1
|
Wen Y, Wang J, Duan M, Li Q, Ma J. SIRT1-Mediated Regulation of STAT3/SOCS3 Inhibits Insulin Resistance in Trophoblast Cells and Improves Gestational Diabetes Mellitus. FASEB J 2025; 39:e70620. [PMID: 40346854 DOI: 10.1096/fj.202500520r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/17/2025] [Accepted: 04/29/2025] [Indexed: 05/12/2025]
Abstract
Gestational diabetes mellitus (GDM) frequently arises during pregnancy and is closely linked to insulin resistance (IR). The objective of this study was to explore the role of SIRT1 and possible processes involved in the IR of GDM trophoblast cells. An IR cell model induced by glucosamine and a GDM mouse model induced by high-fat and high-sugar feeding were constructed. Cell viability was determined by a CCK-8 assay, and the presence of associated proteins was identified through western blotting, immunohistochemistry, and immunofluorescence techniques. Measurements of mouse body mass, fasting blood concentrationsglucose levels, fasting insulin, and oral glucose tolerance (OGTT) were obtained, and placental tissues were examined using HE and PAS staining. The findings revealed reduced expression of SIRT1 in the placental tissues of GDM patients, animals, and IR cell models. The overexpression of SIRT1 can significantly increase the glucose uptake capacity and viability of IR-treated cells; increase the expression of p-IRS1, p-AKT, and GLUT4; and decrease the expression of GLUT1. Furthermore, SIRT1 diminished the expression of SOCS3 by hindering the phosphorylation and acetylation of STAT3, thereby improving IR. In the GDM mouse model, the overexpression of SIRT1 improved IR. Overall,SIRT1 reduces IR in trophoblast cells through the suppression of the STAT3-SOCS3 signaling pathway, providing a new idea for clinical intervention in GDM.
Collapse
Affiliation(s)
- Ya Wen
- Reproductive Genetics Department, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jue Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Mengjia Duan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qianxue Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jing Ma
- Reproductive Genetics Department, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
2
|
Yu L, Yang YX, Gong Z, Wan Q, Du Y, Zhou Q, Xiao Y, Zahr T, Wang Z, Yu Z, Yang K, Geng J, Fried SK, Li J, Haeusler RA, Leong KW, Bai L, Wu Y, Sun L, Wang P, Zhu BT, Wang L, Qiang L. FcRn-dependent IgG accumulation in adipose tissue unmasks obesity pathophysiology. Cell Metab 2025; 37:656-672.e7. [PMID: 39674176 PMCID: PMC11885036 DOI: 10.1016/j.cmet.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/24/2024] [Accepted: 11/01/2024] [Indexed: 12/16/2024]
Abstract
Immunoglobulin G (IgG) is traditionally recognized as a plasma protein that neutralizes antigens for immune defense. However, our research demonstrates that IgG predominantly accumulates in adipose tissue during obesity development, triggering insulin resistance and macrophage infiltration. This accumulation is governed by neonatal Fc receptor (FcRn)-dependent recycling, orchestrated in adipose progenitor cells and macrophages during the early and late stages of diet-induced obesity (DIO), respectively. Targeting FcRn abolished IgG accumulation and rectified insulin resistance and metabolic degeneration in DIO. By integrating artificial intelligence (AI) modeling with in vivo and in vitro experimental models, we unexpectedly uncovered an interaction between IgG's Fc-CH3 domain and the insulin receptor's ectodomain. This interaction hinders insulin binding, consequently obstructing insulin signaling and adipocyte functions. These findings unveil adipose IgG accumulation as a driving force in obesity pathophysiology, providing a novel therapeutic strategy to tackle metabolic dysfunctions.
Collapse
Affiliation(s)
- Lexiang Yu
- Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, NY 10032, USA; Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yong Xiao Yang
- Research Center for Endocrine and Metabolic Diseases, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Zhen Gong
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Qianfen Wan
- Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Yifei Du
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Qiuzhong Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Tarik Zahr
- Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Zhaobin Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhewei Yu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Kangkang Yang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250021, China; Institute for Genome Engineered Animal Models of Human Diseases, College of Integrative Medicine, National Center of Genetically Engineered Animal Models for International Research, Liaoning Province Key Lab of Genetically Engineered Animal Models, Dalian Medical University, Dalian 116044, China
| | - Jinyang Geng
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250021, China; Institute for Genome Engineered Animal Models of Human Diseases, College of Integrative Medicine, National Center of Genetically Engineered Animal Models for International Research, Liaoning Province Key Lab of Genetically Engineered Animal Models, Dalian Medical University, Dalian 116044, China
| | - Susan K Fried
- Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jing Li
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Lin Bai
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yingjie Wu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250021, China; Institute for Genome Engineered Animal Models of Human Diseases, College of Integrative Medicine, National Center of Genetically Engineered Animal Models for International Research, Liaoning Province Key Lab of Genetically Engineered Animal Models, Dalian Medical University, Dalian 116044, China
| | - Lei Sun
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Pan Wang
- Research Center for Endocrine and Metabolic Diseases, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Bao Ting Zhu
- Research Center for Endocrine and Metabolic Diseases, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Liheng Wang
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| | - Li Qiang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| |
Collapse
|
3
|
Urhan E, Karaca Z. Diagnosis of hypoprolactinemia. Rev Endocr Metab Disord 2024; 25:985-993. [PMID: 39037546 PMCID: PMC11624249 DOI: 10.1007/s11154-024-09896-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Prolactin is a polypeptide hormone composed of 199 amino acids, synthesized by lactotroph cells. Its primary effects are on the mammary gland and gonadal axes, but it also influences different organs and systems, particularly metabolic functions. Current literature has mainly focused on the diagnosis, monitoring, and treatment of hyperprolactinemia. Due to the lack of a well-established effective treatment for hypoprolactinemia, it is not clinically emphasized. Therefore, data on its diagnosis is limited. Hypoprolactinemia has been associated with metabolic dysfunctions such as type 2 diabetes mellitus, fatty liver, dyslipidemia, fertility problems, sexual dysfunction, and increased cardiovascular disease. While often seen as a part of combined hormone deficiencies due to pituitary damage, isolated prolactin deficiency is rare. Hypoprolactinemia can serve as a marker for extensive pituitary gland damage and dysfunction.Low or undetectable serum prolactin levels and the absence of a sufficient prolactin peak in the thyrotropin-releasing hormone (TRH) stimulation test are considered diagnostic for hypoprolactinemia. Gender appears to influence both basal prolactin levels and TRH stimulation test responses. Basal prolactin levels of, at least, 5 ng/mL for males and 7 ng/mL for females can be used as cut-off levels for normal prolactin reserve. Minimum peak prolactin responses of 18 ng/mL for males and 41 ng/mL for females to TRH stimulation can exclude hypoprolactinemia. However, larger population studies across different age groups and sexes are needed to better define normal basal prolactin levels and prolactin responses to the TRH stimulation test.
Collapse
Affiliation(s)
- Emre Urhan
- Department of Endocrinology, Erciyes University Medical School, Kayseri, Turkey
| | - Zuleyha Karaca
- Department of Endocrinology, Erciyes University Medical School, Kayseri, Turkey.
| |
Collapse
|
4
|
Cera N, Pinto J, Pignatelli D. What do we know about abnormally low prolactin levels in polycystic ovary syndrome? A narrative review. Rev Endocr Metab Disord 2024; 25:1127-1138. [PMID: 39425884 PMCID: PMC11624252 DOI: 10.1007/s11154-024-09912-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 10/21/2024]
Abstract
Hyper and hypoprolactinemia seem to be related to the occurrence of metabolic alterations in PCOS patients. In contrast, between significantly elevated and significantly low, prolactin levels seem to be protective against metabolic consequences. In the present review, we found 4 studies investigating hypoprolactinemia in patients with PCOS. We also identified 6 additional studies that reported low levels of PRL in PCOS patients. Although its prevalence is not considered high (13.2-13.9%), its contribution is certainly significant to the metabolic alterations observed in PCOS (insulin resistance, obesity, diabetes mellitus, and fatty liver disease). Dopamine inhibits the secretion of prolactin and GnRH. If dopamine levels are low or the dopamine receptor is less expressed or mutated, the levels of prolactin and GnRH increase, and consequently, LH also increases. On the other hand, hyperprolactinemia, in prolactinomas-typical levels, acting through kisspeptin inhibition causes GnRH suppression and hypogonadotropic hypogonadism. In situations of hypoprolactinemia due to excessive dopamine agonist treatment, dosage reduction is important to minimize the decrease in prolactin levels. Nevertheless, there is a lack of prospective studies confirming these hypotheses, as well as randomized clinical trials with appropriate drugs targeting both hyperprolactin and hypoprolactin in patients with PCOS.
Collapse
Affiliation(s)
- Nicoletta Cera
- Faculty of Psychology and Education Sciences, University of Porto, Porto, Portugal
- Research Unit in Medical Imaging and Radiotherapy, Cross I&D Lisbon Research Center, Escola Superior de Saúde da Cruz Vermelha Portuguesa, Lisbon, Portugal
| | - Joana Pinto
- Faculty of Psychology and Education Sciences, University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Duarte Pignatelli
- Faculty of Medicine, University of Porto, Porto, Portugal.
- Department of Endocrinology, University Hospital S João and Faculty of Medicine of the University of Porto, Porto, Portugal.
- I3S Institute for Innovation in Health, Porto, Portugal.
| |
Collapse
|
5
|
Finn BP, Dattani MT. The molecular basis of hypoprolactinaemia. Rev Endocr Metab Disord 2024; 25:967-983. [PMID: 39417960 DOI: 10.1007/s11154-024-09906-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 10/19/2024]
Abstract
Hypoprolactinaemia is an endocrinopathy which is typically encountered as part of a combined pituitary hormone deficiency picture. The vast majority of genetic causes identified to date have been in the context of congenital hypopituitarism with multiple co-existent endocrinopathies. This is primarily with its closest hormonal relation, namely growth hormone. Acquired hypoprolactinaemia is generally rare in paediatric patients, and usually occurs together with other hormonal deficiencies. Congenital hypopituitarism occurs with an incidence of 1:4,000-10,000 cases and mutations in the following transcription factors account for the majority of documented genetic causes: PROP-1, POU1F1, LHX3/4 as well as documented case reports for a smaller subset of transcription factors and other molecules implicated in lactotroph development and prolactin secretion. Isolated prolactin deficiency has been described in a number of sporadic case reports in the literature, but no cases of mutations in the gene have been described to date. A range of genetic polymorphisms affecting multiple components of the prolactin signalling pathway have been identified in the literature, ranging from RNA spliceosome mutations (RNPC3) to loss of function mutations in IGSF-1. As paediatricians gain a greater understanding of the long-term ramifications of hypoprolactinaemia in terms of metabolic syndrome, type 2 diabetes mellitus and impaired fertility, the expectation is that clinicians will measure prolactin more frequently over time. Ultimately, we will encounter further reports of hypoprolactinaemia-related clinical presentations with further genetic mutations, in turn leading to a greater insight into the molecular basis of hypoprolactinaemia in terms of signalling pathways and downstream mediators. In the interim, the greatest untapped reserve of genetic causes remains within the phenotypic spectrum of congenital hypopituitarism.
Collapse
Affiliation(s)
- Bryan Padraig Finn
- Department of Paediatric Endocrinology, Great Ormond Street Children's Hospital, London, UK.
| | - Mehul T Dattani
- Department of Paediatric Endocrinology, Great Ormond Street Children's Hospital, London, UK
- Genetics and Genomic Medicine Research and Teaching Department, UCL GOS Institute of Child Health, London, UK
| |
Collapse
|
6
|
Karaca Z, Unluhizarci K, Kelestimur F. Hypoprolactinemia. Does it matter? Redefining the hypopituitarism and return from a mumpsimus : "Absence of proof is not the proof of absence". Rev Endocr Metab Disord 2024; 25:943-951. [PMID: 37875774 DOI: 10.1007/s11154-023-09847-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/16/2023] [Indexed: 10/26/2023]
Abstract
Prolactin (PRL) is secreted by the lactotroph cells in the anterior pituitary gland which is under inhibitory control of dopamine. The mature human PRL has more than 300 physiological actions including lactation, reproduction, homeostasis, neuroprotection, behavior, water and electrolyte balance, immunoregulation and embryonic and fetal development. PRL is involved in the growth and development of mammary gland, preparation of the breast for lactation in the postpartum period, synthesis of milk, and maintenance of milk secretion. Abnormalities in the synthesis and secretion of PRL may result in hyperprolactinemia or hypoprolactinemia. Although hyperprolactinemia has been extensively investigated in the literature, because of the subtle or unclearly defined symptoms, hypoprolactinemia is a less-known and neglected disorder. Failure of lactation is a well-known clinical manifestation of hypoprolactinemia. Recent studies reveal that hypoprolactinemia may have some effects beyond lactation such as increased risk for metabolic abnormalities including insulin resistance, abnormal lipid profile, obesity and sexual dysfunction. Very low level of PRL is suggested to be avoided in patients receiving dopamin agonist treatment to prevent unwanted effects of hypoprolactinemia. Another important point is that hypoprolactinemia is not included in the classification of hypopituitarism. Anterior pituitary failure is traditionally classified as isolated, partial and complete (panhypopituitarism) hypopituitarism regardless of prolactin level. Therefore, there are two kinds of panhypopituitarism: panhypopituitarism with normal or high PRL level and panhypopituitarism with low PRL level. In this review, we present two personal cases, discuss the diagnosis of hypoprolactinemia, hypoprolactinemia associated clinical picture and suggest to redefine the classification of hypopituitarism.
Collapse
Affiliation(s)
- Zuleyha Karaca
- Faculty of Medicine, Department of Endocrinology Kayseri, Erciyes University, Talas/Kayseri, Turkey
| | - Kursad Unluhizarci
- Faculty of Medicine, Department of Endocrinology Kayseri, Erciyes University, Talas/Kayseri, Turkey
| | - Fahrettin Kelestimur
- Faculty of Medicine, Department of Endocrinology İstanbul, Yeditepe University, Ataşehir/İstanbul, Turkey.
| |
Collapse
|
7
|
Amin M, Wu R, Postolache TT, Gragnoli C. The prolactin receptor ( PRLR) gene is linked to and associated with the comorbidity of depression and type 2 diabetes in Italian families. Genes Dis 2024; 11:101048. [PMID: 38274369 PMCID: PMC10806260 DOI: 10.1016/j.gendis.2023.06.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 03/22/2023] [Accepted: 06/27/2023] [Indexed: 01/27/2024] Open
Affiliation(s)
- Mutaz Amin
- INSERM, US14-Orphanet, Paris 75014, France
- University of Paris, Paris 75013, France
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Al-Neelain University, Khartoum 11121, Sudan
| | - Rongling Wu
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA 17033, USA
- Department of Statistics, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Teodor T Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Denver, CO 80246, USA
- Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, MD 21090, USA
| | - Claudia Gragnoli
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA 17033, USA
- Division of Endocrinology, Department of Medicine, Creighton University School of Medicine, Omaha, NE 68124, USA
- Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, Rome 00197, Italy
| |
Collapse
|
8
|
Yue X, Hao W, Wang M, Fu Y. Astragalus polysaccharide ameliorates insulin resistance in HepG2 cells through activating the STAT5/IGF-1 pathway. Immun Inflamm Dis 2023; 11:e1071. [PMID: 38018587 PMCID: PMC10664394 DOI: 10.1002/iid3.1071] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/13/2023] [Accepted: 10/19/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Insulin resistance (IR) is considered as a major factor initiating type 2 diabetes mellitus and can lead to a reduction in glucose uptake that mainly occurs in the liver. Astragalus polysaccharide (APC), extracted from the traditional Chinese medicine, has been recorded to suppress IR. However, the underlying mechanism remains inadequately explored. METHODS IR was induced in HepG2 cells which further underwent APC treatment. Cell viability was determined by cell counting kit-8 assay. Pretreatment with AG490, an inhibitor of signal transducer and activator of transcription 5 (STAT5) signaling, was performed for investigating the influence of STAT5 on APC. Glucose uptake level was reflected by 2-deoxyglucose-6-phosphate content determined through colorimetric assay. Expression levels of insulin-like growth factor 1 (IGF-1), IGF-1 receptor (IGF-1R), phosphorylated-STAT5/STAT5, and p-protein kinase B (AKT)/AKT in the cells were assessed by Western blot. Radioimmunoassay (RIA) was used to detect IGF-1 secretion in the cells. RESULTS APC at doses of 10 and 20 mg increased the viability of HepG2 cells with/without IR induction, and abrogated IR-induced inhibition of glucose intake. Meanwhile, APC (10 mg) offset IR-induced inhibition on the expressions of IGF-1R and IGF-1, the activation of AKT and STAT5, and the secretion of IGF-1 in HepG2 cells. More importantly, the reversal effect of APC on IR-induced alterations in HepG2 cells was counteracted by AG490. CONCLUSION APC ameliorates IR in HepG2 cells through activating the STAT5/IGF-1 pathway.
Collapse
Affiliation(s)
- Xinxin Yue
- Department of Clinical CollegeHE UniversityShenyangLiaoningChina
| | - Wei Hao
- Department of Clinical CollegeHE UniversityShenyangLiaoningChina
| | - Min Wang
- Department of Clinical CollegeHE UniversityShenyangLiaoningChina
| | - Yang Fu
- Department of Burn and Plastic SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| |
Collapse
|
9
|
Mastnak L, Herman R, Ferjan S, Janež A, Jensterle M. Prolactin in Polycystic Ovary Syndrome: Metabolic Effects and Therapeutic Prospects. Life (Basel) 2023; 13:2124. [PMID: 38004264 PMCID: PMC10672473 DOI: 10.3390/life13112124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/14/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most prevalent endocrine and metabolic disorder in premenopausal women, characterized by hyperandrogenism, ovulatory dysfunction, and polycystic ovaries. Patients frequently present comorbidities, including obesity, insulin resistance, and impaired glucose and lipid metabolism. The diverse clinical presentation may mimic various endocrine disorders, making the diagnosis challenging in some clinical circumstances. Prolactin (PRL) is a recommended biomarker in the initial diagnostic workup to rule out hyperprolactinemia (HPRL). The traditional role of PRL is linked to lactation and the reproductive system. Recent research highlights PRL's emerging role in metabolic homeostasis. PRL influences metabolism directly by interacting with the pancreas, liver, hypothalamus, and adipose tissue. Its influence on an individual's metabolism is intricately tied to its serum concentration. While deficient and very high levels of PRL can negatively affect metabolism, intermediate-normal to moderately high levels may promote metabolic health. In women with PCOS, PRL levels may be altered. Research results on different aspects of the relationship between PCOS and the impact of various levels of PRL on metabolic homeostasis are limited and inconsistent. In this narrative literature review, we comprehensively examined data on serum PRL levels in PCOS patients. We investigated the correlation between a favorable metabolic profile and serum PRL levels in this population. Furthermore, we explored the concept of beneficial PRL effects on metabolism and discussed the potential therapeutic application of dopamine agonists in PCOS treatment. Lastly, we emphasized several promising avenues for future research in this field.
Collapse
Affiliation(s)
- Lara Mastnak
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Rok Herman
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Simona Ferjan
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Andrej Janež
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Mojca Jensterle
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
10
|
Xu C, He Z, Song Y, Shao S, Yang G, Zhao J. Atypical pituitary hormone-target tissue axis. Front Med 2023; 17:1-17. [PMID: 36849623 DOI: 10.1007/s11684-022-0973-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 12/05/2022] [Indexed: 03/01/2023]
Abstract
A long-held belief is that pituitary hormones bind to their cognate receptors in classical target glands to actuate their manifold functions. However, a number of studies have shown that multiple types of pituitary hormone receptors are widely expressed in non-classical target organs. Each pituitary gland-derived hormone exhibits a wide range of nonconventional biological effects in these non-classical target organs. Herein, the extra biological functions of pituitary hormones, thyroid-stimulating hormone, follicle-stimulating hormone, luteinizing hormone, adrenocorticotrophic hormone, and prolactin when they act on non-classical organs were summarized, defined by the novel concept of an "atypical pituitary hormone-target tissue axis." This novel proposal explains the pathomechanisms of abnormal glucose and lipid metabolism, obesity, hypertension, fatty liver, and atherosclerosis while offering a more comprehensive and systematic insights into the coordinated regulation of environmental factors, genetic factors, and neuroendocrine hormones on human biological functions. The continued exploration of the physiology of the "atypical pituitary hormone-target tissue axis" could enable the identification of novel therapeutic targets for metabolic diseases.
Collapse
Affiliation(s)
- Chao Xu
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.,Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China
| | - Zhao He
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.,Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China
| | - Yongfeng Song
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.,Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China
| | - Shanshan Shao
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.,Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China
| | - Guang Yang
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| | - Jiajun Zhao
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China. .,Department of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China.
| |
Collapse
|
11
|
Jiang X, Liu K, Jiang H, Yin H, Wang ED, Cheng H, Yuan F, Xiao F, Wang F, Lu W, Peng B, Shu Y, Li X, Chen S, Guo F. SLC7A14 imports GABA to lysosomes and impairs hepatic insulin sensitivity via inhibiting mTORC2. Cell Rep 2023; 42:111984. [PMID: 36640347 DOI: 10.1016/j.celrep.2022.111984] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/11/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023] Open
Abstract
Lysosomal amino acid accumulation is implicated in several diseases, but its role in insulin resistance, the central mechanism to type 2 diabetes and many metabolic diseases, is unclear. In this study, we show the hepatic expression of lysosomal membrane protein solute carrier family 7 member 14 (SLC7A14) is increased in insulin-resistant mice. The promoting effect of SLC7A14 on insulin resistance is demonstrated by loss- and gain-of-function experiments. SLC7A14 is further demonstrated as a transporter resulting in the accumulation of lysosomal γ-aminobutyric acid (GABA), which induces insulin resistance via inhibiting mTOR complex 2 (mTORC2)'s activity. These results establish a causal link between lysosomal amino acids and insulin resistance and suggest that SLC7A14 inhibition may provide a therapeutic strategy in treating insulin resistance-related and GABA-related diseases and may provide insights into the upstream mechanisms for mTORC2, the master regulator in many important processes.
Collapse
Affiliation(s)
- Xiaoxue Jiang
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, 131 Dong'an Road, Shanghai 200032, China
| | - Kan Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Haizhou Jiang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hanrui Yin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - En-Duo Wang
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Hong Cheng
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Feixiang Yuan
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, 131 Dong'an Road, Shanghai 200032, China
| | - Fei Xiao
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, 131 Dong'an Road, Shanghai 200032, China
| | - Fenfen Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wei Lu
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, 131 Dong'an Road, Shanghai 200032, China
| | - Bo Peng
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, 131 Dong'an Road, Shanghai 200032, China
| | - Yousheng Shu
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, 131 Dong'an Road, Shanghai 200032, China
| | - Xiaoying Li
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, 131 Dong'an Road, Shanghai 200032, China
| | - Shanghai Chen
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, 131 Dong'an Road, Shanghai 200032, China
| | - Feifan Guo
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, 131 Dong'an Road, Shanghai 200032, China; CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
12
|
Ferraris J. Is prolactin receptor signaling a target in dopamine-resistant prolactinomas? Front Endocrinol (Lausanne) 2023; 13:1057749. [PMID: 36714572 PMCID: PMC9877409 DOI: 10.3389/fendo.2022.1057749] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
The hypothalamic neuroendocrine catecholamine dopamine regulates the lactotroph function, including prolactin (PRL) secretion, proliferation, and apoptosis. The treatment of PRL-secreting tumors, formerly known as prolactinomas, has relied mainly on this physiological characteristic, making dopamine agonists the first therapeutic alternative. Nevertheless, the group of patients that do not respond to this treatment has few therapeutical options. Prolactin is another physiological regulator of lactotroph function, acting as an autocrine/paracrine factor that controls PRL secretion and cellular turnover, inducing apoptosis and decreasing proliferation. Furthermore, the signaling pathways related to these effects, mainly JAK/STAT and PI3K/Akt, and MAPK, have been extensively studied in prolactinomas and other tumors as therapeutic targets. In the present work, the relationship between PRL pathophysiology and prolactinoma development is explored, aiming to comprehend the value of PRL and PRLR-associated pathways as exploratory fields alternative to dopamine-related approaches, which are worth physiological characteristics that might be impaired and can be potentially restored or upregulated to provide more options to the patients.
Collapse
Affiliation(s)
- Jimena Ferraris
- Department of Biophysics and Biochemistry, Stockholm University, Stockholm, Sweden
| |
Collapse
|
13
|
Savva C, Helguero LA, González-Granillo M, Melo T, Couto D, Angelin B, Domingues MR, Li X, Kutter C, Korach-André M. Molecular programming modulates hepatic lipid metabolism and adult metabolic risk in the offspring of obese mothers in a sex-specific manner. Commun Biol 2022; 5:1057. [PMID: 36195702 PMCID: PMC9532402 DOI: 10.1038/s42003-022-04022-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 09/22/2022] [Indexed: 11/09/2022] Open
Abstract
Male and female offspring of obese mothers are known to differ extensively in their metabolic adaptation and later development of complications. We investigate the sex-dependent responses in obese offspring mice with maternal obesity, focusing on changes in liver glucose and lipid metabolism. Here we show that maternal obesity prior to and during gestation leads to hepatic steatosis and inflammation in male offspring, while female offspring are protected. Females from obese mothers display important changes in hepatic transcriptional activity and triglycerides profile which may prevent the damaging effects of maternal obesity compared to males. These differences are sustained later in life, resulting in a better metabolic balance in female offspring. In conclusion, sex and maternal obesity drive differently transcriptional and posttranscriptional regulation of major metabolic processes in offspring liver, explaining the sexual dimorphism in obesity-associated metabolic risk. Sex and maternal obesity drive differently transcriptional and posttranscriptional regulation of major metabolic processes in the livers of female and male offspring, contributing to the sexual dimorphism in obesity-associated metabolic risk.
Collapse
Affiliation(s)
- Christina Savva
- Department of Medicine, Cardiometabolic Unit and Integrated Cardio Metabolic Center, Karolinska Institute, Stockholm, Sweden.,Clinical Department of Endocrinology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Luisa A Helguero
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | | | - Tânia Melo
- Mass Spectrometry Centre, Department of Chemistry, University of Aveiro, Aveiro, Portugal.,CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Daniela Couto
- Mass Spectrometry Centre, Department of Chemistry, University of Aveiro, Aveiro, Portugal.,CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Bo Angelin
- Department of Medicine, Cardiometabolic Unit and Integrated Cardio Metabolic Center, Karolinska Institute, Stockholm, Sweden.,Clinical Department of Endocrinology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Maria Rosário Domingues
- Mass Spectrometry Centre, Department of Chemistry, University of Aveiro, Aveiro, Portugal.,CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Xidan Li
- Department of Medicine, Cardiometabolic Unit and Integrated Cardio Metabolic Center, Karolinska Institute, Stockholm, Sweden
| | - Claudia Kutter
- Department of Microbiology, Tumor and Cell Biology, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
| | - Marion Korach-André
- Department of Medicine, Cardiometabolic Unit and Integrated Cardio Metabolic Center, Karolinska Institute, Stockholm, Sweden. .,Department of Gene Technology, Science for Life Laboratory, Royal Institute of Technology (KTH), Stockholm, Sweden.
| |
Collapse
|
14
|
Niu Y, Jiang H, Yin H, Wang F, Hu R, Hu X, Peng B, Shu Y, Li Z, Chen S, Guo F. Hepatokine ERAP1 Disturbs Skeletal Muscle Insulin Sensitivity Via Inhibiting USP33-Mediated ADRB2 Deubiquitination. Diabetes 2022; 71:921-933. [PMID: 35192681 DOI: 10.2337/db21-0857] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022]
Abstract
Chronic inflammation in liver induces insulin resistance systemically and in other tissues, including the skeletal muscle (SM); however, the underlying mechanisms remain largely unknown. RNA sequencing of primary hepatocytes from wild-type mice fed long-term high-fat diet (HFD), which have severe chronic inflammation and insulin resistance revealed that the expression of hepatokine endoplasmic reticulum aminopeptidase 1 (ERAP1) was upregulated by a HFD. Increased ERAP1 levels were also observed in interferon-γ-treated primary hepatocytes. Furthermore, hepatic ERAP1 overexpression attenuated systemic and SM insulin sensitivity, whereas hepatic ERAP1 knockdown had the opposite effects, with corresponding changes in serum ERAP1 levels. Mechanistically, ERAP1 functions as an antagonist-like factor, which interacts with β2 adrenergic receptor (ADRB2) and reduces its expression by decreasing ubiquitin-specific peptidase 33-mediated deubiquitination and thereby interrupts ADRB2-stimulated insulin signaling in the SM. The findings of this study indicate ERAP1 is an inflammation-induced hepatokine that impairs SM insulin sensitivity. Its inhibition may provide a therapeutic strategy for insulin resistance-related diseases, such as type 2 diabetes.
Collapse
Affiliation(s)
- Yuguo Niu
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Haizhou Jiang
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hanrui Yin
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Fenfen Wang
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ronggui Hu
- Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoming Hu
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Bo Peng
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yousheng Shu
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Zhigang Li
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shanghai Chen
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Feifan Guo
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
15
|
Sosa F, Santos JEP, Rae DO, Larson CC, Macchietto M, Abrahante JE, Amaral TF, Denicol AC, Sonstegard TS, Hansen PJ. Effects of the SLICK1 mutation in PRLR on regulation of core body temperature and global gene expression in liver in cattle. Animal 2022; 16:100523. [PMID: 35468510 DOI: 10.1016/j.animal.2022.100523] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 11/01/2022] Open
Abstract
The SLICK1 mutation in bovine PRLR (c.1382del; rs517047387) is a deletion mutation resulting in a protein with a truncated intracellular domain. Cattle carrying at least one allele have a phenotype characterized by a short hair coat (slick phenotype) and increased resistance to heat stress. Given the pleiotropic nature of prolactin, the mutation may affect other physiological characteristics. The liver is one organ that could potentially be affected because of the expression of PRLR. The mutation is a dominant allele, and heterozygous animals have a similar hair coat to that of animals homozygous for the mutation. Present objectives were to determine whether inheritance of the SLICK1 mutation affects liver gene expression and if animals homozygous for the SLICK1 allele differ from heterozygotes in liver gene expression and regulation of body temperature during heat stress. In one experiment, rectal and ruminal temperatures were less for Holstein heifers that were heterozygous for the SLICK1 allele compared with wildtype heifers. There were 71 differentially expressed genes in liver, with 13 upregulated and 58 downregulated in SLICK1 heterozygotes. Among the ontologies characteristic of differentially expressed genes were those related to immune function and fatty acid and amino acid metabolism. In a prospective cohort study conducted with adult Senepol cattle, body temperature and hepatic gene expression were compared between animals heterozygous or homozygous for the SLICK1 mutation. There were no differences in ruminal temperatures between genotypes, rectal temperature was higher in animals homozygous for the SLICK1 mutation, and there was only one gene in liver that was differentially expressed. It was concluded that inheritance of the SLICK1 allele can exert functional changes beyond those related to hair growth although changes in liver gene expression were not extensive. Results are also consistent with the SLICK1 allele being dominant because there were few differences in phenotype between animals inheriting one or two copies of the allele.
Collapse
Affiliation(s)
- Froylan Sosa
- Department of Animal Sciences, D.H. Barron Reproductive and Perinatal Biology Research Program, and Genetics Institute, University of Florida, Gainesville, FL 32611-0910, USA
| | - José E P Santos
- Department of Animal Sciences, D.H. Barron Reproductive and Perinatal Biology Research Program, and Genetics Institute, University of Florida, Gainesville, FL 32611-0910, USA
| | - D Owen Rae
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville 32610-0136, USA
| | - Colleen C Larson
- Okeechobee County Cooperative Extension Service, University of Florida/Institute of Food and Agricultural Sciences, Okeechobee, FL 34972, USA
| | - Marissa Macchietto
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, USA
| | - Juan E Abrahante
- Informatics Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Thiago F Amaral
- Department of Animal Sciences, D.H. Barron Reproductive and Perinatal Biology Research Program, and Genetics Institute, University of Florida, Gainesville, FL 32611-0910, USA
| | - Anna C Denicol
- Department of Animal Science, University of California Davis, Davis, CA 95616, USA
| | | | - Peter J Hansen
- Department of Animal Sciences, D.H. Barron Reproductive and Perinatal Biology Research Program, and Genetics Institute, University of Florida, Gainesville, FL 32611-0910, USA.
| |
Collapse
|
16
|
Du R, Wu N, Bai Y, Tang L, Li L. circMAP3K4 regulates insulin resistance in trophoblast cells during gestational diabetes mellitus by modulating the miR-6795-5p/PTPN1 axis. J Transl Med 2022; 20:180. [PMID: 35449053 PMCID: PMC9022258 DOI: 10.1186/s12967-022-03386-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 04/09/2022] [Indexed: 11/25/2022] Open
Abstract
Background Insulin resistance (IR) during gestational diabetes mellitus (GDM) has been linked to dysregulated insulin-PI3K/Akt pathway. A defective insulin-PI3K/Akt pathway and dysregulated circular RNA (circRNA) levels have been observed in the placentas of patients with GDM; however, the mechanisms underlying this association remain unclear. Methods circRNAs potentially associated with GDM were selected through bioinformatics analysis and initially identified by quantitative real-time PCR (qPCR) in 9 GDM patients and 9 healthy controls, of which circMAP3K4 was further validated in additional 84 samples by qPCR. circMAP3K4 identity and localization were verified. Pearson correlation analysis was applied to evaluate the correlation between circMAP3K4 expression in the placental tissues of GDM patients and IR-related indicators. An IR model of trophoblasts was constructed using glucosamine. Interactions between miR-6795-5p and circMAP3K4 or PTPN1 were confirmed using a dual-luciferase reporter assay. The circMAP3K4/miR-6795-5p/PTPN1 axis and key markers in the insulin-PI3K/Akt pathway in placentas and trophoblasts were evaluated through qRT-PCR, immunofluorescence, and western blotting. The role of circMAP3K4 in glucose metabolism and cell growth in trophoblasts was determined using the glucose uptake and CCK8 assay, respectively. Results circMAP3K4 was highly expressed in the placentas of patients with GDM and the IR trophoblast model; this was associated with a dysregulated insulin-PI3K/Akt pathway. circMAP3K4 in the placentas of GDM patients was positively correlated with weight gain during pregnancy and time-glucose area under the curve of OGTT. circMAP3K4 and PTPN1 could both bind to miR-6795-5p. miR-6795-5p and PTPN1 were downregulated and upregulated, respectively, in the placentas of GDM patients and the IR trophoblast model. circMAP3K4 silencing or miR-6795-5p overexpression partially reversed the decrease in glucose uptake, inhibition in cell growth, and downregulated IRS1 and Akt phosphorylation in IR-trophoblasts; this restoration was reversed upon co-transfection with an miR-6795-5p inhibitor or PTPN1. Conclusion circMAP3K4 could suppress the insulin-PI3K/Akt signaling pathway via miR-6795-5p/PTPN1 axis, probably contributing to GDM-related IR. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03386-8.
Collapse
Affiliation(s)
- Runyu Du
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Na Wu
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Bai
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lei Tang
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ling Li
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
17
|
Von-Hafe M, Borges-Canha M, Vale C, Leite AR, Sérgio Neves J, Carvalho D, Leite-Moreira A. Nonalcoholic Fatty Liver Disease and Endocrine Axes-A Scoping Review. Metabolites 2022; 12:298. [PMID: 35448486 PMCID: PMC9026925 DOI: 10.3390/metabo12040298] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/20/2022] [Accepted: 03/27/2022] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the leading cause of chronic liver disease. NAFLD often occurs associated with endocrinopathies. Evidence suggests that endocrine dysfunction may play an important role in NAFLD development, progression, and severity. Our work aimed to explore and summarize the crosstalk between the liver and different endocrine organs, their hormones, and dysfunctions. For instance, our results show that hyperprolactinemia, hypercortisolemia, and polycystic ovary syndrome seem to worsen NAFLD's pathway. Hypothyroidism and low growth hormone levels also may contribute to NAFLD's progression, and a bidirectional association between hypercortisolism and hypogonadism and the NAFLD pathway looks likely, given the current evidence. Therefore, we concluded that it appears likely that there is a link between several endocrine disorders and NAFLD other than the typically known type 2 diabetes mellitus and metabolic syndrome (MS). Nevertheless, there is controversial and insufficient evidence in this area of knowledge.
Collapse
Affiliation(s)
- Madalena Von-Hafe
- Departamento de Cirurgia e Fisiologia, Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal; (M.V.-H.); (C.V.); (A.R.L.); (J.S.N.); (A.L.-M.)
| | - Marta Borges-Canha
- Departamento de Cirurgia e Fisiologia, Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal; (M.V.-H.); (C.V.); (A.R.L.); (J.S.N.); (A.L.-M.)
- Serviço de Endocrinologia, Diabetes e Metabolismo do Centro Hospitalar Universitário de São João, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal;
| | - Catarina Vale
- Departamento de Cirurgia e Fisiologia, Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal; (M.V.-H.); (C.V.); (A.R.L.); (J.S.N.); (A.L.-M.)
| | - Ana Rita Leite
- Departamento de Cirurgia e Fisiologia, Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal; (M.V.-H.); (C.V.); (A.R.L.); (J.S.N.); (A.L.-M.)
| | - João Sérgio Neves
- Departamento de Cirurgia e Fisiologia, Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal; (M.V.-H.); (C.V.); (A.R.L.); (J.S.N.); (A.L.-M.)
- Serviço de Endocrinologia, Diabetes e Metabolismo do Centro Hospitalar Universitário de São João, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal;
| | - Davide Carvalho
- Serviço de Endocrinologia, Diabetes e Metabolismo do Centro Hospitalar Universitário de São João, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal;
- Investigação e Inovação em Saúde (i3s), Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal
| | - Adelino Leite-Moreira
- Departamento de Cirurgia e Fisiologia, Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal; (M.V.-H.); (C.V.); (A.R.L.); (J.S.N.); (A.L.-M.)
- Serviço de Cirurgia Cardiotorácica do Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal
| |
Collapse
|
18
|
Castillo P, Aisagbonhi O, Saenz CC, ElShamy WM. Novel insights linking BRCA1-IRIS role in mammary gland development to formation of aggressive PABCs: the case for longer breastfeeding. Am J Cancer Res 2022; 12:396-426. [PMID: 35141026 PMCID: PMC8822284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/27/2021] [Indexed: 06/14/2023] Open
Abstract
Pregnancy-associated breast cancer (PABC) is diagnosed during or shortly after pregnancy. Although rare, PABC is a serious occurrence often of the triple negative (TNBC) subtype. Here we show progesterone, prolactin, and RANKL upregulate BRCA1-IRIS (IRIS) in separate and overlapping subpopulations of human mammary epithelial cell lines, which exacerbates the proliferation, survival, and the TNBC-like phenotype in them. Conversely, vitamin D3 reduces IRIS expression in TNBC cell lines, which attenuates growth, survival, and the TNBC-like phenotype in them. In the mouse, Brca1-Iris (Iris, mouse IRIS homolog) is expressed at low-level in nulliparous mice, increases ~10-fold in pregnant/lactating mice, to completely disappear in involuting mice, and reappears at low-level in regressed glands. Mice underwent 3 constitutive pregnancies followed by a forced involution (after 5 days of lactation) contained ~10-fold higher Iris in their mammary glands compared to those underwent physiological involution (after 21 days of lactation). While protein extracts from lactating glands promote proliferation in IRISlow and IRIS overexpressing (IRISOE) cells, extracts from involuting glands promote apoptosis in IRISlow, and aneuploidy in IRISOE cells. In a cohort of breast cancer patients, lack of breastfeeding was associated with formation of chemotherapy resistant, metastatic IRISOE breast cancers. We propose that terminal differentiation triggered by long-term breastfeeding reduces IRIS expression in mammary cells allowing their elimination by the inflammatory microenvironment during physiological involution. No/short-term breastfeeding retains in the mammary gland IRISOE cells that thrive in the inflammatory microenvironment during forced involution to become precursors for aggressive breast cancers shortly after pregnancy.
Collapse
Affiliation(s)
- Patricia Castillo
- Breast Cancer Program, San Diego Biomedical Research Institute, Gynecology and Reproductive Sciences, UC San Diego Health SystemSan Diego, CA 92121, USA
| | - Omonigho Aisagbonhi
- Department of Pathology, Gynecology and Reproductive Sciences, UC San Diego Health SystemSan Diego, CA 92121, USA
| | - Cheryl C Saenz
- Department of Obstetrics, Gynecology and Reproductive Sciences, UC San Diego Health SystemSan Diego, CA 92121, USA
| | - Wael M ElShamy
- Breast Cancer Program, San Diego Biomedical Research Institute, Gynecology and Reproductive Sciences, UC San Diego Health SystemSan Diego, CA 92121, USA
| |
Collapse
|
19
|
Gauthier MF, de Andrade AA, Fisch J, Feistauer V, Morás AM, Reinhardt LS, de Moura AC, Moura DJ, de Almeida S, Guedes RP, Giovenardi M. Dietary interventions in mice affect oxidative stress and gene expression of the Prlr and Esr1 in the adipose tissue and hypothalamus of dams and their offspring. J Physiol Biochem 2022; 78:271-282. [PMID: 35023022 DOI: 10.1007/s13105-021-00862-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 11/22/2021] [Indexed: 11/26/2022]
Abstract
Maternal diet is key to the progeny's health since it may impact on the offspring's adult life. In this study, mice dams received standard (CONT), restrictive (RD), or hypercaloric (HD) diets during mating, pregnancy, and lactation. Male offspring of each group of dams also received these diets: CONT, RD, HD. Aiming to evaluate the oxidative stress in the adipose tissue, reactive oxygen species (ROS) production, catalase (CAT), and superoxide dismutase (SOD) activities were analyzed in dams and offspring. In the adipose tissue and hypothalamus, gene expression of prolactin (Prlr) and estrogen alpha (Esr1) receptors was performed in dams and offspring. Protein expression of Stat5 was evaluated in the adipose tissue of the offspring from RD-fed dams. HD-fed dams increased triglycerides and leptin serum concentrations, and decreased SOD activity in the adipose tissue. In the offspring's adipose tissue, we observed a maternal diet effect caused by HD, with increased ROS production and SOD and CAT activities. Gene expression of Prlr and Esr1 in the offspring's adipose tissue was decreased due to maternal RD. Mice from HD-fed dams showed higher Stat5 expression compared to the offspring from CONT and RD dams in the adipose tissue. In the hypothalamus, we found decreased expression of Prlr in RD and HD dams, compared to CONT; and a maternal diet effect on Prlr and Esr1 gene expression in the offspring. In conclusion, we can affirm that maternal nutrition impacts the redox state and influences the gene expression of Prlr and Esr1, which are involved in energy metabolism, both peripherally and centrally in the adult life of the female offspring.
Collapse
Affiliation(s)
- Mariana Fraga Gauthier
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-CEP, Porto Alegre, 90050-170, Brazil
| | - Andressa Alves de Andrade
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-CEP, Porto Alegre, 90050-170, Brazil
| | - Joana Fisch
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245/308C-CEP, Porto Alegre, 90050-170, Brazil
| | - Vanessa Feistauer
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-CEP, Porto Alegre, 90050-170, Brazil
| | - Ana Moira Morás
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-CEP, Porto Alegre, 90050-170, Brazil
| | - Luiza Steffens Reinhardt
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-CEP, Porto Alegre, 90050-170, Brazil
| | - Ana Carolina de Moura
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-CEP, Porto Alegre, 90050-170, Brazil
| | - Dinara Jaqueline Moura
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-CEP, Porto Alegre, 90050-170, Brazil
| | - Silvana de Almeida
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-CEP, Porto Alegre, 90050-170, Brazil
| | - Renata Padilha Guedes
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-CEP, Porto Alegre, 90050-170, Brazil
| | - Márcia Giovenardi
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-CEP, Porto Alegre, 90050-170, Brazil.
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245/308C-CEP, Porto Alegre, 90050-170, Brazil.
| |
Collapse
|
20
|
Macotela Y, Ruiz-Herrera X, Vázquez-Carrillo DI, Ramírez-Hernandez G, Martínez de la Escalera G, Clapp C. The beneficial metabolic actions of prolactin. Front Endocrinol (Lausanne) 2022; 13:1001703. [PMID: 36213259 PMCID: PMC9539817 DOI: 10.3389/fendo.2022.1001703] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
The role of prolactin (PRL) favoring metabolic homeostasis is supported by multiple preclinical and clinical studies. PRL levels are key to explaining the direction of its actions. In contrast with the negative outcomes associated with very high (>100 μg/L) and very low (<7 μg/L) PRL levels, moderately high PRL levels, both within but also above the classically considered physiological range are beneficial for metabolism and have been defined as HomeoFIT-PRL. In animal models, HomeoFIT-PRL levels counteract insulin resistance, glucose intolerance, adipose tissue hypertrophy and fatty liver; and in humans associate with reduced prevalence of insulin resistance, fatty liver, glucose intolerance, metabolic syndrome, reduced adipocyte hypertrophy, and protection from type 2 diabetes development. The beneficial actions of PRL can be explained by its positive effects on main metabolic organs including the pancreas, liver, adipose tissue, and hypothalamus. Here, we briefly review work supporting PRL as a promoter of metabolic homeostasis in rodents and humans, the PRL levels associated with metabolic protection, and the proposed mechanisms involved. Finally, we discuss the possibility of using drugs elevating PRL for the treatment of metabolic diseases.
Collapse
|
21
|
Kirsch P, Kunadia J, Shah S, Agrawal N. Metabolic effects of prolactin and the role of dopamine agonists: A review. Front Endocrinol (Lausanne) 2022; 13:1002320. [PMID: 36246929 PMCID: PMC9562454 DOI: 10.3389/fendo.2022.1002320] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Prolactin is a polypeptide hormone that is well known for its role in reproductive physiology. Recent studies highlight its role in neurohormonal appetite regulation and metabolism. Elevated prolactin levels are widely associated with worsening metabolic disease, but it appears that low prolactin levels could also be metabolically unfavorable. This review discusses the pathophysiology of prolactin related metabolic changes, and the less commonly recognized effects of prolactin on adipose tissue, pancreas, liver, and small bowel. Furthermore, the effect of dopamine agonists on the metabolic profiles of patients with hyperprolactinemia are discussed as well.
Collapse
Affiliation(s)
- Polly Kirsch
- New York University (NYU) Grossman School of Medicine, NYU Langone Health, New York, NY, United States
| | - Jessica Kunadia
- Department of Medicine, NYU Langone Health, New York, NY, United States
| | - Shruti Shah
- New York University (NYU) Grossman School of Medicine, NYU Langone Health, New York, NY, United States
| | - Nidhi Agrawal
- Department of Medicine, NYU Langone Health, New York, NY, United States
- *Correspondence: Nidhi Agrawal,
| |
Collapse
|
22
|
Towards Understanding the Direct and Indirect Actions of Growth Hormone in Controlling Hepatocyte Carbohydrate and Lipid Metabolism. Cells 2021; 10:cells10102532. [PMID: 34685512 PMCID: PMC8533955 DOI: 10.3390/cells10102532] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
Growth hormone (GH) is critical for achieving normal structural growth. In addition, GH plays an important role in regulating metabolic function. GH acts through its GH receptor (GHR) to modulate the production and function of insulin-like growth factor 1 (IGF1) and insulin. GH, IGF1, and insulin act on multiple tissues to coordinate metabolic control in a context-specific manner. This review will specifically focus on our current understanding of the direct and indirect actions of GH to control liver (hepatocyte) carbohydrate and lipid metabolism in the context of normal fasting (sleep) and feeding (wake) cycles and in response to prolonged nutrient deprivation and excess. Caveats and challenges related to the model systems used and areas that require further investigation towards a clearer understanding of the role GH plays in metabolic health and disease are discussed.
Collapse
|
23
|
Yang H, Lin J, Li H, Liu Z, Chen X, Chen Q. Prolactin Is Associated With Insulin Resistance and Beta-Cell Dysfunction in Infertile Women With Polycystic Ovary Syndrome. Front Endocrinol (Lausanne) 2021; 12:571229. [PMID: 33716958 PMCID: PMC7947819 DOI: 10.3389/fendo.2021.571229] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 01/04/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Our study aimed to investigate if serum prolactin (PRL) levels associated with insulin resistance and beta-cell dysfunction in infertile patients with polycystic ovary syndrome (PCOS). METHODS This was a retrospective cross-sectional study performed in the reproductive medicine center of the first affiliated hospital of Wenzhou Medical University. From January 2007 to August 2018, a total of 792 PCOS and 700 non-PCOS infertile women were included. All patients' prolactin levels were in the normal range. PCOS was diagnosed according to the Rotterdam Criteria. Anthropometric parameters, blood pressure, serum prolactin levels, sex hormones, fasting lipids, fasting plasma glucose (FPG), fasting insulin (FINS) and hepatic biological parameters were measured in all subjects. RESULTS Serum prolactin levels in PCOS women were significantly decreased compared with levels in non-PCOS women after adjusting for age and BMI (P < 0.05). Moreover, we found that prolactin levels were positively associated with high-density lipoprotein cholesterol (HDL-C) and negatively associated with age, BMI, waist circumference (WC), hip circumference (HC), luteinizing hormone/follicle stimulating hormone (LH/FSH), estradiol (E2), FINS, homeostasis model assessment of insulin resistance (HOMA-IR), homeostasis model assessment of β (HOMA-β), triglyceride (TG) and alanine aminotransferase (ALT) (P < 0.05). After adjusting for age and BMI, multiple linear regression analysis revealed that LH, LH/FSH, E2, FINS, HOMA-IR, and HOMA-β were negatively associated with serum PRL (P < 0.05). CONCLUSIONS Low serum PRL levels within the normal range associates with a higher incidence of insulin resistance and beta-cell dysfunction in infertile women with PCOS.
Collapse
Affiliation(s)
- Haiyan Yang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jie Lin
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - He Li
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Zhangwei Liu
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xia Chen
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Qianqian Chen, ; Xia Chen,
| | - Qianqian Chen
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Qianqian Chen, ; Xia Chen,
| |
Collapse
|
24
|
Eveleens Maarse BC, Loh NY, Karpe F, Rosendaal FR, van Heemst D, Mook-Kanamori DO, Willems van Dijk K, Rensen PCN, Kooijman S, Christodoulides C, Noordam R. Associations between outdoor temperature and bright sunlight with metabolites in two population-based European cohorts. Nutr Metab Cardiovasc Dis 2020; 30:2252-2261. [PMID: 32912789 DOI: 10.1016/j.numecd.2020.07.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND AIMS Outdoor temperature and bright sunlight may directly and/or indirectly modulate systemic metabolism. We assessed the associations between outdoor temperature and bright sunlight duration with metabolomics. METHODS AND RESULTS Cross-sectional analyses were undertaken in non-diabetic individuals from the Oxford BioBank (OBB; N = 6368; mean age 47.0 years, males 44%) and the Netherlands Epidemiology of Obesity (NEO; N = 5916; mean age 55.6 years, males 43%) study. Data on mean outdoor bright sunlight and temperature were collected from local weather stations in the week prior to blood sampling. Fasting serum levels of 148 metabolites, including 14 lipoprotein subclasses, were measured using NMR spectroscopy. Linear regression analyses were performed to assess the associations between mean outdoor temperature and bright sunlight duration with metabolomics adjusted for age, sex, body mass index, season and either outdoor temperature or bright sunlight. A higher mean outdoor temperature was associated with increased serum concentrations of lipoprotein (sub)particles (β (SE) = 0.064 (0.018) SD per 5 °C, p = 5.03e-4) and certain amino acids such as phenylalanine (0.066 (0.016) SD, p = 6.44e-05) and leucine (0.111 (0.018) SD, p = 1.25e-09). In contrast, longer duration of bright sunlight was specifically associated with lower concentrations of very low-density lipoprotein (sub)particles (e.g., VLDL cholesterol (-0.024 (0.005) SD per 1-h bright sunlight, p = 8.06e-6)). The direction of effects was generally consistent between the OBB and NEO, although effect sizes were generally larger in the OBB. CONCLUSIONS Increased bright sunlight duration is associated with an improved metabolic profile whilst higher outdoor temperature may adversely impact cardiometabolic health.
Collapse
Affiliation(s)
- Boukje C Eveleens Maarse
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Nellie Y Loh
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ko Willems van Dijk
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Patrick C N Rensen
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Sander Kooijman
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Constantinos Christodoulides
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
25
|
Lopez-Vicchi F, De Winne C, Brie B, Sorianello E, Ladyman SR, Becu-Villalobos D. Metabolic functions of prolactin: Physiological and pathological aspects. J Neuroendocrinol 2020; 32:e12888. [PMID: 33463813 DOI: 10.1111/jne.12888] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/25/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022]
Abstract
Prolactin is named after its vital role of promoting milk production during lactation, although it has been implicated in multiple functions within the body, including metabolism and energy homeostasis. Prolactin has been hypothesised to play a key role in driving many of the adaptations of the maternal body to allow the mother to meet the physiological demands of both pregnancy and lactation, including the high energetic demands of the growing foetus followed by milk production to support the offspring after birth. Prolactin receptors are found in many tissues involved in metabolism and food intake, such as the pancreas, liver, hypothalamus, small intestine and adipose tissue. We review the literature examining the effects of prolactin in these various tissues and how they relate to changes in function in physiological states of high prolactin, such as pregnancy and lactation, and in pathological states of hyperprolactinaemia in the adult. In many cases, whether prolactin promotes healthy metabolism or leads to dysregulation of metabolic functions is highly dependent on the situation. Overall, although prolactin may not play a major role in regulating metabolism and body weight outside of pregnancy and lactation, it definitely has the ability to contribute to metabolic function.
Collapse
Affiliation(s)
- Felicitas Lopez-Vicchi
- Instituto de Biologia y Medicina Experimental, Consejo Nacional de Investigaciones Cientificas y Tecnicas, Buenos Aires, Argentina
| | - Catalina De Winne
- Instituto de Biologia y Medicina Experimental, Consejo Nacional de Investigaciones Cientificas y Tecnicas, Buenos Aires, Argentina
| | - Belen Brie
- Instituto de Biologia y Medicina Experimental, Consejo Nacional de Investigaciones Cientificas y Tecnicas, Buenos Aires, Argentina
| | - Eleonora Sorianello
- Instituto de Biologia y Medicina Experimental, Consejo Nacional de Investigaciones Cientificas y Tecnicas, Buenos Aires, Argentina
| | - Sharon R Ladyman
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre, Auckland, New Zealand
| | - Damasia Becu-Villalobos
- Instituto de Biologia y Medicina Experimental, Consejo Nacional de Investigaciones Cientificas y Tecnicas, Buenos Aires, Argentina
| |
Collapse
|
26
|
Faria de Castro L, Alves Dos Santos Á, Augusto Casulari L, Ansaneli Naves L, Amorim Amato A. Association between variations of physiological prolactin serum levels and the risk of type 2 diabetes: A systematic review and meta-analysis. Diabetes Res Clin Pract 2020; 166:108247. [PMID: 32505717 DOI: 10.1016/j.diabres.2020.108247] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/22/2020] [Accepted: 06/01/2020] [Indexed: 10/24/2022]
Abstract
AIM To determine the pooled association between variations of prolactin serum levels within the physiological range and the risk of type 2 diabetes mellitus (T2D). METHODS Pubmed, Scopus, Web of Science, and grey literature were searched for studies investigating the association between variations of prolactin serum levels in the normal range and the risk of T2D in adults. The risk of prevalent and incident T2D was summarized as the odds ratio or relative risk according to the quartile of prolactin serum concentration, using random-effects meta-analysis. RESULTS Of 2,014 articles identified, 6 met the inclusion criteria. Data were pooled from cross-sectional studies including 6,670 subjects and longitudinal studies involving 13,203 subjects. Men with prolactin levels in the fourth quartile versus those in the first quartile had decreased risk of prevalent T2D (OR 0.52; 95%CI 0.35-077). The same association was seen in women (OR 0.46; 95%CI 0.30-0.73). Conversely, prolactin levels in the fourth versus first quartile were not associated with the risk of incident T2D in men (RR 1.21; 95%CI 0.79-1.87) or women (RR 0.77; 95%CI 0.48-1.22). CONCLUSION Higher prolactin serum levels within the normal range were associated with reduced risk of prevalent but not incident T2D. Further studies are necessary to address whether this association is causal, but these findings raise the discussion regarding the optimal level of prolactin suppression in subjects undergoing therapy with dopaminergic agonists.
Collapse
Affiliation(s)
- Lucas Faria de Castro
- Endocrine Division, University Hospital of Brasilia, University of Brasilia, Brasilia, Brazil; Postgraduate Program in Health Sciences, University of Brasilia, Brazil
| | | | - Luiz Augusto Casulari
- Endocrine Division, University Hospital of Brasilia, University of Brasilia, Brasilia, Brazil; Postgraduate Program in Health Sciences, University of Brasilia, Brazil
| | - Luciana Ansaneli Naves
- Endocrine Division, University Hospital of Brasilia, University of Brasilia, Brasilia, Brazil; Postgraduate Program in Health Sciences, University of Brasilia, Brazil
| | - Angelica Amorim Amato
- Postgraduate Program in Health Sciences, University of Brasilia, Brazil; Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brazil.
| |
Collapse
|
27
|
Macotela Y, Triebel J, Clapp C. Time for a New Perspective on Prolactin in Metabolism. Trends Endocrinol Metab 2020; 31:276-286. [PMID: 32044206 DOI: 10.1016/j.tem.2020.01.004] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/09/2019] [Accepted: 01/09/2020] [Indexed: 12/13/2022]
Abstract
The pituitary hormone prolactin (PRL) regulates a variety of functions beyond reproduction. The association between physiological (pregnancy) and pathological (prolactinoma) hyperprolactinemia and metabolic alterations led to the concept of this hormone being diabetogenic. However, large cohort clinical studies have recently shown that low circulating PRL levels are associated with metabolic disease and represent a risk factor for type 2 diabetes (T2D), whereas high PRL levels are beneficial. Moreover, PRL acts on the pancreas, liver, adipose tissue, and hypothalamus to maintain and promote metabolic homeostasis. By integrating basic and clinical evidence, we hypothesize that upregulation of PRL levels is a mechanism to maintain metabolic homeostasis and, thus, propose that the range of PRL levels considered physiological should be expanded to higher values.
Collapse
Affiliation(s)
- Yazmín Macotela
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, 76230 Querétaro, México.
| | - Jakob Triebel
- Institute for Clinical Chemistry, Laboratory Medicine, and Transfusion Medicine, Nuremberg General Hospital, Paracelsus Medical University, 90419 Nuremberg, Germany
| | - Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, 76230 Querétaro, México
| |
Collapse
|
28
|
Ponce AJ, Galván-Salas T, Lerma-Alvarado RM, Ruiz-Herrera X, Hernández-Cortés T, Valencia-Jiménez R, Cárdenas-Rodríguez LE, Martínez de la Escalera G, Clapp C, Macotela Y. Low prolactin levels are associated with visceral adipocyte hypertrophy and insulin resistance in humans. Endocrine 2020; 67:331-343. [PMID: 31919769 DOI: 10.1007/s12020-019-02170-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 12/21/2019] [Indexed: 01/22/2023]
Abstract
PURPOSE Low prolactin (PRL) serum levels are associated with glucose intolerance and type 2 diabetes in adults, and with metabolic syndrome and obesity in children. In obese rodents, PRL treatment promotes insulin sensitivity by maintaining adipose tissue fitness, and lack of PRL signaling exacerbates obesity-derived metabolic alterations. Since adipose tissue dysfunction is a key factor triggering metabolic alterations, we evaluated whether PRL serum levels are associated with adipocyte hypertrophy (a marker of adipose tissue dysfunction), insulin resistance, and metabolic syndrome in lean, overweight, and obese adult men and women. METHODS Samples of serum and adipose tissue from 40 subjects were obtained to evaluate insulin resistance index (homeostasis model assessment of insulin resistance (HOMA-IR)), signs of metabolic syndrome (glucose levels, high-density lipoproteins, triglycerides, blood pressure, and waist circumference), as well as adipocyte size and gene expression in fat. RESULTS Lower PRL serum levels are associated with adipocyte hypertrophy, in visceral but not in subcutaneous fat, and with a higher HOMA-IR. Furthermore, low systemic PRL levels together with high waist circumference predict an elevated HOMA-IR whereas low serum PRL values in combination with high blood glucose predicts visceral adipocyte hypertrophy. In agreement, visceral fat from insulin resistant subjects shows reduced expression of prolactin receptor. However, there is no association between PRL levels and obesity or signs of metabolic syndrome. CONCLUSIONS Our results support that low levels of PRL are markers of visceral fat dysfunction and insulin resistance, and suggest the potential therapeutic value of medications elevating PRL levels to help maintain metabolic homeostasis.
Collapse
Affiliation(s)
- Antonio J Ponce
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, 76230, Querétaro, México
| | - Tomás Galván-Salas
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, 76230, Querétaro, México
- Hospital General de Querétaro, Servicio de Cirugía General, SESEQ, 76170, Querétaro, México
| | | | - Xarubet Ruiz-Herrera
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, 76230, Querétaro, México
| | - Tomás Hernández-Cortés
- Hospital General de Querétaro, Servicio de Cirugía General, SESEQ, 76170, Querétaro, México
| | | | - Laura E Cárdenas-Rodríguez
- Hospital General de Querétaro, Centro Estatal de Diagnóstico Especializado, SESEQ, 76170, Querétaro, México
| | - Gonzalo Martínez de la Escalera
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, 76230, Querétaro, México
| | - Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, 76230, Querétaro, México
| | - Yazmín Macotela
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, 76230, Querétaro, México.
| |
Collapse
|
29
|
Yang H, Di J, Pan J, Yu R, Teng Y, Cai Z, Deng X. The Association Between Prolactin and Metabolic Parameters in PCOS Women: A Retrospective Analysis. Front Endocrinol (Lausanne) 2020; 11:263. [PMID: 32477263 PMCID: PMC7235367 DOI: 10.3389/fendo.2020.00263] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 04/09/2020] [Indexed: 01/15/2023] Open
Abstract
Background: The aim of this retrospective study was to analyze the association between prolactin (PRL) and metabolic parameters in infertile patients with polycystic ovary syndrome (PCOS). Methods: A total of 2,052 patients with PCOS and 9,696 patients with tubal infertility (non-PCOS) undergoing in vitro fertilization and embryo transfer (IVF-ET) at the reproductive medicine center of the first affiliated hospital of Wenzhou Medical University from January 2007 to July 2017 were enrolled in this study. Serum PRL, basic endocrine hormones, fasting plasma lipid, fasting plasma glucose (FPG), liver function, thyroid hormone and other parameters were measured and analyzed. Result: PRL levels were significantly lower in PCOS patients than controls over all age groups (p < 0.05). In the PCOS patients, serum PRL was significantly and positively correlated with FPG, serum TSH and serum FT4, and significantly and negatively correlated with LH, LH/FSH, TC, TG, LDL-C, AST, ALT, γ-GGT, FT3, and FT3/FT4 (p < 0.05 or 0.01). After adjusted for age and body mass index (BMI), serum PRL was positively correlated with FPG, TSH, and FT4, and negatively correlated with LH and LH/FSH. Conclusion: Low serum PRL may be an important cause of metabolic risk in infertile patients with PCOS.
Collapse
Affiliation(s)
- Haiyan Yang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Reproductive Medicine Center of the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Junbo Di
- Qilu Children's Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiexue Pan
- Reproductive Medicine Center of the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Rong Yu
- Reproductive Medicine Center of the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yili Teng
- Reproductive Medicine Center of the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zhuhua Cai
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaohui Deng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Xiaohui Deng
| |
Collapse
|
30
|
Tian RH, Bai Y, Li JY, Guo KM. Reducing PRLR expression and JAK2 activity results in an increase in BDNF expression and inhibits the apoptosis of CA3 hippocampal neurons in a chronic mild stress model of depression. Brain Res 2019; 1725:146472. [DOI: 10.1016/j.brainres.2019.146472] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 08/27/2019] [Accepted: 09/20/2019] [Indexed: 11/25/2022]
|
31
|
Zhang P, Feng W, Chu X, Sun X, Zhu D, Bi Y. A newly noninvasive model for prediction of non-alcoholic fatty liver disease: utility of serum prolactin levels. BMC Gastroenterol 2019; 19:202. [PMID: 31775658 PMCID: PMC6882057 DOI: 10.1186/s12876-019-1120-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 11/17/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUNDS To investigate the value of prolactin (PRL) in diagnosing non-alcoholic fatty liver disease (NAFLD). METHODS Metabolic parameters and serum PRL levels were measured in 452 males and 421 females, who were randomized to the estimation or the validation group as a 1:1 ratio. Hepatic steatosis was diagnosed via abdominal ultrasound. Variables that significantly associated with NAFLD in univariate analysis were included in multiple logistic regression. We used the receiver operator characteristic (ROC) curves to test the model performance. Besides, 147 patients underwent metabolic and liver biopsy were analyzed to validate the diagnostic value of this model. RESULTS Body mass index, alanine aminotransferase, prolactin, high density lipoprotein cholesterol and HbA1c were included into models. In males, the area under ROC curve (AUC) was 0.86 (95%CI: 0.82-0.91) for the validation group. With two cut-off points (- 0.79 and 1.71), the sensitivity and specificity for predicting NALFD was 95.2 and 91.1% in the validation group, respectively. In females, the AUC was 0.82 (95%CI: 0.76-0.88) for the validation group. With two cut-off points (- 0.68 and 2.16), the sensitivity and specificity for predicting NALFD was 97.1 and 91.4% in the validation group, respectively. In subjects with liver pathology, the AUC was higher than that of fatty liver index. A positive correlation between the scores of the model and the severities of NAFLD was observed. Importantly, we demonstrated a potential value of this model in predicting nonalcoholic steatohepatitis. CONCLUSION We established a mathematic model that can conveniently and effectively diagnose the existence and severities of NAFLD.
Collapse
Affiliation(s)
- Pengzi Zhang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, No 321, Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Wenhuan Feng
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, No 321, Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Xuehui Chu
- Department of General Surgery, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Xitai Sun
- Department of General Surgery, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Dalong Zhu
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, No 321, Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Yan Bi
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, No 321, Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
32
|
Rasheed HA, Al-Kuraishy HM, Al-Gareeb AI, Hussien NR, Al-Nami MS. Effects of diabetic pharmacotherapy on prolactin hormone in patients with type 2 diabetes mellitus: Bane or Boon. J Adv Pharm Technol Res 2019; 10:163-168. [PMID: 31742116 PMCID: PMC6844004 DOI: 10.4103/japtr.japtr_65_19] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Prolactin is a polypeptide hormone secreted from the anterior part of the pituitary gland which was implicated as a diabetogenic factor in the pathogenesis of type 2 diabetes mellitus (T2DM). Therefore, the aim of the present study was to evaluate prolactin serum levels in patients with T2DM regarding the effect of diabetic pharmacotherapy. Eighty patients with T2DM compared with 25 male healthy controls were recruited and divided into four groups: Group I - 29 patients with T2DM treated with metformin, Group II - 30 patients with T2DM treated with glyburide, Group III - 21 patients with T2DM treated with glyburide plus metformin, and Group IV - 25 control male healthy patients. Prolactin serum levels were high in patients with T2DM compared with controls (P < 0.01). Prolactin serum levels were higher in glyburide-treated patients compared with metformin-treated patients (P < 0.01). This study concludes that high prolactin levels in patients with T2DM are linked with diabetic complications and regarded as a beneficial phenomena to overcome IR and diabetic complications. Metformin but not glyburide reduced prolactin levels due to the improvement of insulin resistance.
Collapse
Affiliation(s)
- Huda Abdulbaki Rasheed
- Department of Clinical Pharmacology, Medical Faculty, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Hayder M Al-Kuraishy
- Department of Pharmacology, Toxicology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Pharmacology, Toxicology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Nawar Raad Hussien
- Department of Clinical Pharmacology, Medical Faculty, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Marwa S Al-Nami
- Department of Clinical Pharmacology, Medical Faculty, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| |
Collapse
|
33
|
Li J, Rice MS, Huang T, Hankinson SE, Clevenger CV, Hu FB, Tworoger SS. Circulating prolactin concentrations and risk of type 2 diabetes in US women. Diabetologia 2018; 61:2549-2560. [PMID: 30306190 PMCID: PMC6309828 DOI: 10.1007/s00125-018-4733-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/16/2018] [Indexed: 01/14/2023]
Abstract
AIMS/HYPOTHESIS Prolactin, a multifunctional hormone, is involved in regulating insulin sensitivity and glucose homeostasis in experimental studies. However, whether circulating concentrations of prolactin are associated with risk of type 2 diabetes remains uncertain. METHODS We analysed the prospective relationship between circulating prolactin concentrations and type 2 diabetes risk in the Nurses' Health Study (NHS) and NHSII with up to 22 years of follow-up. Total plasma prolactin was measured using immunoassay in 8615 women free of type 2 diabetes and cardiovascular disease at baseline blood collection (NHS 1989-1990; NHSII 1996-1999) and a subset of 998 NHS women providing a second blood sample during 2000-2002. Baseline bioactive prolactin was measured in a subset of 2478 women using the Nb2 bioassay. HRs were estimated using Cox regression. RESULTS A total of 699 incident type 2 diabetes cases were documented during 156,140 person-years of follow-up. Total plasma prolactin levels were inversely associated with type 2 diabetes risk; the multivariable HR comparing the highest with the lowest quartile was 0.73 (95% CI 0.55, 0.95; ptrend = 0.02). The associations were similar by menopausal status and other risk factors (pinteraction > 0.70). Additional adjustment for sex and growth hormones, adiponectin, and inflammatory and insulin markers did not significantly alter the results. The association of plasma bioactive prolactin with type 2 diabetes risk was non-significantly stronger than that of total prolactin (HR comparing extreme quartiles, 0.53 vs 0.81 among the subset of 2478 women, pdifference = 0.11). The inverse association of total prolactin with type 2 diabetes was significant during the first 9 years after blood draw but waned linearly with time, whereas for bioactive prolactin, the inverse relationship persisted for a longer follow-up time after blood draw. CONCLUSIONS/INTERPRETATION A normally high circulating total prolactin concentration was associated with a lower type 2 diabetes risk within 9-10 years of follow-up since blood draw in US women. Our findings are consistent with experimental evidence, suggesting that among healthy women, prolactin within the biologically normal range may play a protective role in the pathogenesis of type 2 diabetes.
Collapse
Affiliation(s)
- Jun Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Megan S Rice
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Tianyi Huang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Susan E Hankinson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst, Amherst, MA, USA
| | - Charles V Clevenger
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, USA
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Shelley S Tworoger
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Inc., 12902 Magnolia Drive, MRC-CANCONT, Tampa, FL, 33612-9497, USA.
| |
Collapse
|
34
|
Wei S, Zhao J, Wang S, Huang M, Wang Y, Chen Y. Intermittent administration of a leucine-deprived diet is able to intervene in type 2 diabetes in db/db mice. Heliyon 2018; 4:e00830. [PMID: 30294696 PMCID: PMC6169254 DOI: 10.1016/j.heliyon.2018.e00830] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/28/2018] [Accepted: 09/25/2018] [Indexed: 12/19/2022] Open
Abstract
Continuous deficiency of leucine, a member of branched chain amino acids, is able to reduce obesity and improve insulin sensitivity in mice. Intermittent fasting has been shown to be effective in intervention of metabolic disorders including diabetes. However, it is unknown whether intermittent leucine deprivation can intervene in type 2 diabetes progression. We administered leucine-deprived food every other day in db/db mice, a type 2 diabetes model, for a total of eight weeks to investigate the interventional effect of intermittent leucine deprivation. Intermittent leucine deprivation significantly reduces hyperglycemia in db/db mice independent of body weight change, together with improvement in glucose tolerance and insulin sensitivity. The total area of pancreatic islets and β cell number are increased by intermittent leucine deprivation, accompanied by elevated proliferation of β cells. The expression level of Ngn3, a β cell progenitor marker, is also increased by leucine-deleted diet. However, leucine deficiency engenders an increase in fat mass and a decrease in lean mass. Lipid accumulation in the liver is elevated and liver function is compromised by leucine deprivation. In addition, leucine deficiency alters the composition of gut microbiota. Leucine deprivation increases the genera of Bacteroides, Alloprevotella, Rikenellaceae while reduces Lachnospiraceae and these changes are correlated with fasting blood glucose levels of the mice. Collectively, our data demonstrated that intermittent leucine deprivation can intervene in the progression of type 2 diabetes in db/db mice. However, leucine deficiency reduces lean mass and aggravates hepatic steatosis in the mouse.
Collapse
Affiliation(s)
- Siying Wei
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jingyu Zhao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shuo Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Meiqin Huang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yining Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yan Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| |
Collapse
|
35
|
Zhang P, Ge Z, Wang H, Feng W, Sun X, Chu X, Jiang C, Wang Y, Zhu D, Bi Y. Prolactin improves hepatic steatosis via CD36 pathway. J Hepatol 2018; 68:1247-1255. [PMID: 29452209 DOI: 10.1016/j.jhep.2018.01.035] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 01/23/2018] [Accepted: 01/25/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND & AIMS Prolactin (PRL) is a multifunctional polypeptide with effects on metabolism, however, little is known about its effect on hepatic steatosis and lipid metabolism. Herein, we aimed to assess the role of PRL in the development of non-alcoholic fatty liver disease (NAFLD). METHODS The serum PRL levels of 456 patients with NAFLD, 403 controls without NAFLD diagnosed by ultrasound, and 85 individuals with liver histology obtained during metabolic surgery (44 female and 30 male patients with NAFLD and 11 age-matched non-NAFLD female individuals) were evaluated. The expression of the gene encoding the prolactin receptor (PRLR) and signalling molecules involved in hepatic lipid metabolism were evaluated in human liver and HepG2 cells. The effects of overexpression of PRLR or fatty acid translocase (FAT)/CD36 or knockdown of PRLR on hepatic lipid metabolism were tested in free fatty acid (FFA)-treated HepG2 cells. RESULTS Circulating PRL levels were lower in individuals with ultrasound-diagnosed NAFLD (men: 7.9 [range, 5.9-10.3] µg/L; women: 8.7 [range, 6.1-12.4] µg/L) than those with non-NAFLD (men: 9.1 [range, 6.8-13.0] µg/L, p = 0.002; women: 11.6 [range, 8.2-16.1] µg/L, p <0.001). PRL levels in patients with biopsy-proven severe hepatic steatosis were lower compared with those with mild-to-moderate hepatic steatosis in both men (8.3 [range, 5.4-9.5] µg/L vs. 9.7 [range, 7.1-12.3] µg/L, p = 0.031) and women (8.5 [range, 4.2-10.6] µg/L vs. 9.8 [range, 8.2-15.7] µg/L, p = 0.027). Furthermore, hepatic PRLR gene expression was significantly reduced in patients with NAFLD and negatively correlated with CD36 gene expression. In FFA-induced HepG2 cells, PRL treatment or PRLR overexpression significantly reduced the expression of CD36 and lipid content, effects that were abrogated after silencing of PRLR. Furthermore, overexpression of CD36 significantly reduced the PRL-mediated improvement in lipid content. CONCLUSIONS Our results reveal a novel association between the central nervous system and the liver, whereby PRL/PRLR improved hepatic lipid accumulation via the CD36 pathway. LAY SUMMARY Our clinical study suggests a negative association between prolactin (PRL)/prolactin receptor (PRLR) and the presence of non-alcoholic fatty liver disease (NAFLD). Using cell experiments, we found that PRL ameliorates hepatic steatosis via the hepatic PRLR and fatty acid translocase (FAT)/CD36, a key transporter of free fatty acid uptake in liver. Our findings suggest a novel approach to improving NAFLD using PRL and PRLR. Clinical trial number: NCT03296605.
Collapse
Affiliation(s)
- Pengzi Zhang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Zhijuan Ge
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Hongdong Wang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Wenhuan Feng
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Xitai Sun
- Department of General Surgery, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Xuehui Chu
- Department of General Surgery, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Can Jiang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Yan Wang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Dalong Zhu
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China.
| | - Yan Bi
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
36
|
Xiao F, Wang C, Yin H, Yu J, Chen S, Fang J, Guo F. Leucine deprivation inhibits proliferation and induces apoptosis of human breast cancer cells via fatty acid synthase. Oncotarget 2018; 7:63679-63689. [PMID: 27579768 PMCID: PMC5325395 DOI: 10.18632/oncotarget.11626] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 08/08/2016] [Indexed: 01/02/2023] Open
Abstract
Substantial studies on fatty acid synthase (FASN) have focused on its role in regulating lipid metabolism and researchers have a great interest in treating cancer with dietary manipulation of amino acids. In the current study, we found that leucine deprivation caused the FASN-dependent anticancer effect. Here we showed that leucine deprivation inhibited cell proliferation and induced apoptosis of MDA-MB-231 and MCF-7 breast cancer cells. In an in vivo tumor xenograft model, the leucine-free diet suppressed the growth of human breast cancer tumors and triggered widespread apoptosis of the cancer cells. Further study indicated that leucine deprivation decreased expression of lipogenic gene FASN in vitro and in vivo. Over-expression of FASN or supplementation of palmitic acid (the product of FASN action) blocked the effects of leucine deprivation on cell proliferation and apoptosis in vitro and in vivo. Moreover, leucine deprivation suppressed the FASN expression via regulating general control non-derepressible (GCN)2 and sterol regulatory element-binding protein 1C (SREBP1C). Taken together, our study represents proof of principle that anticancer effects can be obtained with strategies to deprive tumors of leucine via suppressing FASN expression, which provides important insights in prevention of breast cancer via metabolic intervention.
Collapse
Affiliation(s)
- Fei Xiao
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of The Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Chunxia Wang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of The Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Hongkun Yin
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of The Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Junjie Yu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of The Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Shanghai Chen
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of The Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Jing Fang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of The Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Feifan Guo
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of The Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
37
|
Vela D, Sopi RB, Mladenov M. Low Hepcidin in Type 2 Diabetes Mellitus: Examining the Molecular Links and Their Clinical Implications. Can J Diabetes 2018; 42:179-187. [DOI: 10.1016/j.jcjd.2017.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/18/2017] [Accepted: 04/21/2017] [Indexed: 01/14/2023]
|
38
|
Shao S, Yao Z, Lu J, Song Y, He Z, Yu C, Zhou X, Zhao L, Zhao J, Gao L. Ablation of prolactin receptor increases hepatic triglyceride accumulation. Biochem Biophys Res Commun 2018. [PMID: 29524401 DOI: 10.1016/j.bbrc.2018.03.048] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Increasing prevalence of non-alcoholic fatty liver disease (NAFLD) worldwide has necessitated a more thorough understanding of it and expanded the scope of research in this field. Women are more resistant to NAFLD than men despite equal exposure to major risk factors, such as obesity or hyperlipidemia. Female resistance is hormone-dependent, as evidenced by the sharp increase in NAFLD incidence in post-menopausal women who do not take hormone replacement therapy. Here, we found that the estrogen-responsive pituitary hormone prolactin (PRL), through specific PRL receptor (PRLR), down-regulates hepatic triglyceride (TG) accumulation. PRL was demonstrated to significantly down-regulate hepatic TG accumulation in female mice and protect male mice from liver steatosis induced by high-fat diet. Interestingly, Ad-shPRLR injected mice, whose hepatic PRLR abundance was effectively decreased at the protein levels, exhibited significantly aggravated liver steatosis. PRL could decrease the expression of stearoyl-coenzyme A desaturase 1 (SCD1), the rate-limiting enzyme in the biosynthesis of monounsaturated fatty acids, in animal models and multiple hepatic cell lines. Following knockdown of PRLR, the changes to PRL-triggered SCD1 expression disappeared. Thus, PRL acted as a previously unrecognized master regulator of liver TG metabolism, indicating that modification of PRL via PRLR might serve as a potential therapeutic target for NAFLD.
Collapse
Affiliation(s)
- Shanshan Shao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, China; Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, China
| | - Zhenyu Yao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, China; Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, China
| | - Jiayu Lu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, China; Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, China
| | - Yongfeng Song
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, China; Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, China
| | - Zhao He
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, China; Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, China
| | - Chunxiao Yu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, China; Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, China
| | - Xiaoming Zhou
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, China; Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, China
| | - Lifang Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, China; Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, China; Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, China
| | - Ling Gao
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, China; Scientific Center, Shandong Provincial Hospital Affiliated to Shandong University, China.
| |
Collapse
|
39
|
de Los Ríos EA, Ruiz-Herrera X, Tinoco-Pantoja V, López-Barrera F, Martínez de la Escalera G, Clapp C, Macotela Y. Impaired prolactin actions mediate altered offspring metabolism induced by maternal high-fat feeding during lactation. FASEB J 2018; 32:3457-3470. [PMID: 29401632 DOI: 10.1096/fj.201701154r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Maternal diet during lactation affects offspring metabolic health throughout life. Prolactin (PRL) is present in high quantities in maternal milk; however, the effects of milk PRL on the offspring remain poorly characterized. In this study, we evaluated whether feeding a high-fat diet (HFD) to rats during lactation alters PRL, both in the mother's serum and in milk, and whether this factor contributes to HFD-induced metabolic dysfunction in the offspring. Maternal HFD resulted in decreased PRL levels in milk (but not in serum), reduced mammary gland (MG) PRL receptor expression, and altered MG structure and function. Offspring from HFD-fed dams had increased body weight and adiposity, and developed fatty liver, hyperinsulinemia, and insulin resistance at weaning. Increasing PRL levels in the HFD-fed mothers by subcutaneous osmotic minipumps releasing PRL normalized MG function and PRL levels in milk. Moreover, PRL treatment in HFD-fed mothers, or directly in their pups via oral PRL administration, increased liver STAT5 phosphorylation, reduced visceral adiposity, ameliorated fatty liver, and improved insulin sensitivity in offspring. Our results show that HFD impairs PRL actions during lactation to negatively affect MG physiology and directly impair offspring metabolism.-De los Ríos, E. A., Ruiz-Herrera, X., Tinoco-Pantoja, V., López-Barrera, F., Martínez de la Escalera, G., Clapp, C., Macotela, Y. Impaired prolactin actions mediate altered offspring metabolism induced by maternal high-fat feeding during lactation.
Collapse
Affiliation(s)
- Ericka A de Los Ríos
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| | - Xarubet Ruiz-Herrera
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| | | | | | | | - Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| | - Yazmín Macotela
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| |
Collapse
|
40
|
Huang W, Jin A, Zhang J, Wang C, Tsang LL, Cai Z, Zhou X, Chen H, Chan HC. Upregulation of CFTR in patients with endometriosis and its involvement in NFκB-uPAR dependent cell migration. Oncotarget 2017; 8:66951-66959. [PMID: 28978008 PMCID: PMC5620148 DOI: 10.18632/oncotarget.16441] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/02/2017] [Indexed: 02/06/2023] Open
Abstract
Endometriotic tissues exhibit high migration ability with the underlying mechanisms remain elusive. Our previous studies have demonstrated that cystic fibrosis transmembrane conductance regulator (CFTR) acts as a tumor suppressor regulating cell migration. In the present study, we explored whether CFTR plays a role in the development of human endometriosis. We found that both mRNA and protein expression levels of CFTR and urokinase-type plasminogen activator receptor (uPAR) were significantly increased in ectopic endometrial tissues from patients with endometriosis compared to normal endometrial tissues from women without endometriosis and positively correlated. In human endometrial Ishikawa (ISK) cells, overexpression of CFTR stimulated cell migration with upregulated NFκB p65 and uPAR. Knockdown of CFTR inhibited cell migration. Furthermore, inhibition of NFκB with its inhibitors (curcumin or Bay) significantly reduced the expression of uPAR and cell migration in the CFTR-overexpressing ISK cells. Collectively, the present results suggest that the CFTR-NFκB-uPAR signaling may contribute to the progression of human endometriosis, and indicate potential targets for diagnosis and treatment.
Collapse
Affiliation(s)
- Wenqing Huang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, PR China.,Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, PR China
| | - Aihong Jin
- Department of Gynecology, The Second People's Hospital of Shenzhen, Shenzhen, PR China
| | - Jieting Zhang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, PR China.,Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, PR China
| | - Chaoqun Wang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, PR China.,Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, PR China
| | - Lai Ling Tsang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, PR China.,Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, PR China
| | - Zhiming Cai
- Department of Gynecology, The Second People's Hospital of Shenzhen, Shenzhen, PR China
| | - Xiaping Zhou
- Department of Gynecology, The Second People's Hospital of Shenzhen, Shenzhen, PR China
| | - Hao Chen
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, PR China.,Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, PR China
| | - Hsiao Chang Chan
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, PR China.,Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, PR China.,Sichuan University - The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, West China Second Hospital, Sichuan University, Chengdu, PR China
| |
Collapse
|
41
|
Vela D, Leshoski J, Gjorgievska ES, Hadzi-Petrushev N, Jakupaj M, Sopi RB, Mladenov M. The Role of Insulin Therapy in Correcting Hepcidin Levels in Patients with Type 2 Diabetes Mellitus. Oman Med J 2017; 32:195-200. [PMID: 28584599 DOI: 10.5001/omj.2017.37] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES Iron overload can cause or contribute to the pathogenesis of type 2 diabetes mellitus (T2DM), but how the major parameters of iron metabolism change in different settings of diabetes are still unclear. The aim of this study was to determine the relationship between iron, ferritin, and hepcidin levels in diabetic patients and the effect of insulin treatment. METHODS The study included 80 subjects, 60 with T2DM and 20 without (control group). Serum hepcidin, insulin, ferritin, and iron levels were determined as well as other clinical parameters. The associations between these parameters were analyzed between both groups. RESULTS Hepcidin levels expressed as mean± standard deviation between groups showed no significant changes (14.4±6.7 ng/mL for the control group, and 18.4±7.9 ng/mL for patients with diabetes, p = 0.069). Parameters of iron metabolism showed modest correlation with the parameters of glucose metabolism. However, the correlation between ferritin and insulin in both groups was statistically significant (p = 0.032; ρ = 0.480 vs. p = 0.011; ρ = 0.328). CONCLUSIONS Our study showed that hepcidin levels in patients with T2DM on insulin therapy do not change, which might be a result of treatment with insulin. In this context, insulin treatment can be used as a novel method for correction of hepcidin levels. By correcting hepcidin levels, we can prevent cellular iron overload and reduce the risk of diabetes.
Collapse
Affiliation(s)
- Driton Vela
- Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
| | - Jovica Leshoski
- Institute of Biology, Faculty of Natural Sciences, Saints Cyril and Methodius University of Skopje, Skopje, Macedonia
| | - Elizabeta S Gjorgievska
- Faculty of Dental Medicine, Saints Cyril and Methodius University of Skopje, Skopje, Macedonia
| | - Nikola Hadzi-Petrushev
- Institute of Biology, Faculty of Natural Sciences, Saints Cyril and Methodius University of Skopje, Skopje, Macedonia
| | | | - Ramadan B Sopi
- Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
| | - Mitko Mladenov
- Institute of Biology, Faculty of Natural Sciences, Saints Cyril and Methodius University of Skopje, Skopje, Macedonia
| |
Collapse
|
42
|
Widespread pre-translational regulation of the inclusion of signal peptides in human proteins. Genomics 2017; 109:113-122. [DOI: 10.1016/j.ygeno.2017.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/16/2017] [Accepted: 01/18/2017] [Indexed: 11/23/2022]
|
43
|
Ochsner SA, Tsimelzon A, Dong J, Coarfa C, McKenna NJ. Research Resource: A Reference Transcriptome for Constitutive Androstane Receptor and Pregnane X Receptor Xenobiotic Signaling. Mol Endocrinol 2016; 30:937-48. [PMID: 27409825 DOI: 10.1210/me.2016-1095] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The pregnane X receptor (PXR) (PXR/NR1I3) and constitutive androstane receptor (CAR) (CAR/NR1I2) members of the nuclear receptor (NR) superfamily of ligand-regulated transcription factors are well-characterized mediators of xenobiotic and endocrine-disrupting chemical signaling. The Nuclear Receptor Signaling Atlas maintains a growing library of transcriptomic datasets involving perturbations of NR signaling pathways, many of which involve perturbations relevant to PXR and CAR xenobiotic signaling. Here, we generated a reference transcriptome based on the frequency of differential expression of genes across 159 experiments compiled from 22 datasets involving perturbations of CAR and PXR signaling pathways. In addition to the anticipated overrepresentation in the reference transcriptome of genes encoding components of the xenobiotic stress response, the ranking of genes involved in carbohydrate metabolism and gonadotropin action sheds mechanistic light on the suspected role of xenobiotics in metabolic syndrome and reproductive disorders. Gene Set Enrichment Analysis showed that although acetaminophen, chlorpromazine, and phenobarbital impacted many similar gene sets, differences in direction of regulation were evident in a variety of processes. Strikingly, gene sets representing genes linked to Parkinson's, Huntington's, and Alzheimer's diseases were enriched in all 3 transcriptomes. The reference xenobiotic transcriptome will be supplemented with additional future datasets to provide the community with a continually updated reference transcriptomic dataset for CAR- and PXR-mediated xenobiotic signaling. Our study demonstrates how aggregating and annotating transcriptomic datasets, and making them available for routine data mining, facilitates research into the mechanisms by which xenobiotics and endocrine-disrupting chemicals subvert conventional NR signaling modalities.
Collapse
Affiliation(s)
- Scott A Ochsner
- Departments of Molecular and Cellular Biology (S.A.O., J.D., C.C., N.J.M.) and Lester and Sue Smith Breast Center (A.T.) and the Nuclear Receptor Signaling Atlas Informatics Group (S.A.O., N.J.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Anna Tsimelzon
- Departments of Molecular and Cellular Biology (S.A.O., J.D., C.C., N.J.M.) and Lester and Sue Smith Breast Center (A.T.) and the Nuclear Receptor Signaling Atlas Informatics Group (S.A.O., N.J.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Jianrong Dong
- Departments of Molecular and Cellular Biology (S.A.O., J.D., C.C., N.J.M.) and Lester and Sue Smith Breast Center (A.T.) and the Nuclear Receptor Signaling Atlas Informatics Group (S.A.O., N.J.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Cristian Coarfa
- Departments of Molecular and Cellular Biology (S.A.O., J.D., C.C., N.J.M.) and Lester and Sue Smith Breast Center (A.T.) and the Nuclear Receptor Signaling Atlas Informatics Group (S.A.O., N.J.M.), Baylor College of Medicine, Houston, Texas 77030
| | - Neil J McKenna
- Departments of Molecular and Cellular Biology (S.A.O., J.D., C.C., N.J.M.) and Lester and Sue Smith Breast Center (A.T.) and the Nuclear Receptor Signaling Atlas Informatics Group (S.A.O., N.J.M.), Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
44
|
Gorvin CM. The prolactin receptor: Diverse and emerging roles in pathophysiology. JOURNAL OF CLINICAL AND TRANSLATIONAL ENDOCRINOLOGY 2015; 2:85-91. [PMID: 29204371 PMCID: PMC5685068 DOI: 10.1016/j.jcte.2015.05.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 05/10/2015] [Indexed: 12/21/2022]
Abstract
Investigations over two decades have revised understanding of the prolactin hormone. Long thought to be merely a lactogenic hormone, its list of functions has been extended to include: reproduction, islet differentiation, adipocyte control and immune modulation. Prolactin functions by binding cell-surface expressed prolactin receptor, initiating signaling cascades, primarily utilizing Janus kinase-signal transducer and activator of transcription (JAK-STAT). Pathway disruption has been implicated in tumorigenesis, reproductive abnormalities, and diabetes. Prolactin can also be secreted from extrapituitary sources adding complexity to understanding of its physiological functions. This review aims to describe how prolactin exerts its pathophysiological roles by endocrine and autocrine means.
Collapse
Affiliation(s)
- Caroline M Gorvin
- Academic Endocrine Unit, University of Oxford, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, Oxford, OX3 7LJ, UK
| |
Collapse
|
45
|
Lemini M, Ruiz-Herrera X, Ledesma-Colunga MG, Díaz-Lezama N, De Los Ríos EA, López-Barrera F, Méndez I, Martínez de la Escalera G, Macotela Y, Clapp C. Prolactin anterior pituitary expression and circulating levels are reduced in obese and diabetic rats: role of TGF-β and TNF-α. Am J Physiol Regul Integr Comp Physiol 2015; 308:R792-R799. [PMID: 25715833 DOI: 10.1152/ajpregu.00327.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 02/17/2015] [Indexed: 02/08/2023]
Abstract
The levels of the hormone prolactin (PRL) are reduced in the circulation of patients with Type 2 diabetes and in obese children, and lower systemic PRL levels correlate with an increased prevalence of diabetes and a higher risk of metabolic syndrome. The secretion of anterior pituitary (AP) PRL in metabolic diseases may be influenced by the interplay between transforming growth factor β (TGF-β) and tumor necrosis factor α (TNF-α), which inhibit and can stimulate AP PRL synthesis, respectively, and are known contributors to insulin resistance and metabolic complications. Here, we show that TGF-β and TNF-α antagonize the effect of each other on the expression and release of PRL by the GH4C1 lactotrope cell line. The levels of AP mRNA and circulating PRL decrease in high-fat diet-induced obese rats in parallel with increased and reduced AP levels of TGF-β and TNF-α mRNA, respectively. Likewise, AP expression and circulating levels of PRL are reduced in streptozotocin-induced diabetic rats and are associated with higher AP expression and protein levels of TGF-β and TNF-α. The opposing effects of the two cytokines on cultured AP cells, together with their altered expression in the AP of obese and diabetic rats suggest they are linked to the reduced PRL production and secretion characteristics of metabolic diseases.
Collapse
Affiliation(s)
- María Lemini
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | - Xarubet Ruiz-Herrera
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | - María G Ledesma-Colunga
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | - Nundehui Díaz-Lezama
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | - Ericka A De Los Ríos
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | - Fernando López-Barrera
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | - Isabel Méndez
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | | | - Yazmín Macotela
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | - Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| |
Collapse
|
46
|
Velázquez-Villegas LA, López-Barradas AM, Torres N, Hernández-Pando R, León-Contreras JC, Granados O, Ortíz V, Tovar AR. Prolactin and the dietary protein/carbohydrate ratio regulate the expression of SNAT2 amino acid transporter in the mammary gland during lactation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:1157-64. [PMID: 25701231 DOI: 10.1016/j.bbamem.2015.02.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 01/24/2015] [Accepted: 02/10/2015] [Indexed: 12/20/2022]
Abstract
The sodium coupled neutral amino acid transporter 2 (SNAT2/SAT2/ATA2) is expressed in the mammary gland (MG) and plays an important role in the uptake of alanine and glutamine which are the most abundant amino acids transported into this tissue during lactation. Thus, the aim of this study was to assess the amount and localization of SNAT2 before delivery and during lactation in rat MG, and to evaluate whether prolactin and the dietary protein/carbohydrate ratio might influence SNAT2 expression in the MG, liver and adipose tissue during lactation. Our results showed that SNAT2 protein abundance in the MG increased during lactation and this increase was maintained along this period, while 24 h after weaning it tended to decrease. To study the effect of prolactin on SNAT2 expression, we incubated MG explants or T47D cells transfected with the SNAT2 promoter with prolactin, and we observed in both studies an increase in the SNAT2 expression or promoter activity. Consumption of a high-protein/low carbohydrate diet increased prolactin concentration, with a concomitant increase in SNAT2 expression not only in the MG during lactation, but also in the liver and adipose tissue. There was a correlation between SNAT2 expression and serum prolactin levels depending on the amount of dietary protein/carbohydrate ratio consumed. These findings suggest that prolactin actively supports lactation providing amino acids to the gland through SNAT2 for the synthesis of milk proteins.
Collapse
Affiliation(s)
- Laura A Velázquez-Villegas
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México D.F. 14000, Mexico
| | - Adriana M López-Barradas
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México D.F. 14000, Mexico
| | - Nimbe Torres
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México D.F. 14000, Mexico
| | - Rogelio Hernández-Pando
- Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México D.F. 14000, Mexico
| | - Juan Carlos León-Contreras
- Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México D.F. 14000, Mexico
| | - Omar Granados
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México D.F. 14000, Mexico
| | - Victor Ortíz
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México D.F. 14000, Mexico
| | - Armando R Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México D.F. 14000, Mexico.
| |
Collapse
|
47
|
Lamming DW. Diminished mTOR signaling: a common mode of action for endocrine longevity factors. SPRINGERPLUS 2014; 3:735. [PMID: 25674466 PMCID: PMC4320218 DOI: 10.1186/2193-1801-3-735] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/07/2014] [Indexed: 12/12/2022]
Abstract
Since the initial observation that a calorie-restricted (CR) diet can extend rodent lifespan, many genetic and pharmaceutical interventions that also extend lifespan in mammals have been discovered. The mechanism by which CR and these other interventions extend lifespan is the subject of significant debate and research. One proposed mechanism is that CR promotes longevity by increasing insulin sensitivity, but recent findings that dissociate longevity and insulin sensitivity cast doubt on this hypothesis. These findings can be reconciled if longevity is promoted not via increased insulin sensitivity, but instead via decreased PI3K/Akt/mTOR pathway signaling. This review presents a unifying hypothesis that explains the lifespan-extending effects of a variety of genetic mutations and pharmaceutical interventions and points towards new molecular pathways which may also be leveraged to promote healthy aging.
Collapse
Affiliation(s)
- Dudley W Lamming
- Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin USA ; William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin USA
| |
Collapse
|
48
|
Xiao F, Xia T, Lv Z, Zhang Q, Xiao Y, Yu J, Liu H, Deng J, Guo Y, Wang C, Li K, Liu B, Chen S, Guo F. Central prolactin receptors (PRLRs) regulate hepatic insulin sensitivity in mice via signal transducer and activator of transcription 5 (STAT5) and the vagus nerve. Diabetologia 2014; 57:2136-44. [PMID: 25064125 DOI: 10.1007/s00125-014-3336-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 06/24/2014] [Indexed: 10/25/2022]
Abstract
AIMS/HYPOTHESIS Recent studies have revealed the crucial role of the central nervous system (CNS), especially the hypothalamus, in the regulation of insulin sensitivity in peripheral tissues. The aim of our current study was to investigate the possible involvement of hypothalamic prolactin receptors (PRLRs) in the regulation of hepatic insulin sensitivity. METHODS We employed overexpression of PRLRs in mouse hypothalamus via intracerebroventricular injection of adenovirus expressing PRLR and inhibition of PRLRs via adenovirus expressing short-hairpin RNA (shRNA) specific for PRLRs in vivo. Selective hepatic vagotomy was employed to verify the important role of the vagus nerve in mediating signals from the brain to peripheral organs. In addition, a genetic insulin-resistant animal model, the db/db mouse, was used in our study to investigate the role of hypothalamic PRLRs in regulating whole-body insulin sensitivity. RESULTS Overexpression of PRLRs in the hypothalamus improved hepatic insulin sensitivity in mice and inhibition of hypothalamic PRLRs had the opposite effect. In addition, we demonstrated that hypothalamic PRLR-improved insulin sensitivity was significantly attenuated by inhibiting the activity of signal transducer and activator of transcription 5 (STAT5) in the CNS and by selective hepatic vagotomy. Finally, overexpression of PRLRs significantly ameliorated insulin resistance in db/db mice. CONCLUSIONS/INTERPRETATION Our study identifies a novel central pathway involved in the regulation of hepatic insulin sensitivity, mediated by hypothalamic PRLR/STAT5 signalling and the vagus nerve, thus demonstrating an important role for hypothalamic PRLRs under conditions of insulin resistance.
Collapse
Affiliation(s)
- Fei Xiao
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|