharmacological approaches to target the mechanisms driving obesity-mediated cancer pathogenesis are emerging and are necessary to better appreciate the interplay between malignancy, adiposity, diet and exercise. Diet, exercise and bariatric surgery are potential strategies to reverse the cancer-promoting effects of obesity; trials of these interventions should be conducted in a scientifically rigorous manner with dose escalation and appropriate selection of tumour phenotypes and have cancer-related clinical and mechanistic endpoints. We are only beginning to understand the mechanisms by which obesity effects cell signalling and systemic factors that contribute to oncogenesis. As the rates of obesity and cancer increase, we must promote the development of non-pharmacological lifestyle trials for the treatment and prevention of malignancy.
Collapse
Affiliation(s)
- Maria M. Rubinstein
- grid.51462.340000 0001 2171 9952Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Kristy A. Brown
- grid.5386.8000000041936877XDepartment of Biochemistry in Medicine, Weill Cornell Medical College, New York, NY USA
| | - Neil M. Iyengar
- grid.51462.340000 0001 2171 9952Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY USA
| |
Collapse
|
9
|
Korac A, Srdic-Galic B, Kalezic A, Stancic A, Otasevic V, Korac B, Jankovic A. Adipokine signatures of subcutaneous and visceral abdominal fat in normal-weight and obese women with different metabolic profiles. Arch Med Sci 2021; 17:323-336. [PMID: 33747267 PMCID: PMC7959090 DOI: 10.5114/aoms/92118] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 06/10/2018] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Metabolic syndrome arises from abnormal adipose function accompanied by insulin resistance. As early factors reflecting/impacting lipid storage dysfunction of adipose tissues, we sought to determine adipokine levels in subcutaneous and visceral adipose tissues (SAT and VAT). MATERIAL AND METHODS Gene and protein expression levels of leptin, adiponectin, and resistin were analysed in SAT and VAT of normal-weight and overweight/obese women, subclassified according to insulin resistance index, triglyceride, total, low-density lipoprotein (LDL) and high-density lipoprotein (HDL) cholesterol levels into metabolically healthy and "at risk" groups. RESULTS Compared with normal-weight women, obese women had higher serum leptin levels (p < 0.05), as well as increased leptin gene and protein expression in VAT. Conversely, expression levels of leptin were lower in SAT of obese women, and minor in the SAT of "at risk" groups of women, compared with weight-matched healthy groups. In addition, lower adiponectin levels were detected in SAT of metabolically healthy obese women (p < 0.01), and lower in SAT and VAT (p < 0.05) of "at risk" obese women compared to healthy, obese women. Significant differences in resistin levels were only observed in obese women; resistin gene expression was higher in VAT and SAT of obese, compared to normal-weight women. However, higher gene expression was not consistent with protein expression of resistin. CONCLUSIONS Low adiponectin in both examined adipose tissues and inappropriate leptin expression levels in SAT appear to be important characteristics of obesity-related metabolic syndrome. Intriguingly, this adipokine dysregulation is primary seen in SAT, suggesting that endocrine dysfunction in this abdominal depot may be an early risk sign of metabolic syndrome.
Collapse
Affiliation(s)
- Aleksandra Korac
- Center for Electron Microscopy, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Biljana Srdic-Galic
- Department of Anatomy, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Andjelika Kalezic
- Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ana Stancic
- Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Vesna Otasevic
- Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Bato Korac
- Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Jankovic
- Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
10
|
Abstract
Leptin is a hormone that plays a major role as mediator of long-term regulation of energy balance, suppressing food intake, and stimulating weight loss. More recently, important physiological roles other than controlling appetite and energy expenditure have been suggested for leptin, including neuroendocrine, reparative, reproductive, and immune functions. These emerging peripheral roles let hypothesize that leptin can modulate also cancer progression. Indeed, many studies have demonstrated that elevated chronic serum concentrations of leptin, frequently seen in obese subjects, represent a stimulatory signal for tumor growth. Current knowledge indicates that also different non-tumoral cells resident in tumor microenvironment may respond to leptin creating a favorable soil for cancer cells. In addition, leptin is produced also within the tumor microenvironment creating the possibility for paracrine and autocrine action. In this review, we describe the main mechanisms that regulate peripheral leptin availability and how leptin can shape tumor microenvironment.
Collapse
|
11
|
Dahan MH, Abbasi F, Reaven G. Relationship between surrogate estimates and direct measurement of insulin resistance in women with polycystic ovary syndrome. J Endocrinol Invest 2019; 42:987-993. [PMID: 30701438 PMCID: PMC6639126 DOI: 10.1007/s40618-019-01014-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/24/2019] [Indexed: 01/19/2023]
Abstract
PURPOSE To evaluate the relationship between surrogate estimates of insulin resistance and a direct measurement of insulin-mediated glucose uptake women with and without PCOS. METHODS Retrospective cohort study of 75 PCOS and 118 controls. Fasting plasma glucose and insulin concentrations, insulin resistance as determined by the insulin suppression test, calculation of multiple surrogate estimates of insulin resistance, total and free testosterone concentrations, and correlations between the direct measure and surrogate estimates of insulin resistance were evaluated. RESULT(S) Surrogate markers of insulin resistance were correlated to a variable, but statistically significant degree with the direct measure of insulin resistance in control population and the women with PCOS. There was no correlation between the surrogate estimates of insulin resistance and total or free plasma testosterone concentrations. CONCLUSION(S) The surrogate estimates of insulin resistance evaluated were significantly related to a direct measure of insulin resistance, and this was true of both the control population and women with PCOS. The magnitude of the relationship between the surrogate estimates and the direct measurement was comparable and not significantly altered by androgen levels. Fasting plasma insulin concentration seems to be at least as accurate as any other surrogate estimate, and is by far the simplest.
Collapse
Affiliation(s)
- M H Dahan
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, McGill University, Montreal, Canada.
| | - F Abbasi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Stanford University, Stanford, USA
| | - G Reaven
- Division of Cardiovascular Medicine, Department of Internal Medicine, Stanford University, Stanford, USA
| |
Collapse
|
12
|
Insulin and Insulin Receptors in Adipose Tissue Development. Int J Mol Sci 2019; 20:ijms20030759. [PMID: 30754657 PMCID: PMC6387287 DOI: 10.3390/ijms20030759] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 12/14/2022] Open
Abstract
Insulin is a major endocrine hormone also involved in the regulation of energy and lipid metabolism via the activation of an intracellular signaling cascade involving the insulin receptor (INSR), insulin receptor substrate (IRS) proteins, phosphoinositol 3-kinase (PI3K) and protein kinase B (AKT). Specifically, insulin regulates several aspects of the development and function of adipose tissue and stimulates the differentiation program of adipose cells. Insulin can activate its responses in adipose tissue through two INSR splicing variants: INSR-A, which is predominantly expressed in mesenchymal and less-differentiated cells and mainly linked to cell proliferation, and INSR-B, which is more expressed in terminally differentiated cells and coupled to metabolic effects. Recent findings have revealed that different distributions of INSR and an altered INSR-A:INSR-B ratio may contribute to metabolic abnormalities during the onset of insulin resistance and the progression to type 2 diabetes. In this review, we discuss the role of insulin and the INSR in the development and endocrine activity of adipose tissue and the pharmacological implications for the management of obesity and type 2 diabetes.
Collapse
|
13
|
Marques-Oliveira GH, Silva TM, Lima WG, Valadares HMS, Chaves VE. Insulin as a hormone regulator of the synthesis and release of leptin by white adipose tissue. Peptides 2018; 106:49-58. [PMID: 29953915 DOI: 10.1016/j.peptides.2018.06.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/20/2018] [Accepted: 06/24/2018] [Indexed: 02/09/2023]
Abstract
Leptin and its receptor are widely distributed in several tissues, mainly in white adipose tissue. The serum leptin is highly correlated with body mass index in rodents and humans, being documented that leptin levels reduces in the fasting state and increase during refeeding, similarly to insulin release by pancreatic islets. Insulin appears to increase leptin mRNA and protein expression and its release by adipocytes. Some studies have suggested that insulin acts through the activation of the transcription factors: sterol regulatory element binding protein 1 (SREBP1), CCAAT enhancer binding protein-α (C/EBP-α) and specificity protein 1 (Sp1). Insulin stimulates the release of preformed and newly synthesized leptin by adipocytes through its signaling cascade. Its effects are blocked by inhibitors of the insulin signaling pathway, as well as by inhibitors of protein synthesis and agents that increase the intracellular cAMP. The literature data suggest that chronic hyperinsulinemia increases serum leptin levels in humans and rodents. In this review, we summarized the most updated knowledge on the effects of insulin on serum leptin levels, presenting the cell mechanisms that control leptin synthesis and release by the white adipose tissue.
Collapse
Affiliation(s)
| | - Thaís Marques Silva
- Laboratory of Physiology, Federal University of São João del-Rei, Divinópolis, Minas Gerais, Brazil
| | - William Gustavo Lima
- Laboratory of Physiology, Federal University of São João del-Rei, Divinópolis, Minas Gerais, Brazil
| | | | - Valéria Ernestânia Chaves
- Laboratory of Physiology, Federal University of São João del-Rei, Divinópolis, Minas Gerais, Brazil.
| |
Collapse
|
14
|
Boudalia S, Belloir C, Miller ML, Canivenc-Lavier MC. Early endocrine disruptors exposure acts on 3T3-L1 differentiation and endocrine activity. ACTA ACUST UNITED AC 2017; 7:83-89. [PMID: 28752072 PMCID: PMC5524989 DOI: 10.15171/bi.2017.11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 02/06/2017] [Accepted: 03/13/2017] [Indexed: 11/09/2022]
Abstract
![]()
Introduction: Data from last years suggested that early exposure to endocrine disruptors (EDs) can predispose newborns to endocrine dysfunction of adipocytes, obesity, and associated disorders. The implication of EDs at low doses on adipocyte development has been poorly investigated. For instance, vinclozolin (V) is a dicarboximide fungicide widely used in agriculture since the 90's, alone or in mixture with genistein (G), an isoflavonoid from Leguminosae. This study aims to identify the effect of vinclozolin alone or with genistein, on adipose tissue properties using cell culture.
Methods: In steroid-free conditions, 3T3-L1 pre-adipocytes were induced to differentiate in the presence of EDs, singularly or in mixtures, for 2 days. DNA and triglyceride (TG) levels were measured on days 0, 2 and 8 of differentiation. Leptin secretion was measured only on the eighth day.
Results: We show that low doses of G (25 µM) and V (0.1 µM) inhibit pre-adipocytes differentiation. This inhibition has been represented by a decreasing in DNA content (µg/well) and decreasing in TG accumulation (mg/mL) in 3T3-L1 cells. Nevertheless, V increased the anti-adipogenic properties of G.
Conclusion: This study confirms that EDs singularly or in mixtures, introduced during early stages of life, could affect the differentiation and the endocrine activity of adipocytes, and can act as potential factors for obesity.
Collapse
Affiliation(s)
- Sofiane Boudalia
- INRA, UMR1324, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France.,CNRS, UMR6265, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France.,Université de Bourgogne, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France.,Département d'Ecologie et Génie de l'Environnement, Université 8 Mai 1945, Guelma, Algérie.,Laboratoire de Biologie, Eau et Environnement, Université 8 Mai 1945, Guelma, Algérie
| | - Christine Belloir
- INRA, UMR1324, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France.,CNRS, UMR6265, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France.,Université de Bourgogne, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France
| | - Marie-Louise Miller
- INRA, UMR1324, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France.,CNRS, UMR6265, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France.,Université de Bourgogne, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France
| | - Marie-Chantal Canivenc-Lavier
- INRA, UMR1324, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France.,CNRS, UMR6265, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France.,Université de Bourgogne, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France
| |
Collapse
|
15
|
Li Y, Liu Y, Chen G. Vitamin A status affects the plasma parameters and regulation of hepatic genes in streptozotocin-induced diabetic rats. Biochimie 2017; 137:1-11. [PMID: 28238841 DOI: 10.1016/j.biochi.2017.02.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 02/22/2017] [Indexed: 02/07/2023]
Abstract
Vitamin A (VA) status regulates metabolism in rats. Whether VA status and availability of retinoic acid (RA) contribute to the insulin-regulated hepatic gene expression remains to be determined. Zucker lean rats with VA sufficient (VAS) or VA deficient (VAD) status were treated with streptozotocin (STZ) to induce insulin-dependent diabetes. They were treated with saline (STZ-VAS-C or STZ-VAD-C), RA (STZ-VAS-RA or STZ-VAD-RA), insulin (STZ-VAS-INS or STZ-VAD-INS), or insulin + RA (STZ-VAS-INS + RA or STZ-VAD-INS + RA) for 3 h. Insulin and insulin + RA treatments reduced tail tip blood glucose, raised plasma insulin and suppressed plasma β-hydroxybutyrate levels in both STZ-VAD and STZ-VAS rats. STZ-VAD-INS and STZ-VAD-INS + RA rats had lower plasma glucose levels than STZ-VAD-C rats had. STZ-VAD-INS and STZ-VAD-INS + RA rats had higher plasma leptin level and lower glucagon level than STZ-VAD-C rats did. Insulin treatment induced Gck, Srebp-1c and Fas and suppressed Pck1 expression levels in the liver of STZ-VAS and STZ-VAD rats. Interestingly, insulin treatment inhibited Cyp26a1 expression in STZ-VAD, but not STZ-VAS rats, whereas RA treatment induced it in both. RA treatment induced Gck expression only in STZ-VAD rats. Insulin + RA treatment further induced the Cyp26a1 and Gck expressions in STZ-VAD rats. The Srebp-1c expression levels of STZ-VAD-INS and STZ-VAD-INS + RA rats were higher than that of STZ-VAS-INS and STZ-VAS-INS + RA rats. The changes of Gck mRNA and glucokinase protein were consistent. In STZ-induced diabetic rats, VA is not required for insulin-regulated Gck, Srebp-1c, Fas and Pck1 expression. However, VA status altered responses of certain genes (Cyp26a1 and Srebp-1c) to insulin treatment.
Collapse
Affiliation(s)
- Yang Li
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, TN, USA
| | - Yang Liu
- Department of Pharmaceutical Engineering, School of Life Science, Wuchang University of Technology, Wuhan, Hubei Province, 430223, China
| | - Guoxun Chen
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, TN, USA.
| |
Collapse
|
16
|
Polak K, Czyzyk A, Simoncini T, Meczekalski B. New markers of insulin resistance in polycystic ovary syndrome. J Endocrinol Invest 2017; 40:1-8. [PMID: 27473078 PMCID: PMC5206255 DOI: 10.1007/s40618-016-0523-8] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 07/21/2016] [Indexed: 02/08/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine-metabolic disorder in women of reproductive age. The diagnostic criteria include two out of three features: hyperandrogenism, polycystic ovaries on ultrasound and menstrual irregularities (Rotterdam Criteria 2003). PCOS patients are more vulnerable to develop diabetes, cardiovascular diseases and metabolic syndrome. Insulin resistance (IR) is prevalent in women with PCOS independently of obesity and is critically involved in reproductive and metabolic complications of the syndrome. Several tests have been developed to measure IR, some very reliable but complex like the hyperinsulinemic euglycemic glucose clamp and others less precise but easier and less invasive like HOMA-IR. New markers are needed to reach a more reliable assessment of insulin resistance. To date, several surrogate markers have been proposed in the literature to facilitate and improve the determination of IR. Many new proteins are strongly involved with PCOS physiopathology and IR, such as some adipocytokines (adiponectin, visfatin, vaspin and apelin), copeptin, irisin, PAI-1 and zonulin. Many other proteins have been proposed as potential new markers of IR in PCOS, such as resistin, leptin, RBP4, kisspetin and ghrelin, but their role is still controversial. In this review, we provide a short characterization of these new markers, recently studied as indicators of metabolic state.
Collapse
Affiliation(s)
- K Polak
- Department of Clinical and Experimental Medicine, Division of Obstetrics and Gynecology, University of Pisa, Pisa, Italy
| | - A Czyzyk
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, ul. Polna 33, Poznan, Poland
| | - T Simoncini
- Department of Clinical and Experimental Medicine, Division of Obstetrics and Gynecology, University of Pisa, Pisa, Italy
| | - B Meczekalski
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, ul. Polna 33, Poznan, Poland.
| |
Collapse
|
17
|
Karimi K, Lindgren TH, Koch CA, Brodell RT. Obesity as a risk factor for malignant melanoma and non-melanoma skin cancer. Rev Endocr Metab Disord 2016; 17:389-403. [PMID: 27832418 DOI: 10.1007/s11154-016-9393-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The dramatic increases in incidence of both obesity and many cancers including skin cancer emphasize the need to better understand the pathophysiology of both conditions and their connections. Melanoma is considered the fastest growing cancer and rates of non-melanoma skin cancer have also increased over the last decade. The molecular mechanisms underlying the association between obesity and skin cancer are not clearly understood but emerging evidence points to changes in the tumor microenvironment including aberrant cell signaling and genomic instability in the chronic inflammatory state many obese individuals experience. This article reviews the literature linking obesity to melanoma and non-melanoma skin cancer.
Collapse
Affiliation(s)
- K Karimi
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - T H Lindgren
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - C A Koch
- Division of Endocrinology, University of Mississippi Medical Center, Jackson, MS, USA
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS, USA
| | - Robert T Brodell
- Department of Dermatology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA.
- Department of Dermatology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
18
|
Kuroda M, Tominaga A, Nakagawa K, Nishiguchi M, Sebe M, Miyatake Y, Kitamura T, Tsutsumi R, Harada N, Nakaya Y, Sakaue H. DNA Methylation Suppresses Leptin Gene in 3T3-L1 Adipocytes. PLoS One 2016; 11:e0160532. [PMID: 27494408 PMCID: PMC4975473 DOI: 10.1371/journal.pone.0160532] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/19/2016] [Indexed: 01/09/2023] Open
Abstract
Leptin is a key regulator of energy intake and expenditure. This peptide hormone is expressed in mouse white adipose tissue, but hardly expressed in 3T3-L1 adipocytes. Using bisulfite sequencing, we found that CpG islands in the leptin promoter are highly methylated in 3T3-L1cells. 5-azacytidine, an inhibitor of DNA methyltransferase, markedly increased leptin expression as pre-adipocytes matured into adipocytes. Remarkably, leptin expression was stimulated by insulin in adipocytes derived from precursor cells exposed to 5-azacytidine, but suppressed by thiazolidinedione and dexamethasone. In contrast, adipocytes derived from untreated precursor cells were unresponsive to both 5-azacytidine and hormonal stimuli, although lipid accumulation was sufficient to boost leptin expression in the absence of demethylation. Taken together, the results suggest that leptin expression in 3T3-L1 cells requires DNA demethylation prior to adipogenesis, transcriptional activation during adipogenesis, and lipid accumulation after adipogenesis.
Collapse
Affiliation(s)
- Masashi Kuroda
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima-city, Tokushima, Japan
- Research Fellow of Japan Society for the Promotion of Science, Chiyoda-ku, Tokyo, Japan
| | - Ayako Tominaga
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima-city, Tokushima, Japan
| | - Kasumi Nakagawa
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima-city, Tokushima, Japan
| | - Misa Nishiguchi
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima-city, Tokushima, Japan
| | - Mayu Sebe
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima-city, Tokushima, Japan
| | - Yumiko Miyatake
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima-city, Tokushima, Japan
| | - Tadahiro Kitamura
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | - Rie Tsutsumi
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima-city, Tokushima, Japan
| | - Nagakatsu Harada
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima-city, Tokushima, Japan
| | - Yutaka Nakaya
- Cardiovascular Medicine, Shikoku Central Hospital of the Mutual aid Association of Public School Teachers, Shikokuchuo-city, Ehime, Japan
| | - Hiroshi Sakaue
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima-city, Tokushima, Japan
- Diabetes Therapeutics and Research Center, Tokushima University, Tokushima-city, Tokushima, Japan
- * E-mail:
| |
Collapse
|
19
|
Noll C, Labbé SM, Pinard S, Shum M, Bilodeau L, Chouinard L, Phoenix S, Lecomte R, Carpentier AC, Gallo-Payet N. Postprandial fatty acid uptake and adipocyte remodeling in angiotensin type 2 receptor-deficient mice fed a high-fat/high-fructose diet. Adipocyte 2016; 5:43-52. [PMID: 27144096 DOI: 10.1080/21623945.2015.1115582] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 10/20/2015] [Accepted: 10/27/2015] [Indexed: 12/16/2022] Open
Abstract
The role of the angiotensin type-2 receptor in adipose physiology remains controversial. The aim of the present study was to demonstrate whether genetic angiotensin type-2 receptor-deficiency prevents or worsens metabolic and adipose tissue morphometric changes observed following a 6-week high-fat/high-fructose diet with injection of a small dose of streptozotocin. We compared tissue uptake of nonesterified fatty acid and dietary fatty acid in wild-type and angiotensin type-2 receptor-deficient mice by using the radiotracer 14(R,S)-[(1) (8)F]-fluoro-6-thia-heptadecanoic acid in mice fed a standard or high-fat diet. Postprandial fatty acid uptake in the heart, liver, skeletal muscle, kidney and adipose tissue was increased in wild-type mice after a high-fat diet and in angiotensin type-2 receptor-deficient mice on both standard and high-fat diets. Compared to the wild-type mice, angiotensin type-2 receptor-deficient mice had a lower body weight, an increase in fasting blood glucose and a decrease in plasma insulin and leptin levels. Mice fed a high-fat diet exhibited increased adipocyte size that was prevented by angiotensin type-2 receptor-deficiency. Angiotensin type-2 receptor-deficiency abolished the early hypertrophic adipocyte remodeling induced by a high-fat diet. The small size of adipocytes in the angiotensin type-2 receptor-deficient mice reflects their inability to store lipids and explains the increase in fatty acid uptake in non-adipose tissues. In conclusion, a genetic deletion of the angiotensin type-2 receptor is associated with metabolic dysfunction of white adipose depots, and indicates that adipocyte remodeling occurs before the onset of insulin resistance in the high-fat fed mouse model.
Collapse
|
20
|
Nasrat H, Patra SK, Goswami B, Jain A, Raghunandan C. Study of Association of Leptin and Insulin Resistance Markers in Patients of PCOS. Indian J Clin Biochem 2015; 31:104-7. [PMID: 26855496 DOI: 10.1007/s12291-015-0499-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 04/12/2015] [Indexed: 10/23/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrinological disorder among women of the reproductive age group with long term sequelae which include diabetes mellitus, hypertension and CAD. The present study was conducted to evaluate the association of leptin-an adipokine playing an important role in carbohydrate metabolism and markers of insulin resistance among women with PCOS. Sixty diagnosed cases of PCOS as per Rotterdam criteria were enrolled in this study after informed written consent. Insulin resistance was estimated using the homeostatic model assessment-insulin resistance (HOMA-IR). HOMA-IR was calculated as the product of the fasting plasma insulin value (mU/ml) and the fasting plasma glucose value (mg/dl), divided by 405 and HOMA β was calculated as 360 × [insulin]/([glucose] - 63) % (glucose in mg/dl). Estimation of serum leptin levels was done by ELISA using leptin ELISA kit from (DRG). A positive correlation of serum leptin levels was observed with markers of insulin resistance. Multiple regression analysis with HOMA-IR as dependent variable demonstrated a statistically significant contribution of fasting insulin levels. This study highlights the role of leptin in alterations in carbohydrate metabolism in patients with PCOS.
Collapse
Affiliation(s)
- Huma Nasrat
- Department of Biochemistry, Lady Hardinge Medical College, New Delhi, India
| | - Surajeet K Patra
- Department of Biochemistry, Lady Hardinge Medical College, New Delhi, India
| | - Binita Goswami
- Department of Biochemistry, Lady Hardinge Medical College, New Delhi, India
| | - Anju Jain
- Department of Biochemistry, Lady Hardinge Medical College, New Delhi, India
| | - Chitra Raghunandan
- Department of Obstetrics & Gynecology, Lady Hardinge Medical College, New Delhi, India
| |
Collapse
|
21
|
Uzun H, Bitik O, Baltu Y, Sönmez Ç, Öztürk Kaymak A. The Effects of Reduction Mammaplasty on Serum Leptin Levels and Insulin Resistance. Int J Endocrinol 2015; 2015:719824. [PMID: 26550014 PMCID: PMC4621362 DOI: 10.1155/2015/719824] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 10/04/2015] [Indexed: 11/21/2022] Open
Abstract
Background. The reduction mammaplasty has been a well-executed and known procedure in which considerable amount of fatty tissue is removed from the body. The authors aimed to show the effects of the reduction mammaplasty on serum leptin levels and insulin resistance. Methods. 42 obese female patients who had gigantomastia were operated on. We recorded patients' demographic and preoperative data, including age, weight, height, and body mass index. Fasting serum leptin, glucose, and insulin levels were noted. Homeostasis model assessment scores were calculated. At the postoperative 8th week, patients were reevaluated in terms of above parameters assessing the presence of any difference. Results. Serum leptin levels were decreased postoperatively and the decrease was statistically significant. We were able to show a decrease in homeostasis model assessment score, which indicated an increase in insulin sensitivity, and this change was statistically significant. A significant correlation between body mass index and leptin change was found postoperatively. Conclusion. Reduction mammaplasty is not solely an aesthetic procedure but it decreases serum leptin levels and increases insulin sensitivity, which may help obese women to reduce their cardiovascular risk.
Collapse
Affiliation(s)
- Hakan Uzun
- Department of Plastic Reconstructive and Aesthetic Surgery, Hacettepe University Faculty of Medicine, Sıhhiye, 06100 Ankara, Turkey
- *Hakan Uzun:
| | - Ozan Bitik
- Department of Plastic Reconstructive and Aesthetic Surgery, Hacettepe University Faculty of Medicine, Sıhhiye, 06100 Ankara, Turkey
| | - Yahya Baltu
- Department of Plastic Reconstructive and Aesthetic Surgery, Ankara Oncology Research and Training Hospital, Yenimahalle, 06200 Ankara, Turkey
| | - Çiğdem Sönmez
- Department of Biochemistry, Ankara Oncology Research and Training Hospital, Yenimahalle, 06200 Ankara, Turkey
| | - Ayşegül Öztürk Kaymak
- Department of Genetics, Ankara Oncology Research and Training Hospital, Yenimahalle, 06200 Ankara, Turkey
| |
Collapse
|
22
|
Upadhyay V, Fu YX. Lymphotoxin organizes contributions to host defense and metabolic illness from innate lymphoid cells. Cytokine Growth Factor Rev 2014; 25:227-33. [PMID: 24411493 PMCID: PMC3999173 DOI: 10.1016/j.cytogfr.2013.12.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 12/15/2013] [Indexed: 01/02/2023]
Abstract
The lymphotoxin (LT)-pathway is a unique constituent branch of the Tumor Necrosis Superfamily (TNFSF). Use of LT is a critical mechanism by which fetal innate lymphoid cells regulate lymphoid organogenesis. Within recent years, adult innate lymphoid cells have been discovered to utilize this same pathway to regulate IL-22 and IL-23 production for host defense. Notably, genetic studies have linked polymorphisms in the genes encoding LTα to several phenotypes contributing to metabolic syndrome. The role of the LT-pathway may lay the foundation for a bridge between host immune response, microbiota, and metabolic syndrome. The contribution of the LT-pathway to innate lymphoid cell function and metabolic syndrome will be visited in this review.
Collapse
Affiliation(s)
- Vaibhav Upadhyay
- Committee on Immunology, University of Chicago, United States; Department of Pathology, University of Chicago, United States
| | - Yang-Xin Fu
- Committee on Immunology, University of Chicago, United States; Department of Pathology, University of Chicago, United States.
| |
Collapse
|
23
|
|
24
|
Chen H, Zhang JP, Huang H, Wang ZH, Cheng R, Cai WB. Leptin promotes fetal lung maturity and upregulates SP-A expression in pulmonary alveoli type-II epithelial cells involving TTF-1 activation. PLoS One 2013; 8:e69297. [PMID: 23894445 PMCID: PMC3718688 DOI: 10.1371/journal.pone.0069297] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 06/12/2013] [Indexed: 11/19/2022] Open
Abstract
The placental hormone leptin has important functions in fetal and neonatal growth, and prevents depressed respiration in leptin-deficient mice. The effect of leptin on respiratory distress suffered by low birth weight and premature infants has been studied. However, it is unclear how leptin enhances lung maturity in the fetus and ameliorates neonatal respiratory distress. In the present study, we found that antenatal treatment with leptin for 2 d significantly enhanced the relative alveolus area and improved the maturity of fetal lungs in a rat model of fetal growth restriction (FGR). Mean birth weight and lung wet weight were higher in the leptin-treated group than in the PBS-treated group, indicating promotion of fetal growth. Leptin upregulated the intracellular expression and extracellular secretion of surfactant protein (SP) A in type-II alveolar epithelial cells (AECs) in vivo and in vitro. Dual positive effects of leptin were found on protein expression and transcriptional activity of thyroid transcription factor-1 (TTF-1), a nuclear transcription essential for branching morphogenesis of the lung and expression of SP-A in type-II AECs. Knockdown of TTF-1 by RNA interference indicated that TTF-1 may play a vital role in leptin-induced SP-A expression. These results suggest that leptin may have great therapeutic potential for the treatment of FGR, and leptin-mediated SP-A induction and lung maturity of the fetus are TTF-1 dependent.
Collapse
Affiliation(s)
- Hui Chen
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, China
| | - Jian-Ping Zhang
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, China
| | - Hui Huang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhen-Hua Wang
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Rui Cheng
- Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, China
| | - Wei-Bin Cai
- Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, China
- Center for Disease Model Animals, Sun Yat-sen University, Guangzhou, China
- * E-mail:
| |
Collapse
|
25
|
Dutta D, Ghosh S, Pandit K, Mukhopadhyay P, Chowdhury S. Leptin and cancer: Pathogenesis and modulation. Indian J Endocrinol Metab 2012; 16:S596-S600. [PMID: 23565495 PMCID: PMC3602989 DOI: 10.4103/2230-8210.105577] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Leptin, a product of Ob gene from adipocytes regulates appetite, energy expenditure and body mass composition by decreasing orexigenic and increasing anorexigenic neuropeptide release from hypothalamus. Research over the past few years have suggested leptin/leptin receptor dysregulation to have a role in the development of a large variety of malignancies like breast ca, thyroid ca, endometrial ca and gastrointestinal malignancies, predominantly through JAK/STAT pathway which modulates PI3K/AKT3 signaling, ERK1/2 signaling, expression of antiapoptotic proteins (like XIAP), systemic inflammation (TNF-α, IL6), angiogenic factors (VEGF) and hypoxia inducible factor-1a (HIF-1a) expression. In this review, the current understanding of leptin's role in carcinogenesis has been elaborated. Also a few agents modulating leptin signaling to inhibit cancer cell growth has been described.
Collapse
Affiliation(s)
- Deep Dutta
- Department of Endocrinology and Metabolism, Institute of Post Graduate Medical Education and Research Kolkata, Kolkata, India
| | - Sujoy Ghosh
- Department of Endocrinology and Metabolism, Institute of Post Graduate Medical Education and Research Kolkata, Kolkata, India
| | - Kaushik Pandit
- Department of Endocrinology and Metabolism, Institute of Post Graduate Medical Education and Research Kolkata, Kolkata, India
| | - Pradip Mukhopadhyay
- Department of Endocrinology and Metabolism, Institute of Post Graduate Medical Education and Research Kolkata, Kolkata, India
| | - Subhankar Chowdhury
- Department of Endocrinology and Metabolism, Institute of Post Graduate Medical Education and Research Kolkata, Kolkata, India
| |
Collapse
|
26
|
Ishihara Y, White CL, Kageyama H, Kageyama A, York DA, Bray GA. Effects of Diet and Time of the Day on Serum and CSF Leptin Levels in Osborne-Mendel and S5B/Pl Rats. ACTA ACUST UNITED AC 2012; 12:1067-76. [PMID: 15292470 DOI: 10.1038/oby.2004.134] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To characterize the effects of dietary fat on the diurnal variation in serum and cerebrospinal fluid (CSF) leptin levels in Osborne-Mendel (OM) and S5B/Pl rats and quantitate the dose response to lower doses of leptin administered into the third cerebral ventricle. RESEARCH METHODS AND PROCEDURES Rats were fitted with implanted vascular ports or third ventricular cannulas and fed either laboratory chow or one of two semipurified high-fat or low-fat diets. Leptin and insulin were measured by immunoassay. RESULTS Serum leptin and insulin levels were positively correlated and had similar patterns of diurnal change. CSF leptin and insulin also had diurnal rhythms, with a peak at 7:00 am, but the diurnal oscillations of leptin and insulin were significantly lower in the S5B/Pl rats than the OM rats. Thus, the ratio of CSF to serum leptin was significantly higher in the S5B/Pl rats than in the OM rats. Dietary fat had no effect on these diurnal patterns. There was a right shift in the dose response to leptin in the OM rats compared with the S5B/P1 rats. S5B/P1 rats treated with leptin had higher signal transduction and translation (STAT-3) mRNA levels compared with pair-fed or saline injected S5B/P1 rats. Hypothalamic suppressors of cytokine signaling mRNA levels were not statistically different between the groups. DISCUSSION The higher CSF-to-serum leptin ratio in the S5B/P1 rats, the enhanced suppression of food intake and body weight with leptin injections, and the higher STAT-3 activity in these animals suggest that S5B/P1 rats are more sensitive to leptin than OM rats.
Collapse
Affiliation(s)
- Yuri Ishihara
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge 70808, USA
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
Excessive caloric intake without a rise in energy expenditure promotes adipocyte hyperplasia and adiposity. The rise in adipocyte number is triggered by signaling factors that induce conversion of mesenchymal stem cells (MSCs) to preadipocytes that differentiate into adipocytes. MSCs, which are recruited from the vascular stroma of adipose tissue, provide an unlimited supply of adipocyte precursors. Members of the BMP and Wnt families are key mediators of stem cell commitment to produce preadipocytes. Following commitment, exposure of growth-arrested preadipocytes to differentiation inducers [insulin-like growth factor 1 (IGF1), glucocorticoid, and cyclic AMP (cAMP)] triggers DNA replication and reentry into the cell cycle (mitotic clonal expansion). Mitotic clonal expansion involves a transcription factor cascade, followed by the expression of adipocyte genes. Critical to these events are phosphorylations of the transcription factor CCATT enhancer-binding protein β (C/EBPβ) by MAP kinase and GSK3β to produce a conformational change that gives rise to DNA-binding activity. "Activated" C/EBPβ then triggers transcription of peroxisome proliferator-activated receptor-γ (PPARγ) and C/EBPα, which in turn coordinately activate genes whose expression produces the adipocyte phenotype.
Collapse
Affiliation(s)
- Qi Qun Tang
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
28
|
Krishnan S, Newman JW, Hembrooke TA, Keim NL. Variation in metabolic responses to meal challenges differing in glycemic index in healthy women: Is it meaningful? Nutr Metab (Lond) 2012; 9:26. [PMID: 22458475 PMCID: PMC3352098 DOI: 10.1186/1743-7075-9-26] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 03/29/2012] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Established clinical tests are commonly used in disease diagnosis, but tools that enhance identification of metabolic dysfunctions are needed. This study was conducted to identify typical and atypical metabolite temporal patterns in response to paired meal challenge tests. DESIGN Metabolic responses to high and low glycemic index (GI) meals were tested in 24 healthy pre-menopausal women, aged 20-50 y, with BMI of 25-30 kg/m2 using a cross-over design. On test days, blood glucose, insulin, leptin and non-esterified fatty acids were measured after an overnight fasting, and for 8 h following test meal consumption. The data were range scaled, and multivariate statistics were used to assess the presence of distinct response groups to the meal challenge tests. RESULTS As expected, participants showed higher circulating glucose and insulin in response to the high GI compared to the low GI meal challenge. However, using range-scaling and Principal Component Analysis, three distinct groups were identified based on differential responses to the paired challenges. Members of the most populated group (n = 18) displayed little deviation from the expected response to the two meal challenges. Two minor groups (n = 3/group) with distinct responses were observed, one suggestive of sub-clinical insulin resistance, and the other suggestive of hyperleptinemia. CONCLUSIONS The differential responses of glucose, insulin and leptin to low and high glycemic test meals revealed three response groups. Dietary intervention studies traditionally evaluate group responses, and aim to identify the overall effect in the population studied. In contrast, our study analyzed the variance in the meal challenge responses, using an integrated physiological approach, rather than a reductionist approach. This phenotyping approach may be useful for detecting subclinical metabolic dysfunctions, and it could contribute to improved personalized nutrition management. This study is registered in ClinicalTrials.gov, record #200210295.
Collapse
Affiliation(s)
- Sridevi Krishnan
- Department of Nutrition, University of California, Davis, CA, USA
| | - John W Newman
- Department of Nutrition, University of California, Davis, CA, USA
- USDA, Agricultural Research Service, Western Human Nutrition Research Center, Obesity and Metabolism Research Unit, Davis, CA, USA
| | - Tara A Hembrooke
- Department of Nutrition, University of California, Davis, CA, USA
- Platinum Performance Inc, Buellton, CA, USA
| | - Nancy L Keim
- Department of Nutrition, University of California, Davis, CA, USA
- USDA, Agricultural Research Service, Western Human Nutrition Research Center, Obesity and Metabolism Research Unit, Davis, CA, USA
- USDA, ARS, Western Human Nutrition Research Center, 430 W. Health Sciences Drive, Davis, CA 95616, USA
| |
Collapse
|
29
|
Evans JJ, Anderson GM. Balancing ovulation and anovulation: integration of the reproductive and energy balance axes by neuropeptides. Hum Reprod Update 2012; 18:313-32. [DOI: 10.1093/humupd/dms004] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
30
|
Engineer DR, Garcia JM. Leptin in anorexia and cachexia syndrome. INTERNATIONAL JOURNAL OF PEPTIDES 2012; 2012:287457. [PMID: 22518191 PMCID: PMC3303568 DOI: 10.1155/2012/287457] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 10/25/2011] [Accepted: 10/28/2011] [Indexed: 12/31/2022]
Abstract
Leptin is a product of the obese (OB) gene secreted by adipocytes in proportion to fat mass. It decreases food intake and increases energy expenditure by affecting the balance between orexigenic and anorexigenic hypothalamic pathways. Low leptin levels are responsible for the compensatory increase in appetite and body weight and decreased energy expenditure (EE) following caloric deprivation. The anorexia-cachexia syndrome is a complication of many chronic conditions including cancer, chronic obstructive pulmonary disease, congestive heart failure, chronic kidney disease, and aging, where the decrease in body weight and food intake is not followed by a compensatory increase in appetite or decreased EE. Crosstalk between leptin and inflammatory signaling known to be activated in these conditions may be responsible for this paradox. This manuscript will review the evidence and potential mechanisms mediating changes in the leptin pathway in the setting of anorexia and cachexia associated with chronic diseases.
Collapse
Affiliation(s)
- Diana R. Engineer
- Division of Diabetes, Endocrinology and Metabolism, Michael E DeBakey Veterans Affairs Medical Center, Houston, TX 77030, USA
- Baylor College of Medicine, 2002 Holcombe Boulevored, Building 109, Room 210, Houston, TX 77030, USA
- Division of Diabetes, Department of Medicine, Endocrinology and Metabolism, St Luke's Episcopal Hospital, Houston, TX 77030, USA
| | - Jose M. Garcia
- Division of Diabetes, Endocrinology and Metabolism, Michael E DeBakey Veterans Affairs Medical Center, Houston, TX 77030, USA
- Baylor College of Medicine, 2002 Holcombe Boulevored, Building 109, Room 210, Houston, TX 77030, USA
- Huffington Center of Aging, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
31
|
Barone I, Catalano S, Gelsomino L, Marsico S, Giordano C, Panza S, Bonofiglio D, Bossi G, Covington KR, Fuqua SAW, Andò S. Leptin mediates tumor-stromal interactions that promote the invasive growth of breast cancer cells. Cancer Res 2012; 72:1416-27. [PMID: 22282662 DOI: 10.1158/0008-5472.can-11-2558] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Obesity confers risks to cancer development and progression but the mechanisms underlying these risks remain unclear. In this study, we identify a role for the obesity cytokine leptin, which has been implicated previously in breast cancer development, as a determinant for the tumor-promoting activity of cancer-associated fibroblasts (CAF) in both wild-type (WT) and K303R mutant estrogen receptor-α (ERα)-expressing breast cancer cells. Human CAFs stimulated a greater increase in the proliferation and migration of breast cancer cells expressing the K303R-ERα hyperactive receptor than WT-ERα-expressing cells. A concomitant increase was seen in leptin receptor isoform expression and activation of the leptin signaling pathway in cells expressing K303R-ERα compared with WT-ERα, correlating with leptin effects on cell growth, motility, and invasiveness in mutant cells. Epidermal growth factor and other factors secreted by K303R-ERα cells stimulated CAF proliferation, migration, and subsequent leptin secretion. Moreover, K303R-ERα expression generated a leptin hypersensitive phenotype in vivo. Together, our results reveal a bidirectional cross-talk between breast cancer cells and "educated" CAFs that drives tumor progression via leptin signaling. In elucidating a mechanism that connects obesity and cancer, these findings reinforce the concept that blocking cancer-stromal cell communication may represent an effective strategy for targeted therapy of breast cancer.
Collapse
Affiliation(s)
- Ines Barone
- Centro Sanitario, University of Calabria, Rende, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Krause S, Jondeau-Cabaton A, Dhimolea E, Soto AM, Sonnenschein C, Maffini MV. Dual regulation of breast tubulogenesis using extracellular matrix composition and stromal cells. Tissue Eng Part A 2011; 18:520-32. [PMID: 21919795 DOI: 10.1089/ten.tea.2011.0317] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Epithelial-mesenchymal interactions during embryogenesis are critical in defining the phenotype of tissues and organs. The initial elongation of the mammary bud represents a central morphological event requiring extensive epithelial-mesenchymal crosstalk. The precise mechanism orchestrating this outgrowth is still unknown and mostly animal models have been relied upon to explore this process. Highly tunable three-dimensional (3D) culture models are a complementary approach to address the question of phenotypic determination. Here, we used a 3D in vitro culture to study the roles of stromal cells and extracellular matrix components during mammary tubulogenesis. Fibroblasts, adipocytes, and type I collagen actively participated in this process, whereas reconstituted basement membrane inhibited tubulogenesis by affecting collagen organization. We conclude that biochemical and biomechanical signals mediate the interaction between cells and matrix components and are necessary to induce tubulogenesis in vitro.
Collapse
Affiliation(s)
- Silva Krause
- Department of Anatomy and Cellular Biology, School of Medicine, Tufts University, Boston, MA 02111, USA
| | | | | | | | | | | |
Collapse
|
33
|
Cold exposure down-regulates adiponutrin/PNPLA3 mRNA expression and affects its nutritional regulation in adipose tissues of lean and obese Zucker rats. Br J Nutr 2011; 107:1283-95. [PMID: 21914237 DOI: 10.1017/s000711451100434x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Adiponutrin/PNPLA3 is a protein highly produced in adipose tissue whose expression is under tight nutritional regulation. It possesses lipogenic/lipolytic capacity and, although adiponutrin polymorphisms are related to obesity, its physiological role is not clear. To help clarify its role, we studied the effect of acute cold exposure on adiponutrin mRNA expression in different adipose tissues of lean/obese Zucker rats subjected to feeding/fasting/refeeding. The effect of cold on the expression of key lipogenic enzymes and on uncoupling protein-1 (UCP1) was evaluated in selected adipose depots. Adiponutrin mRNA levels were also determined in the adipose tissue of isoprenaline-treated rats and in cultured adipocytes treated with noradrenaline, isoprenaline and a selective β3-adrenoceptor (AR) agonist. Adiponutrin expression was strongly down-regulated by cold in the different adipose depots in lean animals, while this down-regulation was impaired in obese rats. Adiponutrin pattern of expression in response to cold correlated positively with that of the lipogenic enzymes and negatively with UCP1 expression. Acute intraperitoneal administration of isoprenaline also produced a decrease in adiponutrin expression in adipose tissue. In vitro data suggest that adiponutrin's inhibitory effect could be mediated, at least in part, by the sympathetic system via β1/β2-AR. In addition, improvement in metabolic parameters related to obesity in cold-exposed animals was related to an improvement in adiponutrin nutritional regulation. Thus, cold inhibition of adiponutrin expression in adipose tissue (which correlates with the response of lipogenic enzymes) supports a physiological role for this protein in lipogenesis. Moreover, alterations in adiponutrin expression and regulation in adipose tissue are related to obesity.
Collapse
|
34
|
Petridou ET, Sergentanis TN, Antonopoulos CN, Dessypris N, Matsoukis IL, Aronis K, Efremidis A, Syrigos C, Mantzoros CS. Insulin resistance: an independent risk factor for lung cancer? Metabolism 2011; 60:1100-6. [PMID: 21251684 DOI: 10.1016/j.metabol.2010.12.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 12/03/2010] [Accepted: 12/08/2010] [Indexed: 12/15/2022]
Abstract
Insulin resistance is closely associated with numerous metabolic disorders. Although studies have supported the importance of insulin resistance in carcinogenesis, the existing data have not established its relevance in the context of lung cancer. The aim of the present case-control study was to evaluate the association between insulin resistance and lung cancer after adjusting for possible confounders. Homeostasis model assessment of insulin resistance (HOMA-IR) and serum leptin and adiponectin levels were determined in 81 lung cancer cases and 162 age- and sex-matched controls; anthropometric and lifestyle variables were recorded. Mean HOMA-IR in the cases was more than 2-fold higher compared with the mean value of controls (P < .001). Among controls, HOMA-IR correlated positively with serum leptin (r = 0.16; P = .04), body mass index (r = 0.43; P = .0001), and waist-to-hip ratio (r = 0.21; P = .01) but negatively with serum adiponectin (r = -0.29; P = .0002). As expected, smoking was associated with an approximately 10-fold increase in lung cancer risk in multiple logistic regression models. A positive association between HOMA-IR, treated as continuous variable, and lung cancer (odds ratio [OR] = 1.52, 95% confidence interval [CI]: 1.16-1.99, P = .002, model 1) was demonstrated, which persisted after adjustment for somatometric and lifestyle variables (OR = 2.36, 95% CI: 1.00-5.55, P = .05, model 2). When serum adiponectin was also taken into account, the association seemed fairly robust (OR = 2.58, 95% CI: 1.11-6.01, P = .03, model 3); on the contrary, when serum leptin was added, the association remained positive, but lost its statistical significance (OR = 1.76, 95% CI: 0.78-3.98, P = .17, model 4). In the fully adjusted model, HOMA-IR was still positively, but only marginally, associated with lung cancer risk (OR = 2.02, 95% CI: 0.88-4.65, P = .10, model 5). Insulin resistance may represent a meaningful risk factor for lung cancer.
Collapse
Affiliation(s)
- Eleni Th Petridou
- Department of Hygiene, Epidemiology and Medical Statistics, Athens University Medical School, 115 27 Athens, Greece.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Rigamonti A, Brennand K, Lau F, Cowan CA. Rapid cellular turnover in adipose tissue. PLoS One 2011; 6:e17637. [PMID: 21407813 PMCID: PMC3047582 DOI: 10.1371/journal.pone.0017637] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Accepted: 02/04/2011] [Indexed: 11/24/2022] Open
Abstract
It was recently shown that cellular turnover occurs within the human adipocyte population. Through three independent experimental approaches — dilution of an inducible histone 2B-green fluorescent protein (H2BGFP), labeling with the cell cycle marker Ki67 and incorporation of BrdU — we characterized the degree of cellular turnover in murine adipose tissue. We observed rapid turnover of the adipocyte population, finding that 4.8% of preadipocytes are replicating at any time and that between 1–5% of adipocytes are replaced each day. In light of these findings, we suggest that adipose tissue turnover represents a possible new avenue of therapeutic intervention against obesity.
Collapse
Affiliation(s)
- Alessandra Rigamonti
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts, United States of America
- Doctorate of Prenatal Science, Fetal Diagnosis and Therapy, University of Milan, Milan, Italy
| | - Kristen Brennand
- Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Frank Lau
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts, United States of America
| | - Chad A. Cowan
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
36
|
Ravanan P, Harry GJ, Awada R, Hoareau L, Tallet F, Roche R, d’Hellencourt CL. Exposure to an organometal compound stimulates adipokine and cytokine expression in white adipose tissue. Cytokine 2011; 53:355-62. [PMID: 21194965 PMCID: PMC3418814 DOI: 10.1016/j.cyto.2010.11.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Revised: 08/13/2010] [Accepted: 11/19/2010] [Indexed: 12/25/2022]
Abstract
OBJECTIVE White adipose tissue (WAT) is now considered a defined tissue capable of interactions with other organ systems. WAT role in elevating the level of systemic chronic inflammation suggests that alterations in this tissue as the result of disease or environmental factors may influence the development and progression of various obesity-related pathologies. This study investigated WAT cell-specific responses to an organometal compound, trimethyltin (TMT), to determine possible contribution to induced inflammation. METHODS Human primary mature adipocytes and macrophage differentiated THP-1 cells were cultured in TMT presence and relative toxicities and different adipokine levels were determined. The inflammatory response was examined in TMT presence for primary cells from obese ob/ob mice WAT, and after TMT injection in ob/ob mice. RESULTS Both adipocytes and macrophages were resistant to cell death induced by TMT. However, adipocytes cultured in TMT presence showed increased expression of TNFα and IL-6, and modified leptin levels. In macrophage cultures, TMT also increased TNFα and IL-6, while MCP-1 and MIP-1α were decreased. In vivo, a single injection of TMT in ob/ob mice, elevated TNFα, MIP-1α and adiponectin in WAT. CONCLUSIONS Elevation of the inflammatory related products can be induced by chemical exposure in adipocytes and macrophages, as well as murine WAT. These data suggest that numerous factors, including a systemic chemical exposure, can induce an inflammatory response from the WAT. Furthermore, when characterizing both chemical-induced toxicity and the progression of the chronic inflammation associated with elevated WAT content, such responses in this target tissue should be taken into consideration.
Collapse
Affiliation(s)
- Palaniyandi Ravanan
- Laboratoire de Biochimie et de Génétique Moléculaire, Groupe d’Etude de l’Inflammation Chronique et de l’Obésité (GEICO), Université de La Réunion, Faculté des Sciences, 15 avenue R. Cassin and Plateforme CYROI, 2 rue Maxime Rivière, 97490 Sainte Clotilde, France
| | - G. Jean Harry
- Neurotoxicology Group, Laboratory of Molecular Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Dept of Health and Human Services, Research Triangle Park, NC, USA
| | - Rana Awada
- Laboratoire de Biochimie et de Génétique Moléculaire, Groupe d’Etude de l’Inflammation Chronique et de l’Obésité (GEICO), Université de La Réunion, Faculté des Sciences, 15 avenue R. Cassin and Plateforme CYROI, 2 rue Maxime Rivière, 97490 Sainte Clotilde, France
| | - Laurence Hoareau
- Laboratoire de Biochimie et de Génétique Moléculaire, Groupe d’Etude de l’Inflammation Chronique et de l’Obésité (GEICO), Université de La Réunion, Faculté des Sciences, 15 avenue R. Cassin and Plateforme CYROI, 2 rue Maxime Rivière, 97490 Sainte Clotilde, France
| | - Frank Tallet
- Laboratoire de Biochimie UF4130, Centre Hospitalier Régional Félix Guyon, La Réunion, France
| | - Régis Roche
- Laboratoire de Biochimie et de Génétique Moléculaire, Groupe d’Etude de l’Inflammation Chronique et de l’Obésité (GEICO), Université de La Réunion, Faculté des Sciences, 15 avenue R. Cassin and Plateforme CYROI, 2 rue Maxime Rivière, 97490 Sainte Clotilde, France
| | - Christian Lefebvre d’Hellencourt
- Laboratoire de Biochimie et de Génétique Moléculaire, Groupe d’Etude de l’Inflammation Chronique et de l’Obésité (GEICO), Université de La Réunion, Faculté des Sciences, 15 avenue R. Cassin and Plateforme CYROI, 2 rue Maxime Rivière, 97490 Sainte Clotilde, France
| |
Collapse
|
37
|
Corr M, De Souza MJ, Toombs RJ, Williams NI. Circulating leptin concentrations do not distinguish menstrual status in exercising women. Hum Reprod 2011; 26:685-94. [DOI: 10.1093/humrep/deq375] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
38
|
|
39
|
Sánchez-Margalet V, González-Yanes C, Najib S, Santos-Álvarez J. Reprint of: Metabolic effects and mechanism of action of the chromogranin A-derived peptide pancreastatin. ACTA ACUST UNITED AC 2010; 165:71-7. [PMID: 20934461 DOI: 10.1016/j.regpep.2010.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Revised: 02/09/2010] [Accepted: 02/11/2010] [Indexed: 01/12/2023]
Abstract
Pancreastatin is one of the regulatory peptides derived from intracellular and/or extracellular processing of chromogranin A, the soluble acidic protein present in the secretory granules of the neuroendocrine system. While the intracellular functions of chromogranin A include formation and maturation of the secretory granule, the major extracellular functions are generation of biologically active peptides with demonstrated autocrine, paracrine or endocrine activities. In this review, we will focus on the metabolic function of one of these peptides, pancreastatin, and the mechanisms underlying its effects. Many different reported effects have implicated PST in the modulation of energy metabolism, with a general counterregulatory effect to that of insulin. Pancreastatin induces glycogenolysis in liver and lipolysis in adipocytes. Metabolic effects have been confirmed in humans. Moreover, naturally occurring human variants have been found, one of which (Gly297Ser) occurs in the functionally important carboxy-terminus of the peptide, and substantially increases the peptide's potency to inhibit cellular glucose uptake. Thus, qualitative hereditary alterations in pancreastatin's primary structure may give rise to interindividual differences in glucose and lipid metabolism. Pancreastatin activates a receptor signaling system that belongs to the seven-spanning transmembrane receptor coupled to a Gq-PLCβ-calcium-PKC signaling pathway. Increased pancreastatin plasma levels, correlating with catecholamines levels, have been found in insulin resistance states, such as gestational diabetes or essential hypertension. Pancreastatin plays important physiological role in potentiating the metabolic effects of catecholamines, and may also play a pathophysiological role in insulin resistance states with increased sympathetic activity.
Collapse
Affiliation(s)
- Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, Spain.
| | | | | | | |
Collapse
|
40
|
Shankar K, Kang P, Harrell A, Zhong Y, Marecki JC, Ronis MJJ, Badger TM. Maternal overweight programs insulin and adiponectin signaling in the offspring. Endocrinology 2010; 151:2577-89. [PMID: 20371699 PMCID: PMC2875830 DOI: 10.1210/en.2010-0017] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 03/11/2010] [Indexed: 12/15/2022]
Abstract
Gestational exposure to maternal overweight (OW) influences the risk of obesity in adult life. Male offspring from OW dams gain greater body weight and fat mass and develop insulin resistance when fed high-fat diets (45% fat). In this report, we identify molecular targets of maternal OW-induced programming at postnatal d 21 before challenge with the high-fat diet. We conducted global transcriptome profiling, gene/protein expression analyses, and characterization of downstream signaling of insulin and adiponectin pathways in conjunction with endocrine and biochemical characterization. Offspring born to OW dams displayed increased serum insulin, leptin, and resistin levels (P < 0.05) at postnatal d 21 preceding changes in body composition. A lipogenic transcriptome signature in the liver, before development of obesity, was evident in OW-dam offspring. A coordinated locus of 20 sterol regulatory element-binding protein-1-regulated target genes was induced by maternal OW. Increased nuclear levels of sterol regulatory element-binding protein-1 and recruitment to the fatty acid synthase promoter were confirmed via ELISA and chromatin immunoprecipitation analyses, respectively. Higher fatty acid synthase and acetyl coenzyme A carboxylase protein and pAKT (Thr(308)) and phospho-insulin receptor-beta were confirmed via immunoblotting. Maternal OW also attenuated AMP kinase/peroxisome proliferator-activated receptor-alpha signaling in the offspring liver, including transcriptional down-regulation of several peroxisome proliferator-activated receptor-alpha-regulated genes. Hepatic mRNA and circulating fibroblast growth factor-21 levels were significantly lower in OW-dam offspring. Furthermore, serum levels of high-molecular-weight adiponectin (P < 0.05) were decreased in OW-dam offspring. Phosphorylation of hepatic AMP-kinase (Thr(172)) was significantly decreased in OW-dam offspring, along with lower AdipoR1 mRNA. Our results strongly suggest that gestational exposure to maternal obesity programs multiple aspects of energy-balance regulation in the offspring.
Collapse
Affiliation(s)
- Kartik Shankar
- Arkansas Children's Nutrition Center, 15 Children's Way, Slot 512-20B, Little Rock, Arkansas 72202, USA.
| | | | | | | | | | | | | |
Collapse
|
41
|
Sánchez-Margalet V, González-Yanes C, Najib S, Santos-Alvarez J. Metabolic effects and mechanism of action of the chromogranin A-derived peptide pancreastatin. REGULATORY PEPTIDES 2010; 161:8-14. [PMID: 20184923 DOI: 10.1016/j.regpep.2010.02.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Revised: 02/09/2010] [Accepted: 02/11/2010] [Indexed: 12/20/2022]
Abstract
Pancreastatin is one of the regulatory peptides derived from intracellular and/or extracellular processing of chromogranin A, the soluble acidic protein present in the secretory granules of the neuroendocrine system. While the intracellular functions of chromogranin A include formation and maturation of the secretory granule, the major extracellular functions are generation of biologically active peptides with demonstrated autocrine, paracrine or endocrine activities. In this review, we will focus on the metabolic function of one of these peptides, pancreastatin, and the mechanisms underlying its effects. Many different reported effects have implicated PST in the modulation of energy metabolism, with a general counterregulatory effect to that of insulin. Pancreastatin induces glycogenolysis in liver and lipolysis in adipocytes. Metabolic effects have been confirmed in humans. Moreover, naturally occurring human variants have been found, one of which (Gly297Ser) occurs in the functionally important carboxy-terminus of the peptide, and substantially increases the peptide's potency to inhibit cellular glucose uptake. Thus, qualitative hereditary alterations in pancreastatin's primary structure may give rise to interindividual differences in glucose and lipid metabolism. Pancreastatin activates a receptor signaling system that belongs to the seven-spanning transmembrane receptor coupled to a Gq-PLCbeta-calcium-PKC signaling pathway. Increased pancreastatin plasma levels, correlating with catecholamines levels, have been found in insulin resistance states, such as gestational diabetes or essential hypertension. Pancreastatin plays important physiological role in potentiating the metabolic effects of catecholamines, and may also play a pathophysiological role in insulin resistance states with increased sympathetic activity.
Collapse
Affiliation(s)
- Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, Spain.
| | | | | | | |
Collapse
|
42
|
Abstract
BACKGROUND Leptin has physiological roles in multiple systems, and has possible effects on several carcinogenesis steps. The aim of this study was to investigate the leptin levels in thyroid papillary carcinoma (TPC) patients. METHODS Forty-three female TPC patients and 30 healthy female control subjects were recruited for the study. TPC was diagnosed by fine needle aspiration biopsy. TPC patients had a bilateral total thyroidectomy operation and their leptin levels were measured before and 20 days after the operation. RESULTS Serum leptin levels of TPC patients were higher than in control group subjects (21.15 +/- 14.12 ng/mL vs. 9.89 +/- 0.21 ng/mL, p < 0.05). The leptin levels decreased after total thyroidectomy (13.92 +/- 10.55 ng/mL) compared to prethyroidectomy levels (22.94 +/- 14.67 ng/mL) in 34 patients who came to the follow-up visit (p < 0.05). However, the decreased post-thyroidectomy levels of leptin were still statistically significantly higher than the control group levels. Multivariate regression analysis showed that the leptin levels in TPC patients were not related to age, menopausal status or pathologic occult status but were directly related to the cancer group. CONCLUSION Leptin levels were elevated in thyroid cancer, decreased after total thyroidectomy, and might be associated with thyroid papillary carcinogenesis.
Collapse
Affiliation(s)
- Melih Akinci
- Department of General Surgery, Ankara Diskapi Education and Research Hospital, Ankara, Turkey.
| | | | | | | | | | | |
Collapse
|
43
|
Luvizotto RAM, Conde SJ, Síbio MT, Nascimento AF, Lima-Leopoldo AP, Leopoldo AS, Padovani CR, Cicogna AC, Nogueira CR. Administration of physiologic levels of triiodothyronine increases leptin expression in calorie-restricted obese rats, but does not influence weight loss. Metabolism 2010; 59:1-6. [PMID: 19846169 DOI: 10.1016/j.metabol.2009.06.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Revised: 06/24/2009] [Accepted: 06/27/2009] [Indexed: 11/30/2022]
Abstract
Obesity has become a major public health problem, most commonly treated via dietary restriction to promote weight loss. Although leptin and thyroid hormones are involved in the regulation of energy balance, the role of these hormones after the stabilization of weight loss remains unclear. This study was designed to analyze the effect of thyroid hormone on sustained weight loss and leptin gene expression in obese animals after a loss of 5% to 10% of body weight. Thirty-day-old male Wistar rats were separated into 4 groups: control, obese, calorie restriction (CR), and calorie restriction with triiodothyronine administration (CRT). The obese group had increased weight and adiposity, leptin and insulin levels, and leptin gene expression. Dietary restriction in the CR group resulted in decreased body weight and adiposity, diminished leptin, and increased thyroid hormone receptor beta expression. The CRT group, submitted to dietary restriction with concomitant administration of a physiologic triiodothyronine dose, had thyroid hormone receptor beta expression at levels comparable with those observed in the control group and simultaneously increased leptin expression as compared with that in the CR group, suggesting that thyroid hormone modulates leptin expression under conditions of calorie restriction. Increased leptin expression in the CRT group did not result in increased circulating leptin or a statistically significant reduction in body weight during the treatment period. These data provide impetus for further study, as a longer treatment period may result in increased circulating leptin and, thus, further reduction in body weight during calorie restriction in an obesity model.
Collapse
Affiliation(s)
- Renata A M Luvizotto
- Department of Clinical Medicine, Botucatu School of Medicine, University of São Paulo State-UNESP, CEP: 18618-000 Botucatu, SP, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Hwang EY, Hong JH, Choi JH, Choi EJ, Lee IS. Study on Anti-obesity and Hypoglycemic Effects of Lycium chinense Mill Extracts. ACTA ACUST UNITED AC 2009. [DOI: 10.3746/jkfn.2009.38.11.1528] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
45
|
Ohlsson C, Mohan S, Sjögren K, Tivesten A, Isgaard J, Isaksson O, Jansson JO, Svensson J. The role of liver-derived insulin-like growth factor-I. Endocr Rev 2009; 30:494-535. [PMID: 19589948 PMCID: PMC2759708 DOI: 10.1210/er.2009-0010] [Citation(s) in RCA: 309] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Accepted: 07/01/2009] [Indexed: 02/08/2023]
Abstract
IGF-I is expressed in virtually every tissue of the body, but with much higher expression in the liver than in any other tissue. Studies using mice with liver-specific IGF-I knockout have demonstrated that liver-derived IGF-I, constituting a major part of circulating IGF-I, is an important endocrine factor involved in a variety of physiological and pathological processes. Detailed studies comparing the impact of liver-derived IGF-I and local bone-derived IGF-I demonstrate that both sources of IGF-I can stimulate longitudinal bone growth. We propose here that liver-derived circulating IGF-I and local bone-derived IGF-I to some extent have overlapping growth-promoting effects and might have the capacity to replace each other (= redundancy) in the maintenance of normal longitudinal bone growth. Importantly, and in contrast to the regulation of longitudinal bone growth, locally derived IGF-I cannot replace (= lack of redundancy) liver-derived IGF-I for the regulation of a large number of other parameters including GH secretion, cortical bone mass, kidney size, prostate size, peripheral vascular resistance, spatial memory, sodium retention, insulin sensitivity, liver size, sexually dimorphic liver functions, and progression of some tumors. It is clear that a major role of liver-derived IGF-I is to regulate GH secretion and that some, but not all, of the phenotypes in the liver-specific IGF-I knockout mice are indirect, mediated via the elevated GH levels. All of the described multiple endocrine effects of liver-derived IGF-I should be considered in the development of possible novel treatment strategies aimed at increasing or reducing endocrine IGF-I activity.
Collapse
Affiliation(s)
- Claes Ohlsson
- Division of Endocrinology, Institute of Medicine, Sahlgrenska University Hospital, Göteborg, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Briana DD, Malamitsi-Puchner A. Reviews: adipocytokines in normal and complicated pregnancies. Reprod Sci 2009; 16:921-37. [PMID: 19474287 DOI: 10.1177/1933719109336614] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Human pregnancy is characterized by insulin resistance, traditionally attributed to the effects of placental hormones. Normal pregnancy-induced insulin resistance is further enhanced in pregnancy complications, associated with disturbed placental function, such as gestational diabetes mellitus, preeclampsia, and intrauterine growth restriction. Compelling evidence suggests that these pregnancy disorders are associated with future development of maternal metabolic syndrome. However, the pathogenetic mechanisms underlying the association between abnormal placental development, insulin resistance, and maternal metabolic syndrome are not fully understood. A large body of evidence has recently supported the role of adipose tissue in the regulation of insulin resistance in both nonpregnant and pregnant participants. In this respect, adipocytokines, which are adipocyte-derived hormones, have been implicated in the regulation of maternal metabolism and gestational insulin resistance. Adipocytokines, including leptin, adiponectin, tumor necrosis factor alpha, interleukin 6, as well as the newly discovered resistin, visfatin, and apelin, are also known to be produced within the intrauterine environment. However, data concerning the pattern of adipocytokines secretion in normal and complicated pregnancies are still limited and partially contradictory. Given the importance of adipose tissue and its hormones in terms of adequate metabolic control and energy homeostasis, we present a review of published data related to the role of adipocytokines in pregnancy, especially in relation to pregnancy complications. Focus will be placed on the functions and other potential roles of the novel adipocytokines resistin, visfatin, and apelin.
Collapse
Affiliation(s)
- Despina D Briana
- Neonatal Division, 2nd Department of Obstetrics and Gynecology, Athens University Medical School, Athens, Greece
| | | |
Collapse
|
47
|
Harris RBS. Nature or nurture?: focus on "Preadipocyte transplantation: an in vivo study of direct leptin signaling on adipocyte morphogenesis and cell size". Am J Physiol Regul Integr Comp Physiol 2009; 296:R1336-8. [PMID: 19261917 DOI: 10.1152/ajpregu.00101.2009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
48
|
Liu TC, Liu YY, Lee SD, Huang CY, Chien KY, Cheng IS, Lin CY, Kuo CH. Effects of short-term detraining on measures of obesity and glucose tolerance in elite athletes. J Sports Sci 2008; 26:919-25. [PMID: 18569557 DOI: 10.1080/02640410801885925] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Athletes frequently adjust their training volume in line with their athletic competition schedule, onset of sport injury, and retirement. Whether maintenance of partial training activity during the detraining period can preserve optimal body composition and insulin sensitivity is currently unknown. Sixteen elite kayak athletes (mean VO2max: 58.5 ml.kg(-1).min(-1), s = 1.77) were randomly assigned to a totally detrained group (age: 20.8 years, s = 0.7; body mass index: 23.74, s = 0.54) or partially detrained group (age: 21.8 years, s = 0.7; body mass index: 23.20, s = 1.02), whereby totally detrained participants terminated their training routine completely and the partially detrained participants preserved approximately 50% of their previous training duration with equivalent intensity for one month. Body mass, waist circumference, oral glucose tolerance test, insulin, leptin, cortisol, and testosterone were measured during the trained state and after detraining. Waist circumferences for both the partially detrained and totally detrained groups were significantly elevated after detraining, with no group difference. However, body mass was reduced in both groups. Significant elevations in the area under the curve for insulin and fasted leptin with detraining were observed. These changes were greater in the totally detrained participants. In conclusion, the present results show that maintaining partial training activity cannot prevent an increase in waist circumference. During the detraining period, the magnitude of increase in plasma insulin and leptin concentrations was regulated in an activity-dependent manner.
Collapse
Affiliation(s)
- Te-Chih Liu
- Laboratory of Exercise Biochemistry, Taipei Physical Education College, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Obesity results from an abnormal accumulation of fat in the white adipose tissue. Recent research utilizing genetic models of obesity in rodents has implicated a major role of leptin as a controller of obesity. Leptin is a 167-amino acid peptide hormone encoded by the obesity gene (ob), which is secreted by adipocytes and plays an important role in regulating food intake, energy expenditure and adiposity. Leptin receptors (OB-R) are expressed in the central nervous system mainly in afferent satiety centres of hypothalamus and in peripheral organs such as adipose tissues, skeletal muscles, pancreatic beta-cells and liver, thus indicating the autocrine and paracrine role of leptin in energy regulation. In human beings, a highly organized circadian pattern of leptin secretion is observed with peak levels in the midnight probably resulting from cumulative hyperinsulinemia of entire day. Leptin has a dual role in weight maintenance. Leptin reflects total body adipose tissue mass whereas in conditions of negative and positive energy balance, the dynamic changes in plasma leptin concentration function as a sensor of energy balance and influence the efferent energy regulation pathways. Many effects of leptin on metabolism are mediated by interaction with Insulin and also by synergistic action with cholecystokinin. Besides physiological roles, leptin may influence pathological conditions like obesity-associated atherosclerosis, oxidative stress and cancers. The purpose of the present review is to summarize the important aspects of the biology, actions, and regulation of leptin and to serve as an update of new information.
Collapse
|
50
|
Abstract
White adipose tissue is a key endocrine and secretory organ, releasing multiple adipokines, many of which are linked to inflammation and immunity. During the expansion of adipose tissue mass in obesity there is a major inflammatory response in the tissue with increased expression and release of inflammation-related adipokines, including IL-6, leptin, monocyte chemoattractant protein-1 and TNF-α, together with decreased adiponectin production. We proposed in 2004 (Trayhurn & Wood, Br J Nutr92, 347–355) that inflammation in adipose tissue in obesity is a response to hypoxia in enlarged adipocytes distant from the vasculature. Hypoxia has now been directly demonstrated in adipose tissue of several obese mouse models (ob/ob, KKAy, diet-induced) and molecular studies indicate that the level of the hypoxia-inducible transcription factor, hypoxia-inducible factor-1α, is increased, as is expression of the hypoxia-sensitive marker gene, GLUT1. Cell- culture studies on murine and human adipocytes show that hypoxia (induced by low O2 or chemically) leads to stimulation of the expression and secretion of a number of inflammation-related adipokines, including angiopoietin-like protein 4, IL-6, leptin, macrophage migration inhibitory factor and vascular endothelial growth factor. Hypoxia also stimulates the inflammatory response of macrophages and inhibits adipocyte differentiation from preadipocytes. GLUT1 gene expression, protein level and glucose transport by human adipocytes are markedly increased by hypoxia, indicating that low O2 tension stimulates glucose utilisation. It is suggested that hypoxia has a pervasive effect on adipocyte metabolism and on overall adipose tissue function, underpinning the inflammatory response in the tissue in obesity and the subsequent development of obesity-associated diseases, particularly type 2 diabetes and the metabolic syndrome.
Collapse
|