This site uses cookies, tags and tracking settings to store information that help give you the very best browsing experience. If you don't change your settings, we'll assume you're happy with this. More information Dismiss this warning

Browse

You are looking at 121 - 130 of 14,371 items for

  • Refine by access: All content x
Clear All
Hikari Hirakida Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan

Search for other papers by Hikari Hirakida in
Google Scholar
PubMed
Close
,
Taiga Okumura Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan

Search for other papers by Taiga Okumura in
Google Scholar
PubMed
Close
,
Ryosuke Fujita Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan

Search for other papers by Ryosuke Fujita in
Google Scholar
PubMed
Close
,
Yoshiki Kuse Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan

Search for other papers by Yoshiki Kuse in
Google Scholar
PubMed
Close
,
Takahiro Mizoguchi Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan

Search for other papers by Takahiro Mizoguchi in
Google Scholar
PubMed
Close
,
Satoshi Inagaki Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan

Search for other papers by Satoshi Inagaki in
Google Scholar
PubMed
Close
,
Shinsuke Nakamura Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan

Search for other papers by Shinsuke Nakamura in
Google Scholar
PubMed
Close
,
Masamitsu Shimazawa Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
Lab. of Collaborative Research for Innovative Drug Discovery, Gifu Pharmaceutical University, Gifu, Japan

Search for other papers by Masamitsu Shimazawa in
Google Scholar
PubMed
Close
, and
Hideaki Hara Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
Lab. of Collaborative Research for Innovative Drug Discovery, Gifu Pharmaceutical University, Gifu, Japan

Search for other papers by Hideaki Hara in
Google Scholar
PubMed
Close

VGF nerve growth factor inducible (VGF) is a secreted polypeptide involved in metabolic regulation. VGF-derived peptides have been reported to regulate insulin secretion in the plasma of patients with type 2 diabetes and model mice. However, the protective effects of VGF on pancreatic β-cells in diabetic model are not well understood. In this study, we aimed to elucidate the β-cell protective effect of VGF on a streptozotocin (STZ)-induced diabetic model using VGF-overexpressing (OE) mice and also examined the therapeutic effect by a small molecule, SUN N8075 which is an inducer of VGF. VGF-OE mice improved blood glucose levels and maintained β-cell mass compared to wild-type (WT) mice on STZ-induced diabetic model. In addition, VGF-OE mice showed better glucose tolerance than WT mice. In culture, AQEE-30, a VGF-derived peptide, suppressed STZ-induced β-cell death in vitro and attenuated the decrease in the phosphorylation of Akt and GSK3β. Furthermore, SUN N8075 suppressed the blood glucose levels and increased VGF expression in the pancreatic islet. SUN N8075 also protected STZ-induced β-cell death in vitro. These findings indicate that VGF plays a hypoglycemic role in response to blood glucose levels in diabetes and protects β-cells from STZ-induced cell death. Therefore, VGF and its inducer have the therapeutic potential by preserving β-cells in diabetes.

Restricted access
Renea A Taylor Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
Department of Physiology, Biomedicine Discovery Institute, Cancer Program, Monash University, Melbourne, Victoria, Australia
Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
Cabrini Institute, Cabrini Health, Malvern, Victoria, Australia

Search for other papers by Renea A Taylor in
Google Scholar
PubMed
Close
,
Mitchell G Lawrence Department of Physiology, Biomedicine Discovery Institute, Cancer Program, Monash University, Melbourne, Victoria, Australia
Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
Cabrini Institute, Cabrini Health, Malvern, Victoria, Australia
Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Cancer Program, Monash University, Melbourne, Victoria, Australia

Search for other papers by Mitchell G Lawrence in
Google Scholar
PubMed
Close
, and
Gail P Risbridger Department of Physiology, Biomedicine Discovery Institute, Cancer Program, Monash University, Melbourne, Victoria, Australia
Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
Cabrini Institute, Cabrini Health, Malvern, Victoria, Australia
Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Cancer Program, Monash University, Melbourne, Victoria, Australia

Search for other papers by Gail P Risbridger in
Google Scholar
PubMed
Close

There is longstanding interest in the role of androgens in the aetiology of prostate cancer, one of the most common malignancies worldwide. In this review, we reflect on the ways that knowledge of prostate development and hormone action have catalysed advances in the management of patients with prostate cancer. The use of hormone therapies to treat prostate cancer has changed significantly over time, including the emergence of androgen receptor signalling inhibitors (ARSI). These compounds have improved outcomes for patients with castration-resistant prostate cancer, which was once considered ‘androgen-independent’ but is clearly still driven by androgen receptor signalling in many cases. There is also a need for new therapies to manage neuroendocrine prostate cancer, which is not responsive to hormonal agents. One of the major gaps is understanding how treatment-induced neuroendocrine prostate cancer emerges and whether it can be re-sensitised to treatment. Patient-derived models, including patient-derived xenografts (PDXs), will be instrumental in facilitating future discoveries in these areas. Developments in the use of PDXs have been fostered by lessons from the field of endocrinology, such as the role of stroma and hormones in normal and developmental tissues. Thus, there is ongoing reciprocity between the discoveries in endocrinology and advances in prostate cancer research and treatment.

Free access
N Zlocowski Centro de Microscopía Electrónica, Facultad de Ciencias Médicas (CME-FCM) - Instituto de Investigaciones en Ciencias de la Salud, Consejo Nacional de Investigaciones Científicas Técnicas (INICSA-CONICET), Universidad Nacional de Córdoba, Córdoba, ArgentinaFacultad de Ciencias Médicas (CME-FCM) - Instituto de Investigaciones en Ciencias de la Salud, Consejo Nacional de Investigaciones Científicas Técnicas (INICSA-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina

Search for other papers by N Zlocowski in
Google Scholar
PubMed
Close
,
L d V Sosa Centro de Microscopía Electrónica, Facultad de Ciencias Médicas (CME-FCM) - Instituto de Investigaciones en Ciencias de la Salud, Consejo Nacional de Investigaciones Científicas Técnicas (INICSA-CONICET), Universidad Nacional de Córdoba, Córdoba, ArgentinaFacultad de Ciencias Médicas (CME-FCM) - Instituto de Investigaciones en Ciencias de la Salud, Consejo Nacional de Investigaciones Científicas Técnicas (INICSA-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina

Search for other papers by L d V Sosa in
Google Scholar
PubMed
Close
,
B De la Cruz-Thea Cellular and Molecular Neurobiology Department, CONICET - Universidad Nacional de Córdoba - Instituto de Investigación Médica Mercedes y Martín Ferreyra, Córdoba, Argentina

Search for other papers by B De la Cruz-Thea in
Google Scholar
PubMed
Close
,
C B Guido Centro de Microscopía Electrónica, Facultad de Ciencias Médicas (CME-FCM) - Instituto de Investigaciones en Ciencias de la Salud, Consejo Nacional de Investigaciones Científicas Técnicas (INICSA-CONICET), Universidad Nacional de Córdoba, Córdoba, ArgentinaFacultad de Ciencias Médicas (CME-FCM) - Instituto de Investigaciones en Ciencias de la Salud, Consejo Nacional de Investigaciones Científicas Técnicas (INICSA-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina

Search for other papers by C B Guido in
Google Scholar
PubMed
Close
,
M G Martín Cellular and Molecular Neurobiology Department, CONICET - Universidad Nacional de Córdoba - Instituto de Investigación Médica Mercedes y Martín Ferreyra, Córdoba, Argentina

Search for other papers by M G Martín in
Google Scholar
PubMed
Close
,
J H Mukdsi Centro de Microscopía Electrónica, Facultad de Ciencias Médicas (CME-FCM) - Instituto de Investigaciones en Ciencias de la Salud, Consejo Nacional de Investigaciones Científicas Técnicas (INICSA-CONICET), Universidad Nacional de Córdoba, Córdoba, ArgentinaFacultad de Ciencias Médicas (CME-FCM) - Instituto de Investigaciones en Ciencias de la Salud, Consejo Nacional de Investigaciones Científicas Técnicas (INICSA-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina

Search for other papers by J H Mukdsi in
Google Scholar
PubMed
Close
,
A I Torres Centro de Microscopía Electrónica, Facultad de Ciencias Médicas (CME-FCM) - Instituto de Investigaciones en Ciencias de la Salud, Consejo Nacional de Investigaciones Científicas Técnicas (INICSA-CONICET), Universidad Nacional de Córdoba, Córdoba, ArgentinaFacultad de Ciencias Médicas (CME-FCM) - Instituto de Investigaciones en Ciencias de la Salud, Consejo Nacional de Investigaciones Científicas Técnicas (INICSA-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina

Search for other papers by A I Torres in
Google Scholar
PubMed
Close
, and
J P Petiti Centro de Microscopía Electrónica, Facultad de Ciencias Médicas (CME-FCM) - Instituto de Investigaciones en Ciencias de la Salud, Consejo Nacional de Investigaciones Científicas Técnicas (INICSA-CONICET), Universidad Nacional de Córdoba, Córdoba, ArgentinaFacultad de Ciencias Médicas (CME-FCM) - Instituto de Investigaciones en Ciencias de la Salud, Consejo Nacional de Investigaciones Científicas Técnicas (INICSA-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina

Search for other papers by J P Petiti in
Google Scholar
PubMed
Close

Interest in epigenetics has gained substantial momentum as a result of their identified role in the regulation of tumor progression as well as their ability to pharmacologically target genes. Pituitary neuroendocrine tumors (PitNETs) tend to be inactivated via epigenetic modification, and although emerging evidence has suggested a role for epigenetic factors in PitNET tumorigenesis, the degree to which these factors may be targeted by new therapeutic strategies still remains poorly understood. The objective of the present study was to examine the participation of the EZH2/H3K27me3 axis in the proliferation of lactotroph tumor cells. We demonstrated that the levels of EZH2 and H3K27me3 were increased in murine experimental prolactin (PRL) tumors with respect to a control pituitary, in contrast with the low p21 mRNA levels encountered, with an H3K27me3 enrichment being observed in its promoter region in a GH3 tumor cell. Furthermore, specific EZH2/H3K27me3 axis inhibition blocked the proliferation of primary tumor cell culture and GH3 cells, thereby making it an attractive therapeutic target for PRL PitNETs.

Restricted access
Bettina Geidl-Flueck Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Switzerland

Search for other papers by Bettina Geidl-Flueck in
Google Scholar
PubMed
Close
and
Philipp A Gerber Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Switzerland

Search for other papers by Philipp A Gerber in
Google Scholar
PubMed
Close

Despite the existence of numerous studies supporting a pathological link between fructose consumption and the development of the metabolic syndrome and its sequelae, such as non-alcoholic fatty liver disease (NAFLD), this link remains a contentious issue. With this article, we shed a light on the impact of sugar/fructose intake on hepatic de novo lipogenesis (DNL), an outcome parameter known to be dysregulated in subjects with type 2 diabetes and/or NAFLD. In this review, we present findings from human intervention studies using physiological doses of sugar as well as mechanistic animal studies. There is evidence from both human and animal studies that fructose is a more potent inducer of hepatic lipogenesis than glucose. This is most likely due to the liver’s prominent physiological role in fructose metabolism, which may be disrupted under pathological conditions by increased hepatic expression of fructolytic and lipogenic enzymes. Increased DNL may not only contribute to ectopic fat deposition (i.e. in the liver), but it may also impair several metabolic processes through DNL-related fatty acids (e.g. beta-cell function, insulin secretion, or insulin sensitivity).

Open access
Vicki Chen Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada

Search for other papers by Vicki Chen in
Google Scholar
PubMed
Close
,
Gia V Shelp Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada

Search for other papers by Gia V Shelp in
Google Scholar
PubMed
Close
,
Jacob L Schwartz Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada

Search for other papers by Jacob L Schwartz in
Google Scholar
PubMed
Close
,
Niklas D J Aardema Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, Utah, United States

Search for other papers by Niklas D J Aardema in
Google Scholar
PubMed
Close
,
Madison L Bunnell Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, Utah, United States

Search for other papers by Madison L Bunnell in
Google Scholar
PubMed
Close
, and
Clara E Cho Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada

Search for other papers by Clara E Cho in
Google Scholar
PubMed
Close

Micronutrients consumed in excess or imbalanced amounts during pregnancy may increase the risk of metabolic diseases in offspring, but the mechanisms underlying these effects are unknown. Serotonin (5-hydroxytryptamine, 5-HT), a multifunctional indoleamine in the brain and the gut, may have key roles in regulating metabolism. We investigated the effects of gestational micronutrient intakes on the central and peripheral serotonergic systems as modulators of the offspring's metabolic phenotypes. Pregnant Wistar rats were fed an AIN-93G diet with 1-fold recommended vitamins (RV), high 10-fold multivitamins (HV), high 10-fold folic acid with recommended choline (HFolRC), or high 10-fold folic acid with no choline (HFolNC). Male and female offspring were weaned to a high-fat RV diet for 12 weeks. We assessed the central function using the 5-HT2C receptor agonist, 1-(3-chlorophenyl)piperazine (mCPP), and found that male offspring from the HV- or HFolRC-fed dams were less responsive (P < 0.05) whereas female HFolRC offspring were more responsive to mCPP (P < 0.01) at 6 weeks post-weaning. Male and female offspring from the HV and HFolNC groups, and male HFolRC offspring had greater food intake (males P < 0.001; females P < 0.001) and weight gain (males P < 0.0001; females P < 0.0001), elevated colon 5-HT (males P < 0.01; females P < 0.001) and fasting glucose concentrations (males P < 0.01; females P < 0.01), as well as body composition toward obesity (males P < 0.01; females P < 0.01) at 12 weeks post-weaning. Colon 5-HT was correlated with fasting glucose concentrations (males R2=0.78, P < 0.0001; females R2=0.71, P < 0.0001). Overall, the serotonergic systems are sensitive to the composition of gestational micronutrients, with alterations consistent with metabolic disturbances in offspring.

Open access
Sy-Ying Leu Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan, ROC

Search for other papers by Sy-Ying Leu in
Google Scholar
PubMed
Close
,
Yi-Ling Tsang Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
Institute of Physiological Chemistry and Pathobiochemistry and Cells in Motion Interfaculty Centre, University of Münster, Münster, Germany

Search for other papers by Yi-Ling Tsang in
Google Scholar
PubMed
Close
,
Li-Chun Ho School of Medicine, I-Shou University, Kaohsiung, Taiwan, ROC
Division of General Medicine, Department of Internal Medicine, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan, ROC

Search for other papers by Li-Chun Ho in
Google Scholar
PubMed
Close
,
Ching-Chun Yang Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC

Search for other papers by Ching-Chun Yang in
Google Scholar
PubMed
Close
,
Ai-Ning Shao Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC

Search for other papers by Ai-Ning Shao in
Google Scholar
PubMed
Close
,
Chia-Yu Chang Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC

Search for other papers by Chia-Yu Chang in
Google Scholar
PubMed
Close
,
Hui-Kuan Lin Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA

Search for other papers by Hui-Kuan Lin in
Google Scholar
PubMed
Close
,
Pei-Jane Tsai Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC

Search for other papers by Pei-Jane Tsai in
Google Scholar
PubMed
Close
,
Junne-Ming Sung Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan, ROC

Search for other papers by Junne-Ming Sung in
Google Scholar
PubMed
Close
, and
Yau-Sheng Tsai Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
Clinical Medicine Research Center, National Cheng Kung University Hospital, Tainan, Taiwan, ROC

Search for other papers by Yau-Sheng Tsai in
Google Scholar
PubMed
Close

The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is an oligomeric complex that assembles in response to exogenous signals of pathogen infection and endogenous danger signals of non-microbial origin. When NLRP3 inflammasome assembly activates caspase-1, it promotes the maturation and release of the inflammatory cytokines interleukin-1B and IL-18. Aberrant activation of the NLRP3 inflammasome has been implicated in various diseases, including chronic inflammatory, metabolic, and cardiovascular diseases. The NLRP3 inflammasome can be activated through several principal mechanisms, including K+ efflux, lysosomal damage, and the production of mitochondrial reactive oxygen species. Interestingly, metabolic danger signals activate the NLRP3 inflammasome to induce metabolic diseases. NLRP3 contains three crucial domains: an N-terminal pyrin domain, a central nucleotide-binding domain, and a C-terminal leucine-rich repeat domain. Protein–protein interactions act as a ‘pedal or brake’ to control the activation of the NLRP3 inflammasome. In this review, we present the mechanisms underlying NLRP3 inflammasome activation after induction by metabolic danger signals or via protein–protein interactions with NLRP3 that likely occur in metabolic diseases. Understanding these mechanisms will enable the development of specific inhibitors to treat NLRP3-related metabolic diseases.

Free access
Timothy J Dreyer Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK

Search for other papers by Timothy J Dreyer in
Google Scholar
PubMed
Close
,
Jacob AC Keen Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK

Search for other papers by Jacob AC Keen in
Google Scholar
PubMed
Close
,
Leah M Wells Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK

Search for other papers by Leah M Wells in
Google Scholar
PubMed
Close
, and
Scott J Roberts Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK

Search for other papers by Scott J Roberts in
Google Scholar
PubMed
Close

As a key regulator of bone homeostasis, sclerostin has garnered a lot of interest over the last two decades. Although sclerostin is primarily expressed by osteocytes and is well known for its role in bone formation and remodelling, it is also expressed by a number of other cells and potentially plays a role in other organs. Herein, we aim to bring together recent sclerostin research and discuss the effect of sclerostin on bone, cartilage, muscle, liver, kidney and the cardiovascular and immune systems. Particular focus is placed on its role in diseases, such as osteoporosis and myeloma bone disease, and the novel development of sclerostin as a therapeutic target. Anti-sclerostin antibodies have recently been approved for the treatment of osteoporosis. However, a cardiovascular signal was observed, prompting extensive research into the role of sclerostin in vascular and bone tissue crosstalk. The study of sclerostin expression in chronic kidney disease was followed by the investigation of its role in liver–lipid–bone interactions, and the recent discovery of sclerostin as a myokine prompted new research into sclerostin within the bone–muscle relationship. Potentially, the effects of sclerostin reach beyond that of bone alone. We further summarise recent developments in the use of sclerostin as a potential therapeutic for osteoarthritis, osteosarcoma and sclerosteosis. Overall, these new treatments and discoveries illustrate progress within the field, however, also highlight remaining gaps in our knowledge.

Free access
Ken KY Ho Garvan Institute of Medical Research, St. Vincent’s Hospital and the UNSW Sydney, Sydney, New South Wales, Australia

Search for other papers by Ken KY Ho in
Google Scholar
PubMed
Close
,
Anthony J O’Sullivan St. George Hospital and the Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia

Search for other papers by Anthony J O’Sullivan in
Google Scholar
PubMed
Close
, and
Morton G Burt Southern Adelaide Diabetes and Endocrine, Flinders Medical Centre and College of Medicine and Public Health, and Flinders University, Adelaide, South Australia, Australia

Search for other papers by Morton G Burt in
Google Scholar
PubMed
Close

The fact that growth hormone (GH) plays an important role in health after the cessation of growth requiring replacement therapy in adult life has only been recognised in the last three decades. This has only been made possible by recombinant technology providing GH supplies required to undertake investigations in the physiology of GH action and the benefits of replacement therapy in patients identified by rigorously validated diagnostic tests for GH deficiency (GHD). Human studies have revealed important regulatory roles in substrate metabolism, sodium homeostasis, body composition, and physical function. GH-induced anabolism is achieved by stimulating amino acid incorporation into protein while reducing oxidative loss simultaneously enhancing lipid utilisation by stimulating fatty acid oxidation and reducing lipid storage. Sodium and fluid retention are enhanced by activating the renin–angiotensin system and distal renal tubular reabsorption. GH stimulates the aerobic and anaerobic energy systems that underpin muscle and cardiovascular function. These pleiotropic actions explain the clinical picture of increased adiposity, reduced lean mass, and impaired physical and psychological function in the GHD adult, all of which are reversed when GH is replaced. Women require a greater replacement dose of GH than men. This is because androgens enhance while oestrogens attenuate GH action. The oestrogen effect is route-dependent, occurring with oral delivery blunting the liver-mediated actions of GH by directly inhibiting GH receptor signalling, global experience spanning over 30 years has attested to the safety, efficacy, and benefits of replacement therapy for adults with GHD.

Free access
Yuta Kasahara Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan

Search for other papers by Yuta Kasahara in
Google Scholar
PubMed
Close
,
Hiroshi Kishi Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan

Search for other papers by Hiroshi Kishi in
Google Scholar
PubMed
Close
,
Ryo Yokomizo Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan

Search for other papers by Ryo Yokomizo in
Google Scholar
PubMed
Close
, and
Aikou Okamoto Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan

Search for other papers by Aikou Okamoto in
Google Scholar
PubMed
Close

There are many previous reports on the effects of ethanol on physiological function, including reports of elevated blood estrogen levels in women who drank alcohol. However, the mechanism of ethanol's effects on ovarian functions, such as follicle development and hormone secretion, has not been fully clarified. Therefore, in this study, we investigated the impacts of ethanol on these phenomena and their mechanisms using a primary culture system of rat ovarian granulosa cells (GCs). In the present experiment, groups were created in which follicle-stimulating hormone (FSH) or ethanol was added alone or FSH and ethanol were co-added, and mRNA and protein expression in each group was measured for luteinizing hormone receptor (LHR) and sex steroid hormone synthase, as well as for estradiol (E2) production, cAMP production, and FSH receptor (FSHR) internalization rate. The addition of FSH induced mRNA expression of LHR and aromatase, which led to membrane LHR expression and E2 production. The coexistence of ethanol enhanced all these responses. The action of FSH is exerted via cAMP, and the co-addition of ethanol enhanced this cAMP production. Ethanol alone did not induce cAMP production. The enhancing effect of ethanol was also observed for cAMP induced by cholera toxin. Ethanol had no significant effect on the internalization rate of FSHR. In conclusion, ethanol increased FSH-stimulated cAMP production by increasing the activity of adenylyl cyclase, which enhanced FSH actions in rat GCs. Alcohol is an exacerbating factor in several female hormone-related diseases, and the mechanism of ethanol-induced increase in estrogen secretion revealed in this study may be involved in the pathogenesis of these diseases.

Restricted access
Jitendra Vishwakarma Developmental Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India
Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India

Search for other papers by Jitendra Vishwakarma in
Google Scholar
PubMed
Close
,
Keerti Gupta Developmental Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India
Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India

Search for other papers by Keerti Gupta in
Google Scholar
PubMed
Close
,
Juhi Mishra Developmental Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India

Search for other papers by Juhi Mishra in
Google Scholar
PubMed
Close
,
Asmita Garg Developmental Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India
Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India

Search for other papers by Asmita Garg in
Google Scholar
PubMed
Close
,
Rafat Malik Developmental Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India

Search for other papers by Rafat Malik in
Google Scholar
PubMed
Close
,
Amit Kashyap Developmental Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India

Search for other papers by Amit Kashyap in
Google Scholar
PubMed
Close
,
Manoj Shukla Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow-226014, Uttar Pradesh, India

Search for other papers by Manoj Shukla in
Google Scholar
PubMed
Close
,
Dhirendra Singh Central Pathology Laboratory, Regulatory Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR–IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India

Search for other papers by Dhirendra Singh in
Google Scholar
PubMed
Close
, and
Sanghamitra Bandyopadhyay Developmental Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India
Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India

Search for other papers by Sanghamitra Bandyopadhyay in
Google Scholar
PubMed
Close

Thyroid hormones (TH) are vital for brain functions, while TH deficiency, i.e. hypothyroidism, induces neurological impairment in children and adults. Cerebellar neuronal apoptosis and motor deficits are crucial events in hypothyroidism; however, the underlying mechanism is less-known. Using a methimazole-treated hypothyroidism rat model, we investigated cerebellar autophagy, growth factor, and apoptotic mechanisms that participate in motor functions. We first identified that methimazole up-regulated cerebellar autophagy, marked by enhanced LC3B-II, Beclin-1, ATG7, ATG5-12, p-AMPKα/AMPKα, and p62 degradation as well as reduced p-AKT/AKT, p-mTOR/mTOR, and p-ULK1/ULK1 in developing and young adult rats. We probed upstream effectors of this abnormal autophagy and detected a methimazole-induced reduction in cerebellar phospho-epidermal growth factor receptor (p-EGFR)/EGFR and heparin-binding EGF-like growth factor (HB-EGF). Here, while a thyroxine-induced TH replenishment alleviated autophagy process and restored HB-EGF/EGFR, HB-EGF treatment regulated AKT-mTOR and autophagy signaling in the cerebellum. Moreover, neurons of the rat cerebellum demonstrated this reduced HB-EGF-dependent increased autophagy in hypothyroidism. We further checked whether the above events were related to cerebellar neuronal apoptosis and motor functions. We detected that comparable to thyroxine, treatment with HB-EGF or autophagy inhibitor, 3-MA, reduced methimazole-induced decrease in Nissl staining and increase in c-Caspase-3 and TUNEL-+ve apoptotic count of cerebellar neurons. Additionally, 3-MA, HB-EGF, and thyroxine attenuated the methimazole-induced diminution in riding time on rota-rod and grip strength for the motor performance of rats. Overall, our study enlightens HB-EGF/EGFR-dependent autophagy mechanism as a key to cerebellar neuronal loss and functional impairments in developmental hypothyroidism, which may be inhibited by HB-EGF and 3-MA treatments, like thyroxine.

Restricted access