1
|
Hwang J, Okada J, Liu L, Pessin JE, Schwartz GJ, Jo YH. The development of hepatic steatosis depends on the presence of liver-innervating parasympathetic cholinergic neurons in mice fed a high-fat diet. PLoS Biol 2024; 22:e3002865. [PMID: 39436946 PMCID: PMC11530026 DOI: 10.1371/journal.pbio.3002865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 11/01/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
Hepatic lipid metabolism is regulated by the autonomic nervous system of the liver, with the sympathetic innervation being extensively studied, while the parasympathetic efferent innervation is less understood despite its potential importance. In this study, we investigate the consequences of disrupted brain-liver communication on hepatic lipid metabolism in mice exposed to obesogenic conditions. We found that a subset of hepatocytes and cholangiocytes are innervated by parasympathetic nerve terminals originating from the dorsal motor nucleus of the vagus. The elimination of the brain-liver axis by deleting parasympathetic cholinergic neurons innervating the liver prevents hepatic steatosis and promotes browning of inguinal white adipose tissue (ingWAT). The loss of liver-innervating cholinergic neurons increases hepatic Cyp7b1 expression and fasting serum bile acid levels. Furthermore, knockdown of the G protein-coupled bile acid receptor 1 gene in ingWAT reverses the beneficial effects of the loss of liver-innervating cholinergic neurons, leading to the reappearance of hepatic steatosis. Deleting liver-innervating cholinergic neurons has a small but significant effect on body weight, which is accompanied by an increase in energy expenditure. Taken together, these data suggest that targeting the parasympathetic cholinergic innervation of the liver is a potential therapeutic approach for enhancing hepatic lipid metabolism in obesity and diabetes.
Collapse
Affiliation(s)
- Jiyeon Hwang
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Junichi Okada
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Li Liu
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Jeffrey E. Pessin
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Gary J. Schwartz
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Young-Hwan Jo
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Neuroscince, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
2
|
Hwang J, Okada J, Liu L, Pessin JE, Schwartz GJ, Jo YH. The development of hepatic steatosis depends on the presence of liver-innervating parasympathetic cholinergic neurons in mice fed a high-fat diet. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.03.565494. [PMID: 38260695 PMCID: PMC10802435 DOI: 10.1101/2023.11.03.565494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Hepatic lipid metabolism is regulated by the autonomic nervous system of the liver, with the sympathetic innervation being extensively studied, while the parasympathetic efferent innervation is less understood despite its potential importance. In this study, we investigate the consequences of disrupted brain-liver communication on hepatic lipid metabolism in mice exposed to obesogenic conditions. We found that a subset of hepatocytes and cholangiocytes are innervated by parasympathetic nerve terminals originating from the dorsal motor nucleus of the vagus. The elimination of the brain-liver axis by deleting parasympathetic cholinergic neurons innervating the liver prevents hepatic steatosis and promotes browning of inguinal white adipose tissue (ingWAT). The loss of liver-innervating cholinergic neurons increases hepatic Cyp7b1 expression and fasting serum bile acid levels. Furthermore, knockdown of the G protein-coupled bile acid receptor 1 gene in ingWAT reverses the beneficial effects of the loss of liver-innervating cholinergic neurons, leading to the reappearance of hepatic steatosis. Deleting liver-innervating cholinergic neurons has a small but significant effect on body weight, which is accompanied by an increase in energy expenditure. Taken together, these data suggest that targeting the parasympathetic cholinergic innervation of the liver is a potential therapeutic approach for enhancing hepatic lipid metabolism in obesity and diabetes.
Collapse
|
3
|
Goździk M, Żelaźniewicz A, Nowak-Kornicka J, Pawłowska-Seredyńska K, Umławska W, Pawłowski B. Autoimmune Hashimoto's Disease and Feminization Level-Testing the Immunocompetence Hypothesis. EVOLUTIONARY PSYCHOLOGY 2024; 22:14747049241259187. [PMID: 39238450 PMCID: PMC11378202 DOI: 10.1177/14747049241259187] [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] [Indexed: 09/07/2024] Open
Abstract
Morphological femininity depends mainly on estrogen levels at puberty and is perceived as a cue of a woman's biological condition. Due to the immunostimulant properties of estradiol, estradiol-dependent feminine traits are expected to be positively related to immunity. However, heightened immunity in women may increase the risk of autoimmune disease, thus the relationship between femininity and immune quality may be complex. This study aimed to assess the relationship between morphological femininity and both the occurrence and severity of Hashimoto thyroiditis (HT) in women of reproductive age. Moreover, 95 women with HT and 84 without HT (all between 20 and 37 years) participated in the study. Morphological femininity was assessed based on somatic measurements of sexually dimorphic traits (2D:4D ratio, WHR, breast size, facial sexual dimorphism). The occurrence and severity of HT were assessed by serum TPOAb levels. The results showed that only the 2D:4D ratio of the right hand was higher in the HT group, indicating higher femininity in these women. However, there was also a positive relationship between facial femininity and TPOAb level in women with HT, indicating a higher severity of the disease. The results suggest that prenatal and pubertal exposure to estrogens may increase the probability or severity of autoimmune diseases in adulthood, but the relationship is tentative.
Collapse
Affiliation(s)
- Malwina Goździk
- Department of Human Biology, University of Wrocław, Wroclaw, Poland
| | | | | | | | - Wioleta Umławska
- Department of Human Biology, University of Wrocław, Wroclaw, Poland
| | | |
Collapse
|
4
|
Jo YH. Differential transcriptional profiles of vagal sensory neurons in female and male mice. Front Neurosci 2024; 18:1393196. [PMID: 38808032 PMCID: PMC11131592 DOI: 10.3389/fnins.2024.1393196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/24/2024] [Indexed: 05/30/2024] Open
Abstract
Introduction Differences in metabolic homeostasis, diabetes, and obesity between males and females are evident in rodents and humans. Vagal sensory neurons in the vagus nerve ganglia innervate a variety of visceral organs and use specialized nerve endings to sense interoceptive signals. This visceral organ-brain axis plays a role in relaying interoceptive signals to higher brain centers, as well as in regulating the vago-vagal reflex. I hypothesized that molecularly distinct populations of vagal sensory neurons would play a role in causing differences in metabolic homeostasis between the sexes. Methods SnRNA-Seq was conducted on dissociated cells from the vagus nerve ganglia using the 10X Genomics Chromium platform. Results Single-nucleus RNA sequencing analysis of vagal sensory neurons from female and male mice revealed differences in the transcriptional profiles of cells in the vagus nerve ganglia. These differences are linked to the expression of sex-specific genes such as Xist, Tsix, and Ddx3y. Among the 13 neuronal clusters, one-fourth of the neurons in male mice were located in the Ddx3y-enriched VN1 and VN8 clusters, which displayed higher enrichment of Trpv1, Piezo2, Htr3a, and Vip genes. In contrast, 70% of the neurons in females were found in Xist-enriched clusters VN4, 6, 7, 10, 11, and 13, which showed enriched genes such as Fgfr1, Lpar1, Cpe, Esr1, Nrg1, Egfr, and Oprm1. Two clusters of satellite cells were identified, one of which contained oligodendrocyte precursor cells in male mice. A small population of cells expressed Ucp1 and Plin1, indicating that they are epineural adipocytes. Discussion Understanding the physiological implications of distinct transcriptomic profiles in vagal sensory neurons on energy balance and metabolic homeostasis would help develop sex-specific treatments for obesity and metabolic dysregulation.
Collapse
Affiliation(s)
- Young-Hwan Jo
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York, NY, United States
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
5
|
Lemos GAA, Santos AC, Brito DCC, Novaes MAS, Assis Neto AC. Steroidogenic activity and morphological characterization of prenatal testes and epididymis of guinea pig (Cavia porcellus). Anim Reprod Sci 2024; 261:107407. [PMID: 38217925 DOI: 10.1016/j.anireprosci.2023.107407] [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: 08/20/2023] [Accepted: 12/20/2023] [Indexed: 01/15/2024]
Abstract
The present study aims to establish the morphological, morphometric, and immunostaining patterns of the steroidogenic enzymes 17β-HSD and 5α-reductase and androgen receptors (AR) during the prenatal development of the male gonad and epididymis of Cavia porcellus. Fetuses at 22, 25, 30, 40, 45, 50, and 60 days of gestation (DG) were used. Specimens were dissected and subjected to macroscopic, histological, histomorphometric, and immunohistochemical analyses. Genital and scrotal protrusions were identified in 22 DG embryos. Gonocytes were identified at 25 DG and the formation of primary testicular cords was observed at 30 DG. Through anatomical evaluation, we observed differentiation of the epididymis into the head, body, and tail at 45 DG. During development, there is a progressive decrease in the diameters of the testicular cords and epididymal ducts. 17β-HSD enzyme immunostaining was observed in Leydig cells at all ages, while 5α-reductase was observed in Leydig cell cytoplasm and gonocytes at 40, 50, and 60 DG. AR shows gonocyte labeling at 30 DG. Thus, from the second trimester of pregnancy, it is possible to observe patterns of anatomical development, such as genital and scrotal prominence (22 DG), the appearance of gonocytes in the testicular cords at 25 DG, and the beginning of the organization of primary testicular cords at 30 DG, suggesting sexual differentiation. The 17β-HSD, 5α-reductase, and ARs play an essential role in sexual development and differentiation, presenting immunostaining at different reproductive process times.
Collapse
Affiliation(s)
- G A A Lemos
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - A C Santos
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - D C C Brito
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil; Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - M A S Novaes
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - A C Assis Neto
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
6
|
Kaneko H, Maezawa Y, Tsukagoshi‐Yamaguchi A, Koshizaka M, Takada‐Watanabe A, Nakamura R, Funayama S, Aono K, Teramoto N, Sawada D, Maeda Y, Minamizuka T, Hayashi A, Ide K, Ide S, Shoji M, Kitamoto T, Takemoto M, Kato H, Yokote K. Sex differences in symptom presentation and their impact on diagnostic accuracy in Werner syndrome. Geriatr Gerontol Int 2024; 24:161-167. [PMID: 38062994 PMCID: PMC11503585 DOI: 10.1111/ggi.14752] [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: 07/01/2023] [Revised: 10/27/2023] [Accepted: 11/09/2023] [Indexed: 01/05/2024]
Abstract
AIM Whether sex differences exist in hereditary progeroid syndromes remains unclear. In this study, we investigated sex differences in patients with Werner syndrome (WS), a model of human aging, using patient data at the time of diagnosis. METHODS The presence of six cardinal signs in the diagnostic criteria was retrospectively evaluated. RESULTS We found that the percentage of patients with all cardinal signs was higher in males than in females (54.2% vs. 21.2%). By the age of 40 years, 57.1% of male patients with WS presented with all the cardinal signs, whereas none of the female patients developed all of them. In particular, the frequency of having a high-pitched, hoarse voice, a characteristic of WS, was lower in female patients. The positive and negative predictive values for clinical diagnosis were 100% for males and females, indicating the helpfulness of diagnostic criteria regardless of sex. More female patients than male (86.7% vs. 64%) required genetic testing for their diagnosis because their clinical symptoms were insufficient, suggesting the importance of genetic testing for females even if they do not show typical symptoms of WS. Finally, the frequency of abnormal voice was lower in patients with WS harboring the c.3139-1G > C homozygous mutation. CONCLUSION These results indicate, for the first time, that there are sex differences in the phenotypes of hereditary progeroid syndromes. The analysis of this mechanism in this human model of aging may lead to the elucidation of sex differences in the various symptoms of normal human aging. Geriatr Gerontol Int 2024; 24: 161-167.
Collapse
Affiliation(s)
- Hiyori Kaneko
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Yoshiro Maezawa
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Ayano Tsukagoshi‐Yamaguchi
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Masaya Koshizaka
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Aki Takada‐Watanabe
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Rito Nakamura
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Shinichiro Funayama
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Kazuto Aono
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Naoya Teramoto
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Daisuke Sawada
- Department of PediatricsChiba University Graduate School of MedicineChibaJapan
| | - Yukari Maeda
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Takuya Minamizuka
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Aiko Hayashi
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Kana Ide
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Shintaro Ide
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Mayumi Shoji
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Takumi Kitamoto
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Minoru Takemoto
- Department of DiabetesMetabolism and Endocrinology, International University of Health and WelfareChibaJapan
| | - Hisaya Kato
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| | - Koutaro Yokote
- Department of EndocrinologyHematology and Gerontology, Chiba University Graduate School of MedicineChibaJapan
| |
Collapse
|
7
|
de Souza GO, Teixeira PDS, Câmara NOS, Donato J. mTORC1 Signaling in AgRP Neurons Is Not Required to Induce Major Neuroendocrine Adaptations to Food Restriction. Cells 2023; 12:2442. [PMID: 37887286 PMCID: PMC10605346 DOI: 10.3390/cells12202442] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/04/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
Hypothalamic mTORC1 signaling is involved in nutrient sensing. Neurons that express the agouti-related protein (AgRP) are activated by food restriction and integrate interoceptive and exteroceptive signals to control food intake, energy expenditure, and other metabolic responses. To determine whether mTORC1 signaling in AgRP neurons is necessary for regulating energy and glucose homeostasis, especially in situations of negative energy balance, mice carrying ablation of the Raptor gene exclusively in AgRP-expressing cells were generated. AgRPΔRaptor mice showed no differences in body weight, fat mass, food intake, or energy expenditure; however, a slight improvement in glucose homeostasis was observed compared to the control group. When subjected to 5 days of food restriction (40% basal intake), AgRPΔRaptor female mice lost less lean body mass and showed a blunted reduction in energy expenditure, whereas AgRPΔRaptor male mice maintained a higher energy expenditure compared to control mice during the food restriction and 5 days of refeeding period. AgRPΔRaptor female mice did not exhibit the food restriction-induced increase in serum corticosterone levels. Finally, although hypothalamic fasting- or refeeding-induced Fos expression showed no differences between the groups, AgRPΔRaptor mice displayed increased hyperphagia during refeeding. Thus, some metabolic and neuroendocrine responses to food restriction are disturbed in AgRPΔRaptor mice.
Collapse
Affiliation(s)
- Gabriel O. de Souza
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, SP, Brazil; (G.O.d.S.); (P.D.S.T.)
| | - Pryscila D. S. Teixeira
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, SP, Brazil; (G.O.d.S.); (P.D.S.T.)
| | - Niels O. S. Câmara
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, SP, Brazil;
| | - Jose Donato
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, SP, Brazil; (G.O.d.S.); (P.D.S.T.)
| |
Collapse
|
8
|
Li C, Zhang Y, Gao F, Zhao N, Wang G, Xu J, Zhao X. Sex differences in serum pigment epithelium-derived factor in healthy individuals. Medicine (Baltimore) 2023; 102:e34368. [PMID: 37713893 PMCID: PMC10508582 DOI: 10.1097/md.0000000000034368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/26/2023] [Indexed: 09/17/2023] Open
Abstract
To investigate sexual dimorphism of serum pigment epithelium-derived factor (PEDF) and its influencing factors in healthy individuals. A total of 162 healthy people (69 males, 93 females) who underwent health examinations in our department were selected. Serum PEDF, estradiol and other metabolic indices were measured, and homeostasis model assessment of insulin resistance (HOMA-IR) and homeostasis model assessment of β-cell function (HOMA-β) were calculated to evaluate insulin resistance and β-cell function, respectively. Subjects were divided into < 50 years and ≥ 50 years groups to explore the sexual dimorphism of serum PEDF in different age groups. We found no statistically significant difference in serum PEDF levels between men and women in total. However, in the group of subjects under 50 years old, men had significantly higher PEDF levels than women (9.32 ± 2.07 μg/mL vs 8.24 ± 2.29 μg/mL, P < .05), and no sex difference was found in the ≥ 50 years group. In women, serum PEDF levels were significantly higher in subjects aged 50 years and over than in those younger than 50 years of age (9.56 ± 3.05 μg/mL vs 8.25 ± 2.30 μg/mL, P < .05). In men, there was no significant difference in serum PEDF levels between the 2 age groups. In women, correlation analysis showed that serum PEDF levels were significantly correlated with body mass index, waist circumference, diastolic blood pressure (DBP), 2-h postprandial glucose, fasting and 2-h postprandial insulin, HOMA-β, HOMA-IR, aminotransferase, triacylglycerol, and estradiol. Elevated triacylglycerol and aminotransferase and decreased estradiol were significant predictors of increased PEDF concentrations in women. There is sexual dimorphism in circulating PEDF levels, which may be related to estrogen status.
Collapse
Affiliation(s)
- Cuiliu Li
- The Second Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, China
| | - Yunna Zhang
- The Second Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, China
| | - Fang Gao
- The Second Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, China
| | - Nairui Zhao
- The Second Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, China
| | - Guangya Wang
- The Second Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, China
| | - Jinxiu Xu
- The Second Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, China
| | - Xiaopeng Zhao
- The Second Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|
9
|
Wasinski F, Tavares MR, Gusmao DO, List EO, Kopchick JJ, Alves GA, Frazao R, Donato J. Central growth hormone action regulates neuroglial and proinflammatory markers in the hypothalamus of male mice. Neurosci Lett 2023; 806:137236. [PMID: 37030549 PMCID: PMC10133206 DOI: 10.1016/j.neulet.2023.137236] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/10/2023]
Abstract
Growth hormone (GH) action in specific neuronal populations regulates neuroendocrine responses, metabolism, and behavior. However, the potential role of central GH action on glial function is less understood. The present study aims to determine how the hypothalamic expression of several neuroglial markers is affected by central GH action in male mice. The dwarf GH- and insulin-like growth factor-1 (IGF-1)-deficient Ghrhrlit/lit mice showed decreased mRNA expression of Nes (Nestin), Gfap, Iba1, Adgre1 (F4/80), and Tnf (TNFα) in the hypothalamus, compared to wild-type animals. In contrast, transgenic overexpression of GH led to high serum GH and IGF-1 levels, and increased hypothalamic expression of Nes, Gfap, Adgre1, Iba1, and Rax. Hepatocyte-specific GH receptor (GHR) knockout mice, which are characterized by high serum GH levels, but reduced IGF-1 secretion, showed increased mRNA expression of Gfap, Iba1, Tnf, and Sox10, demonstrating that the increase in GH levels alters the hypothalamic expression of glial markers associated with neuroinflammation, independently of IGF-1. Conversely, brain-specific GHR knockout mice showed reduced expression of Gfap, Adgre1, and Vim (vimentin), indicating that brain GHR signaling is necessary to mediate GH-induced changes in the expression of several neuroglial markers. In conclusion, the hypothalamic mRNA levels of several neuroglial markers associated with inflammation are directly modulated by GHR signaling in male mice.
Collapse
Affiliation(s)
- Frederick Wasinski
- Department of Neurology and Neurosurgery, Universidade Federal de Sao Paulo, Sao Paulo, SP 04039-032, Brazil
| | - Mariana R Tavares
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Daniela O Gusmao
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Edward O List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Guilherme A Alves
- Department of Anatomy, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-900, Brazil
| | - Renata Frazao
- Department of Anatomy, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-900, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil.
| |
Collapse
|
10
|
Fernández Núñez MB, Campos Suzman L, Maneja R, Bach A, Marquet O, Anguelovski I, Knobel P. The Differences by Sex and Gender in the Relationship Between Urban Greenness and Cardiometabolic Health: A Systematic Review. J Urban Health 2022; 99:1054-1067. [PMID: 36180810 PMCID: PMC9727044 DOI: 10.1007/s11524-022-00685-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In an increasingly urbanized world, where cardiometabolic issues in cities have raised public health concerns, urban greenness is known to be beneficial for some of the most common health issues. However, the examination of the contribution of sex and gender regarding the benefits of urban greenness for people's cardiometabolic health is lacking. For that reason, we conducted a systematic review of previous literature on the topic following the PRISMA methodology. Additionally, we assessed the quality of the included articles, which we found satisfactory as most papers were of very good or good quality. We explored the relationship between urban greenness exposure and cardiovascular risk factors, cardiovascular diseases, and mortality from cardiovascular diseases. Results suggest that urban greenness is protective against cardiovascular risk factors, diseases, and mortality. When stratifying results by sex and gender, findings point to urban greenness being more beneficial for women and females in stroke and cardiovascular risk factors, except for hypertension and lipid accumulation product. On the other hand, males were more protected by urban greenness in terms of cardiovascular diseases and CVD-related mortality, thus proving that sex and gender health inequalities exist. Furthermore, looking towards the future, research needs to use the proper terminology for sex and gender and policy makers should design urban greenness with a gender perspective.
Collapse
Affiliation(s)
| | - Lia Campos Suzman
- Environmental Science and Technology Institute (ICTA), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Roser Maneja
- Forest Science and Technology Center of Catalonia, Ctra. de St. Llorenç de Morunys, km 2, 25280, Solsona, Spain
- Environment and Human Health Laboratory (EH2 Lab), Ctra. de St. Llorenç de Morunys, km 2, 25280, Solsona, Spain
- Geography Department, Autonomous University of Barcelona, Barcelona, Spain
| | - Albert Bach
- Forest Science and Technology Center of Catalonia, Ctra. de St. Llorenç de Morunys, km 2, 25280, Solsona, Spain
- Environment and Human Health Laboratory (EH2 Lab), Ctra. de St. Llorenç de Morunys, km 2, 25280, Solsona, Spain
| | - Oriol Marquet
- Geography Department, Autonomous University of Barcelona, Barcelona, Spain
| | - Isabelle Anguelovski
- Environmental Science and Technology Institute (ICTA), Universitat Autònoma de Barcelona, Barcelona, Spain
- ICREA, Institució Catalana de Recerca I Estudis Avançats, Barcelona, Spain
| | - Pablo Knobel
- Icahn School of Medicine at Mount Sinai, Department of Environmental Medicine and Public Health, 1 Gustave L. Levy Place, Box 1057, New York, NY, 10029, USA.
| |
Collapse
|
11
|
Merone L, Tsey K, Russell D, Nagle C. Mind the Gap: Reporting and Analysis of Sex and Gender in Health Research in Australia, a Cross-Sectional Study. WOMEN'S HEALTH REPORTS (NEW ROCHELLE, N.Y.) 2022; 3:759-767. [PMID: 36185072 PMCID: PMC9518796 DOI: 10.1089/whr.2022.0033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 07/19/2022] [Indexed: 12/20/2022]
Abstract
Introduction Historically, medical studies have underrepresented female participants and most research data have been collected from males and generalized to other genders. This article aims to determine if there is a sex and/or gender gap in recent Australian health research. Methods This descriptive cross-sectional study of the published literature examines recent Australian-based clinical trials for inclusion of sex and gender. Medians and interquartile ranges (IQRs) were calculated for study sample sizes and female:male representation. Proportion of sex and/or gender was analyzed by the clinical specialty of the trials. t-Tests were used to ascertain significance of any difference in recruitment of female and males. Results A total of 88 articles were included in the analysis. Most studies (n = 63) were randomized clinical controlled trials. Overall women constituted 55% (IQR 30% of all participants). Of the 71 mixed-sex studies, only 8.9% (n = 7) analyzed the data by sex. Women were significantly underrepresented in cardiology and nephrology studies and overrepresented in psychiatry, care of the elderly, and orthopedic studies. Conclusions When analyzed by specialty, women are overrepresented in specialties considered to be female patient dominated, such as psychiatry and care of the elderly, and underrepresented in specialties such as cardiology and nephrology. The overrepresentation of women in some specialties can reinforce gender stereotypes, potentially harming women. In addition, exclusion of males from these areas of research may be of disservice to men's health.
Collapse
Affiliation(s)
- Lea Merone
- College of Healthcare Sciences, James Cook University, Townsville, Queensland, Australia
| | - Komla Tsey
- College of Arts, Society and Education, James Cook University, Smithfield, Queensland, Australia
| | - Darren Russell
- Cairns Sexual Health Service, Cairns North, Queensland, Australia
| | - Cate Nagle
- College of Healthcare Sciences, James Cook University, Townsville, Queensland, Australia
| |
Collapse
|
12
|
Fox R. X-linked genes exhibit skewed expression in Sjogren's disease (SjD): a further step toward understanding the female predominance of autoimmune disease. J Mol Med (Berl) 2022; 100:1267-1269. [PMID: 35982186 PMCID: PMC9402745 DOI: 10.1007/s00109-022-02223-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 05/16/2022] [Accepted: 06/13/2022] [Indexed: 11/18/2022]
Affiliation(s)
- Robert Fox
- Division of Rheumatology, Scripps Memorial Hospital and Research Foundation-Ximed, La Jolla, CA, USA.
| |
Collapse
|
13
|
Kinzenbaw DA, Langmack L, Faraci FM. Angiotensin II-induced endothelial dysfunction: Impact of sex, genetic background, and rho kinase. Physiol Rep 2022; 10:e15336. [PMID: 35681278 PMCID: PMC9184751 DOI: 10.14814/phy2.15336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 06/15/2023] Open
Abstract
The renin-angiotensin system (RAS) contributes to vascular disease with multiple cardiovascular risk factors including hypertension. As a major effector within the RAS, angiotensin II (Ang II) activates diverse signaling mechanisms that affect vascular biology. Despite the impact of such vascular pathophysiology, our understanding of the effects of Ang II in relation to the function of endothelial cells is incomplete. Because genetic background and biological sex can be determinants of vascular disease, we performed studies examining the direct effects of Ang II using carotid arteries from male and female mice on two genetic backgrounds, C57BL/6J and FVB/NJ. Although FVB/NJ mice are much less susceptible to atherosclerosis than C57BL/6J, the effects of Ang II on endothelial cells in FVB/NJ are poorly defined. Overnight incubation of isolated arteries with Ang II (10 nmol/L), impaired endothelial function in both strains and sexes by approximately one-half (p < 0.05). To examine the potential mechanistic contribution of Rho kinase (ROCK), we treated arteries with SLX-2119, an inhibitor with high selectivity for ROCK2. In both male and female mice of both strains, SLX-2119 largely restored endothelial function to normal, compared to vessels treated with vehicle. Thus, Ang II-induced endothelial dysfunction was observed in both FVB/NJ and C57BL/6J mice. This effect was sex-independent. In all groups, effects of Ang II were reversed by inhibition of ROCK2 with SLX-2119. These studies provide the first evidence that ROCK2 may be a key contributor to Ang II-induced endothelial dysfunction in both sexes and in mouse strains that differ in relation to other major aspects of vascular disease.
Collapse
Affiliation(s)
- Dale A. Kinzenbaw
- Departments of Internal MedicineFrancois M. Abboud Cardiovascular CenterThe University of Iowa Carver College of MedicineIowa CityIowaUSA
| | - Lucy Langmack
- Departments of Internal MedicineFrancois M. Abboud Cardiovascular CenterThe University of Iowa Carver College of MedicineIowa CityIowaUSA
| | - Frank M. Faraci
- Departments of Internal MedicineFrancois M. Abboud Cardiovascular CenterThe University of Iowa Carver College of MedicineIowa CityIowaUSA
- Departments of Neuroscience and PharmacologyThe University of Iowa Carver College of MedicineIowa CityIowaUSA
| |
Collapse
|
14
|
de Souza GO, Wasinski F, Donato J. Characterization of the metabolic differences between male and female C57BL/6 mice. Life Sci 2022; 301:120636. [PMID: 35568227 DOI: 10.1016/j.lfs.2022.120636] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 01/22/2023]
Abstract
AIMS The present study aims to compare the responses between male and female C57BL/6 mice to multiple metabolic challenges to understand the importance of sex in the control of energy homeostasis. MAIN METHODS Male and female C57BL/6 mice were subjected to nutritional and hormonal challenges, such as food restriction and refeeding, diet-induced obesity, feeding response to ghrelin and leptin, ghrelin-induced growth hormone secretion, and central responsiveness to ghrelin and leptin. The hypothalamic expression of transcripts that control energy homeostasis was also evaluated. KEY FINDINGS Male mice lost more weight and lean body mass in response to food restriction, compared to females. During refeeding, males accumulated more body fat and exhibited lower energy expenditure and glycemia, as compared to females. Additionally, female mice exhibited a higher protection against diet-induced obesity and related metabolic imbalances in comparison to males. Low dose ghrelin injection elicited higher food intake and growth hormone secretion in male mice, whereas the acute anorexigenic effect of leptin was more robust in females. However, the sex differences in the feeding responses to ghrelin and leptin were not explained by variations in the central responsiveness to these hormones nor by differences in the fiber density from arcuate nucleus neurons. Female, but not male, mice exhibited compensatory increases in hypothalamic Pomc mRNA levels in response to diet-induced obesity. SIGNIFICANCE Our findings revealed several sexually differentiated responses to metabolic challenges in C57BL/6 mice, highlighting the importance of taking into account sex differences in metabolic studies.
Collapse
Affiliation(s)
- Gabriel O de Souza
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil
| | - Frederick Wasinski
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil
| | - Jose Donato
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil..
| |
Collapse
|
15
|
Li C, Zhang Y, Gao F. Serum Pigment Epithelium-Derived Factor Levels are Associated with Estradiol and Decrease After Adjusting for Alanine Aminotransferase in Chinese Women Based on Multiple Linear Regression Analysis. Diabetes Metab Syndr Obes 2022; 15:2901-2909. [PMID: 36177339 PMCID: PMC9514778 DOI: 10.2147/dmso.s378561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/13/2022] [Indexed: 01/10/2023] Open
Abstract
PURPOSE To assess changes in pigment epithelium-derived factor (PEDF) levels in patients with metabolic syndrome (MetS) and to investigate sexual dimorphism in serum PEDF levels and their relationships with estradiol. METHODS A total of 318 individuals (145 men, 173 women) who underwent health examinations in our department were selected. Serum PEDF, estradiol and other metabolic parameters were determined. Homeostasis model assessment of insulin resistance (HOMA- IR) and homeostasis model assessment of β-cell function (HOMA-β) were calculated to evaluate insulin resistance and β-cell function, respectively. Multiple linear regression analysis was used to analyse the factors influencing serum PEDF. RESULTS Serum PEDF levels were significantly higher in subjects with MetS in both men and women (12.09±2.75 vs 8.97±3.19 μg/mL in men and 11.31±2.79 vs 8.40±2.32 μg/mL in women, MetS vs non-MetS, P<0.001). Correlation analysis showed that serum PEDF levels were significantly correlated with body mass index (BMI), waist circumference, waist-to-hip ratio, diastolic blood pressure (DBP), fasting and 2-h postprandial glucose, fasting and 2-h postprandial insulin, HOMA-β, HOMA-IR, hemoglobin A1c (HbA1c), alanine aminotransferase (ALT), aspartate aminotransferase (AST), uric acid (UA), triacylglycerol (TG) and high-density lipoprotein cholesterol (HDL-C). Elevated ALT, HOMA-IR and TG were significant predictors of increased PEDF concentrations. In women, estradiol was inversely correlated with PEDF levels (r=-0.25, P=0.011), and the association was no longer significant after adjustment for ALT. CONCLUSION PEDF could be used as a biomarker of MetS in both men and women. This study reported for the first time that circulating PEDF displays sexual dimorphism, which could be related to estrogen. The association between estrogen and circulating PEDF levels was attenuated after adjusting for ALT.
Collapse
Affiliation(s)
- Cuiliu Li
- The Second Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, 061001, People’s Republic of China
- Correspondence: Cuiliu Li, Tel +8603172075935, Email
| | - Yunna Zhang
- The Second Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, 061001, People’s Republic of China
| | - Fang Gao
- The Second Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, 061001, People’s Republic of China
| |
Collapse
|
16
|
Keestra SM, Male V, Salali GD. Out of balance: the role of evolutionary mismatches in the sex disparity in autoimmune disease. Med Hypotheses 2021; 151:110558. [PMID: 33964604 DOI: 10.1016/j.mehy.2021.110558] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/07/2021] [Accepted: 03/02/2021] [Indexed: 12/28/2022]
Abstract
Over the past century autoimmune disease incidence has increased rapidly in (post-) industrialised, affluent societies, suggesting that changes in ecology and lifestyle are driving this development. Epidemiological studies show that (i) 80% of autoimmune disease patients are female, (ii) autoimmune diseases co-occur more often in women, and (iii) the incidence of some autoimmune diseases is increasing faster in women than in men. The female preponderance in autoimmunity is most pronounced between puberty and menopause, suggesting that diverging sex hormone levels during the reproductive years are implicated in autoimmune disease development. Using an evolutionary perspective, we build on the hypotheses that female immunity is cyclical in menstruating species and that natural selection shaped the female immune system to optimise the implantation and gestation of a semi-allogeneic foetus. We propose that cyclical immunomodulation and female immune tolerance mechanisms are currently out of balance because of a mismatch between the conditions under which they evolved and (post-)industrialised, affluent lifestyles. We suggest that current changes in autoimmune disease prevalence may be caused by increases in lifetime exposure to cyclical immunomodulation and ovarian hormone exposure, reduced immune challenges, increased reproductive lifespan, changed reproductive patterns, and enhanced positive energy balance associated with (post-)industrialised, affluent lifestyles. We discuss proximate mechanisms by which oestrogen and progesterone influence tolerance induction and immunomodulation, and review the effect of the menstrual cycle, pregnancy, and contraceptive use on autoimmune disease incidence and symptoms.
Collapse
Affiliation(s)
- Sarai M Keestra
- Amsterdam UMC, University of Amsterdam, the Netherlands; Department of Global Health & Development, London School of Hygiene and Tropical Medicine, UK.
| | - Victoria Male
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, UK
| | | |
Collapse
|
17
|
Vandel J, Dubois-Chevalier J, Gheeraert C, Derudas B, Raverdy V, Thuillier D, Gaal L, Francque S, Pattou F, Staels B, Eeckhoute J, Lefebvre P. Hepatic Molecular Signatures Highlight the Sexual Dimorphism of Nonalcoholic Steatohepatitis (NASH). Hepatology 2021; 73:920-936. [PMID: 32394476 PMCID: PMC8048532 DOI: 10.1002/hep.31312] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Nonalcoholic steatohepatitis (NASH) is considered as a pivotal stage in nonalcoholic fatty liver disease (NAFLD) progression, given that it paves the way for severe liver injuries such as fibrosis and cirrhosis. The etiology of human NASH is multifactorial, and identifying reliable molecular players and/or biomarkers has proven difficult. Together with the inappropriate consideration of risk factors revealed by epidemiological studies (altered glucose homeostasis, obesity, ethnicity, sex, etc.), the limited availability of representative NASH cohorts with associated liver biopsies, the gold standard for NASH diagnosis, probably explains the poor overlap between published "omics"-defined NASH signatures. APPROACH AND RESULTS Here, we have explored transcriptomic profiles of livers starting from a 910-obese-patient cohort, which was further stratified based on stringent histological characterization, to define "NoNASH" and "NASH" patients. Sex was identified as the main factor for data heterogeneity in this cohort. Using powerful bootstrapping and random forest (RF) approaches, we identified reliably differentially expressed genes participating in distinct biological processes in NASH as a function of sex. RF-calculated gene signatures identified NASH patients in independent cohorts with high accuracy. CONCLUSIONS This large-scale analysis of transcriptomic profiles from human livers emphasized the sexually dimorphic nature of NASH and its link with fibrosis, calling for the integration of sex as a major determinant of liver responses to NASH progression and responses to drugs.
Collapse
Affiliation(s)
- Jimmy Vandel
- Univ. LilleInserm, CHU LilleInstitut Pasteur de LilleU1011-EGIDLilleFrance
| | | | - Céline Gheeraert
- Univ. LilleInserm, CHU LilleInstitut Pasteur de LilleU1011-EGIDLilleFrance
| | - Bruno Derudas
- Univ. LilleInserm, CHU LilleInstitut Pasteur de LilleU1011-EGIDLilleFrance
| | | | | | - Luc Gaal
- Department of Endocrinology, Diabetology and MetabolismAntwerp University HospitalEdegem (Antwerp)Belgium.,Laboratory of Experimental Medicine and Pediatrics (LEMP)University of AntwerpWilrijk (Antwerp)Belgium
| | - Sven Francque
- Laboratory of Experimental Medicine and Pediatrics (LEMP)University of AntwerpWilrijk (Antwerp)Belgium.,Department of Gastroenterology and HepatologyAntwerp University HospitalEdegem (Antwerp)Belgium
| | | | - Bart Staels
- Univ. LilleInserm, CHU LilleInstitut Pasteur de LilleU1011-EGIDLilleFrance
| | - Jérôme Eeckhoute
- Univ. LilleInserm, CHU LilleInstitut Pasteur de LilleU1011-EGIDLilleFrance
| | - Philippe Lefebvre
- Univ. LilleInserm, CHU LilleInstitut Pasteur de LilleU1011-EGIDLilleFrance
| |
Collapse
|
18
|
Santos AC, Conley AJ, Oliveira MF, Assis Neto AC. Steroidogenesis during prenatal testicular development in Spix's cavy Galea spixii. Reprod Fertil Dev 2021; 33:392-400. [PMID: 33685580 DOI: 10.1071/rd20293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 01/28/2021] [Indexed: 12/21/2022] Open
Abstract
Spix's cavy is a potentially good experimental model for research on reproductive biology and sexual development. The aim of the present study was to evaluate the ontogeny of the steroidogenic enzymes involved in testicular androgen synthesis during prenatal development. Testes were investigated on Days 25, 30, 40 and >50 of gestation. Immunohistochemistry and immunoblotting were used to establish the site and relative amount of androgenic enzymes, including 5α-reductase, cytosolic 17β-hydroxysteroid dehydrogenase (17β-HSDI) and mitochondrial microsomal 3β-hydroxysteroid dehydrogenase (3β-HSDII), throughout prenatal development. The testicular parenchyma began to organise on Day 25 of gestation, with the development of recognisable testicular cords. The mesonephros was established after Day 25 of gestation and the ducts differentiated to form the epididymis, as testicular cords were beginning to proliferate and the interstitium to organise by Day 30 of gestation, continuing thereafter. The androgen-synthesising enzymes 5α-reductase, 17β-HSDI and 3β-HSDII were evident in Leydig cells as they differentiated at all subsequent gestational ages studied. In addition, immunoblotting showed an increase in immunoreactivity for the enzymes at Days 30 and 40 of gestation (P<0.05) and a decrease at Day 50 of gestation (P<0.05). It is concluded that the increase in androgenic enzymes in Leydig cells coincides with the functional differentiation of the testes, and with the stabilisation and differentiation of mesonephric ducts forming the epididymis.
Collapse
Affiliation(s)
- A C Santos
- School of Veterinary Medicine and Animal Science, University of Sao Paulo. Av. Prof. Dr. Orlando de Marques Paiva, 87; ZC 05508 270; São Paulo - Brazil
| | - A J Conley
- Population Health & Reproduction, School of Veterinary Medicine, University of California, 3223 VM3B, Davis, CA 95616, USA
| | - M F Oliveira
- Department of Animal Science, Federal Rural University of Semiarid. Av. Francisco Mota, 572, 59625 900, Mossoro, Rio Grande do Norte, Brazil
| | - A C Assis Neto
- School of Veterinary Medicine and Animal Science, University of Sao Paulo. Av. Prof. Dr. Orlando de Marques Paiva, 87; ZC 05508 270; São Paulo - Brazil; and Corresponding author.
| |
Collapse
|
19
|
Tebani A, Gummesson A, Zhong W, Koistinen IS, Lakshmikanth T, Olsson LM, Boulund F, Neiman M, Stenlund H, Hellström C, Karlsson MJ, Arif M, Dodig-Crnković T, Mardinoglu A, Lee S, Zhang C, Chen Y, Olin A, Mikes J, Danielsson H, von Feilitzen K, Jansson PA, Angerås O, Huss M, Kjellqvist S, Odeberg J, Edfors F, Tremaroli V, Forsström B, Schwenk JM, Nilsson P, Moritz T, Bäckhed F, Engstrand L, Brodin P, Bergström G, Uhlen M, Fagerberg L. Integration of molecular profiles in a longitudinal wellness profiling cohort. Nat Commun 2020; 11:4487. [PMID: 32900998 PMCID: PMC7479148 DOI: 10.1038/s41467-020-18148-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 08/03/2020] [Indexed: 12/19/2022] Open
Abstract
An important aspect of precision medicine is to probe the stability in molecular profiles among healthy individuals over time. Here, we sample a longitudinal wellness cohort with 100 healthy individuals and analyze blood molecular profiles including proteomics, transcriptomics, lipidomics, metabolomics, autoantibodies and immune cell profiling, complemented with gut microbiota composition and routine clinical chemistry. Overall, our results show high variation between individuals across different molecular readouts, while the intra-individual baseline variation is low. The analyses show that each individual has a unique and stable plasma protein profile throughout the study period and that many individuals also show distinct profiles with regards to the other omics datasets, with strong underlying connections between the blood proteome and the clinical chemistry parameters. In conclusion, the results support an individual-based definition of health and show that comprehensive omics profiling in a longitudinal manner is a path forward for precision medicine. An important aspect of precision medicine is to probe the stability in molecular profiles among healthy individuals over time. Here, the authors sample a longitudinal wellness cohort and analyse blood molecular profiles as well as gut microbiota composition.
Collapse
Affiliation(s)
- Abdellah Tebani
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Anders Gummesson
- Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Genetics and Genomics, Gothenburg, Sweden
| | - Wen Zhong
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Ina Schuppe Koistinen
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden.,Center for Translational Microbiome Research, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Tadepally Lakshmikanth
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Lisa M Olsson
- Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Boulund
- Center for Translational Microbiome Research, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Maja Neiman
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Hans Stenlund
- Swedish Metabolomics Centre, Department of Molecular Biology, Umeå University, 901 87, Umeå, Sweden
| | - Cecilia Hellström
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Max J Karlsson
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Muhammad Arif
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Tea Dodig-Crnković
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden.,Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| | - Sunjae Lee
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Yang Chen
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Axel Olin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Jaromir Mikes
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Hanna Danielsson
- Center for Translational Microbiome Research, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Kalle von Feilitzen
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Per-Anders Jansson
- Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital, Department of Internal Medicine, Gothenburg, Sweden
| | - Oskar Angerås
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital, Department of Cardiology, Gothenburg, Sweden
| | - Mikael Huss
- Codon Consulting, 118 26, Stockholm, Sweden.,Department of Learning, Informatics, Management and Ethics, Karolinska Institutet, Stockholm, Sweden
| | - Sanela Kjellqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jacob Odeberg
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Fredrik Edfors
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Valentina Tremaroli
- Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Björn Forsström
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Jochen M Schwenk
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Peter Nilsson
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Thomas Moritz
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, 907 36, Umeå, Sweden
| | - Fredrik Bäckhed
- Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Physiology, Gothenburg, Sweden.,Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Engstrand
- Center for Translational Microbiome Research, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Petter Brodin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Göran Bergström
- Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Physiology, Gothenburg, Sweden
| | - Mathias Uhlen
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden.,Center for Biosustainability, Danish Technical University, Copenhagen, Denmark
| | - Linn Fagerberg
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
20
|
Loh R, Stamatakis E, Folkerts D, Allgrove JE, Moir HJ. Effects of Interrupting Prolonged Sitting with Physical Activity Breaks on Blood Glucose, Insulin and Triacylglycerol Measures: A Systematic Review and Meta-analysis. Sports Med 2020; 50:295-330. [PMID: 31552570 PMCID: PMC6985064 DOI: 10.1007/s40279-019-01183-w] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Physical activity (PA) breaks in sitting time might attenuate metabolic markers relevant to the prevention of type 2 diabetes. OBJECTIVES The primary aim of this paper was to systematically review and meta-analyse trials that compared the effects of breaking up prolonged sitting with bouts of PA throughout the day (INT) versus continuous sitting (SIT) on glucose, insulin and triacylglycerol (TAG) measures. A second aim was to compare the effects of INT versus continuous exercise (EX) on glucose, insulin and TAG measures. METHODS The review followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) recommendations. Eligibility criteria consisted of trials comparing INT vs. SIT or INT vs. one bout of EX before or after sitting, in participants aged 18 or above, who were classified as either metabolically healthy or impaired, but not with other major health conditions such as chronic obstructive pulmonary disease or peripheral arterial disease. RESULTS A total of 42 studies were included in the overall review, whereas a total of 37 studies were included in the meta-analysis. There was a standardised mean difference (SMD) of - 0.54 (95% CI - 0.70, - 0.37, p = 0.00001) in favour of INT compared to SIT for glucose. With respect to insulin, there was an SMD of - 0.56 (95% CI - 0.74, - 0.38, p = 0.00001) in favour of INT. For TAG, there was an SMD of - 0.26 (95% CI - 0.44, - 0.09, p = 0.002) in favour of INT. Body mass index (BMI) was associated with glucose responses (β = - 0.05, 95% CI - 0.09, - 0.01, p = 0.01), and insulin (β = - 0.05, 95% CI - 0.10, - 0.006, p = 0.03), but not TAG (β = 0.02, 95% CI - 0.02, 0.06, p = 0.37). When energy expenditure was matched, there was an SMD of - 0.26 (95% CI - 0.50, - 0.02, p = 0.03) in favour of INT for glucose, but no statistically significant SMDs for insulin, i.e. 0.35 (95% CI - 0.37, 1.07, p = 0.35), or TAG i.e. 0.08 (95% CI - 0.22, 0.37, p = 0.62). It is worth noting that there was possible publication bias for TAG outcomes when PA breaks were compared with sitting. CONCLUSION The use of PA breaks during sitting moderately attenuated post-prandial glucose, insulin, and TAG, with greater glycaemic attenuation in people with higher BMI. There was a statistically significant small advantage for PA breaks over continuous exercise for attenuating glucose measures when exercise protocols were energy matched, but no statistically significant differences for insulin and TAG. PROSPERO Registration: CRD42017080982. PROSPERO REGISTRATION CRD42017080982.
Collapse
Affiliation(s)
- Roland Loh
- School of Life Sciences, Pharmacy and Chemistry, Faculty of Science, Engineering and Computing, Kingston University, Penrhyn Road, Kingston upon Thames, Surrey, London, KT1 2EE, UK.
| | - Emmanuel Stamatakis
- Charles Perkins Centre, Prevention Research Collaboration, School of Public Health, University of Sydney, Sydney, NSW, Australia
| | - Dirk Folkerts
- School of Life Sciences, Pharmacy and Chemistry, Faculty of Science, Engineering and Computing, Kingston University, Penrhyn Road, Kingston upon Thames, Surrey, London, KT1 2EE, UK.,Faculty of Sport and Exercise Sciences, University of Muenster, Münster, Germany
| | - Judith E Allgrove
- School of Life Sciences, Pharmacy and Chemistry, Faculty of Science, Engineering and Computing, Kingston University, Penrhyn Road, Kingston upon Thames, Surrey, London, KT1 2EE, UK
| | - Hannah J Moir
- School of Life Sciences, Pharmacy and Chemistry, Faculty of Science, Engineering and Computing, Kingston University, Penrhyn Road, Kingston upon Thames, Surrey, London, KT1 2EE, UK.
| |
Collapse
|
21
|
Kirlangic OF, Yilmaz-Oral D, Kaya-Sezginer E, Toktanis G, Tezgelen AS, Sen E, Khanam A, Oztekin CV, Gur S. The Effects of Androgens on Cardiometabolic Syndrome: Current Therapeutic Concepts. Sex Med 2020; 8:132-155. [PMID: 32201216 PMCID: PMC7261691 DOI: 10.1016/j.esxm.2020.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/31/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Cardiometabolic syndrome (CMS), as a bunch of metabolic disorders mainly characterized by type 2 diabetes mellitus (T2DM), hypertension, atherosclerosis, central adiposity, and abdominal obesity triggering androgen deficiency, is one of the most critical threats to men. Although many significant preclinical and clinical findings explain CMS, new approaches toward common pathophysiological mechanisms and reasonable therapeutic targets are lacking. AIM To gain a further understanding of the role of androgen levels in various facets of CMS such as the constellation of cardiometabolic risk factors including central adiposity, dyslipidemia, insulin resistance, diabetes, and arterial hypertension and to define future directions for development of effective therapeutic modalities. METHODS Clinical and experimental data were searched through scientific literature databases (PubMed) from 2009 to October 2019. MAIN OUTCOME MEASURE Evidence from basic and clinical research was gathered with regard to the causal impact and therapeutic roles of androgens on CMS. RESULTS There are important mechanisms implicated in androgen levels and the risk of CMS. Low testosterone levels have many signs and symptoms on cardiometabolic and glycometabolic risks as well as abdominal obesity in men. CLINICAL IMPLICATIONS The implications of the findings can shed light on future improvements in androgen levels and add potentially predictive risk for CMS, as well as T2DM, abdominal obesity to guide clinical management in the early stage. STRENGTHS & LIMITATIONS This comprehensive review refers to the association between androgens and cardiovascular health. A limitation of this study is the lack of large, prospective population-based studies that analyze the effects of testosterone treatment on CMS or mortality. CONCLUSION Low testosterone levels have several common features with metabolic syndrome. Thus, testosterone may have preventive role in the progress of metabolic syndrome and subsequent T2DM, abdominal obesity, and cardiovascular disease and likely affect aging men's health mainly through endocrine and vascular mechanisms. Further studies are necessary to evaluate the therapeutic interventions directed at preventing CMS in men. Kirlangic OF, Yilmaz-Oral D, Kaya-Sezginer E, et al. The Effects of Androgens on Cardiometabolic Syndrome: Current Therapeutic Concepts. Sex Med 2020;8:132-155.
Collapse
Affiliation(s)
- Omer Faruk Kirlangic
- University of Health Sciences, Gulhane Faculty of Medicine, Department of Medical Biochemistry, Ankara, Turkey
| | - Didem Yilmaz-Oral
- Cukurova University, Faculty of Pharmacy, Department of Pharmacology, Adana, Turkey
| | - Ecem Kaya-Sezginer
- Ankara University, Faculty of Pharmacy, Department of Biochemistry, Ankara, Turkey
| | - Gamze Toktanis
- Ankara University, Faculty of Pharmacy, Department of Pharmacology, Ankara, Turkey
| | | | - Ekrem Sen
- Ankara University, Faculty of Pharmacy, Department of Pharmacology, Ankara, Turkey
| | - Armagan Khanam
- Ankara University, Faculty of Pharmacy, Department of Pharmacology, Ankara, Turkey
| | - Cetin Volkan Oztekin
- Department of Urology, Faculty of Medicine, University of Kyrenia, Girne, Turkish Republic of North Cyprus, Mersin 10, Turkey
| | - Serap Gur
- Ankara University, Faculty of Pharmacy, Department of Pharmacology, Ankara, Turkey.
| |
Collapse
|
22
|
Brandenburg JS, Clark RM, Coffman B, Sharma G, Hathaway HJ, Prossnitz ER, Howdieshell TR. Sex differences in murine myocutaneous flap revascularization. Wound Repair Regen 2020; 28:470-479. [PMID: 32428975 DOI: 10.1111/wrr.12812] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 12/01/2019] [Accepted: 01/03/2020] [Indexed: 12/22/2022]
Abstract
Sex differences in susceptibility to ischemia/reperfusion injury have been documented in humans. Premenopausal women have a lower risk of ischemic heart disease than age-matched men, whereas after menopause, the risk is similar or even higher in women. However, little is known about the effects of sex on myocutaneous ischemia/reperfusion. To explore sex differences in wound revascularization, we utilized a murine myocutaneous flap model of graded ischemia. A cranial-based, peninsular-shaped, myocutaneous flap was surgically created on the dorsum of male and female mice. Physiological, pathological, immunohistochemical, and molecular parameters were analyzed. Flaps created on female mice were re-attached to the recipient site resulting in nearly complete viability at post-operative day 10. In contrast, distal full-thickness myocutaneous necrosis was evident at 10 days post-surgery in male mice. Over the 10 day study interval, laser speckle imaging documented functional revascularization in all flap regions in female mice, but minimal distal flap reperfusion in male mice. Day 10 immunostained histologic sections confirmed significant increases in distal flap vessel count and vascular surface area in female compared to male mice. RT-PCR demonstrated significant differences in growth factor and metabolic gene expression between female and male mice at day 10. In conclusion, in a graded-ischemia wound healing model, flap revascularization was more effective in female mice. The recognition and identification of sex-specific wound healing differences may lead to a better understanding of the underlying mechanisms of myocutaneous revascularization and drive novel discovery to improve soft tissue wound healing following tissue transfer for traumatic injury and cancer resection.
Collapse
Affiliation(s)
- Jacquelyn S Brandenburg
- Department of Surgery, AU/UGA Medical Partnership, University of Georgia Health Sciences Campus, Athens, GA
| | - Ross M Clark
- Department of Surgery, AU/UGA Medical Partnership, University of Georgia Health Sciences Campus, Athens, GA
| | - Brittany Coffman
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Geetanjali Sharma
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Helen J Hathaway
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Eric R Prossnitz
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Thomas R Howdieshell
- Department of Surgery, AU/UGA Medical Partnership, University of Georgia Health Sciences Campus, Athens, GA
| |
Collapse
|
23
|
Cui M, Xiao H, Li Y, Zhang S, Dong J, Wang B, Zhu C, Jiang M, Zhu T, He J, Wang H, Fan S. Sexual Dimorphism of Gut Microbiota Dictates Therapeutics Efficacy of Radiation Injuries. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901048. [PMID: 31728280 PMCID: PMC6839645 DOI: 10.1002/advs.201901048] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/14/2019] [Indexed: 05/15/2023]
Abstract
Accidental or iatrogenic ionizing radiation exposure precipitates acute and chronic radiation injuries. The traditional paradigm of mitigating radiotherapy-associated adverse side effects has ignored the gender-specific dimorphism of patients' divergent responses. Here, the effects of sexual dimorphism on curative efficiencies of therapeutic agents is examined in murine models of irradiation injury. Oral gavage of simvastatin ameliorates radiation-induced hematopoietic injury and gastrointestinal tract dysfunction in male mice, but adversely deteriorates these radiation syndromes in female animals. In a sharp contrast, feeding animals with high-fat diet (HFD) elicites explicitly contrary results. High-throughput sequencing of microbial 16S rRNA, host miRNA, and mRNA shows that simvastatin or HFD administration preventes radiation-altered enteric bacterial taxonomic structure, preserves miRNA expression profile, and reprogrammes the spectrum of mRNA expression in small intestines of male or female mice, respectively. Notably, faecal microbiota transplantation of gut microbes from opposite sexual donors abrogates the curative effects of simvastatin or HFD in respective genders of animals. Together, these findings demonstrate that curative efficiencies of therapeutic strategies mitigating radiation toxicity might be dependent on the gender of patients, thus simvastatin or HFD might be specifically useful for fighting against radiation toxicity in a sex-dependent fashion partly based on sex-distinct gut microbiota composition in preclinical settings.
Collapse
Affiliation(s)
- Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine Chinese Academy of Medical Sciences and Peking Union Medical College 238 Baidi Road Tianjin 300192 China
| | - Huiwen Xiao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine Chinese Academy of Medical Sciences and Peking Union Medical College 238 Baidi Road Tianjin 300192 China
| | - Yuan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine Chinese Academy of Medical Sciences and Peking Union Medical College 238 Baidi Road Tianjin 300192 China
| | - Shuqin Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine Chinese Academy of Medical Sciences and Peking Union Medical College 238 Baidi Road Tianjin 300192 China
| | - Jiali Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine Chinese Academy of Medical Sciences and Peking Union Medical College 238 Baidi Road Tianjin 300192 China
| | - Bin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine Chinese Academy of Medical Sciences and Peking Union Medical College 238 Baidi Road Tianjin 300192 China
| | - Changchun Zhu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine Chinese Academy of Medical Sciences and Peking Union Medical College 238 Baidi Road Tianjin 300192 China
| | - Mian Jiang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine Chinese Academy of Medical Sciences and Peking Union Medical College 238 Baidi Road Tianjin 300192 China
| | - Tong Zhu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine Chinese Academy of Medical Sciences and Peking Union Medical College 238 Baidi Road Tianjin 300192 China
| | - Junbo He
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine Chinese Academy of Medical Sciences and Peking Union Medical College 238 Baidi Road Tianjin 300192 China
| | - Haichao Wang
- Laboratory of Emergency Medicine Feinstein Institute for Medical Research Manhasset NY 11030 USA
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine Chinese Academy of Medical Sciences and Peking Union Medical College 238 Baidi Road Tianjin 300192 China
| |
Collapse
|
24
|
Camporez JP, Lyu K, Goldberg EL, Zhang D, Cline GW, Jurczak MJ, Dixit VD, Petersen KF, Shulman GI. Anti-inflammatory effects of oestrogen mediate the sexual dimorphic response to lipid-induced insulin resistance. J Physiol 2019; 597:3885-3903. [PMID: 31206703 DOI: 10.1113/jp277270] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 05/20/2019] [Indexed: 12/15/2022] Open
Abstract
KEY POINTS Oestrogen has been shown to play an important role in the regulation of metabolic homeostasis and insulin sensitivity in both human and rodent studies. Insulin sensitivity is greater in premenopausal women compared with age-matched men, and metabolism-related cardiovascular diseases and type 2 diabetes are less frequent in these same women. Both female and male mice treated with oestradiol are protected against obesity-induced insulin resistance. The protection against obesity-induced insulin resistance is associated with reduced ectopic lipid content in liver and skeletal muscle. These results were associated with increased insulin-stimulated suppression of white adipose tissue lipolysis and reduced inflammation. ABSTRACT Oestrogen has been shown to play an important role in the regulation of metabolic homeostasis and insulin sensitivity in both human and rodent studies. Overall, females are protected against obesity-induced insulin resistance; yet, the mechanisms responsible for this protection are not well understood. Therefore, the aim of the present work was to evaluate the underlying mechanism(s) by which female mice are protected against obesity-induced insulin resistance compared with male mice. We studied male and female mice in age-matched or body weight-matched conditions. They were fed a high-fat diet (HFD) or regular chow for 4 weeks. We also studied HFD male mice treated with oestradiol or vehicle. Both HFD female and HFD male mice treated with oestradiol displayed increased whole-body insulin sensitivity, associated with reduction in ectopic hepatic and muscle lipid content compared to HFD male mice. Reductions in ectopic lipid content in these mice were associated with increased insulin-stimulated suppression of white adipose tissue (WAT) lipolysis. Both HFD female and HFD male mice treated with oestradiol also displayed striking reductions in WAT inflammation, represented by reductions in plasma and adipose tissue tumour necrosis factor α and interleukin 6 concentrations. Taken together these data support the hypothesis that HFD female mice are protected from obesity-induced insulin resistance due to oestradiol-mediated reductions in WAT inflammation, leading to improved insulin-mediated suppression of WAT lipolysis and reduced ectopic lipid content in liver and skeletal muscle.
Collapse
Affiliation(s)
- João Paulo Camporez
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil, 05508-000
| | - Kun Lyu
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Emily L Goldberg
- Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Dongyan Zhang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Gary W Cline
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Michael J Jurczak
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Vishwa Deep Dixit
- Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Kitt Falk Petersen
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT, 06520, USA.,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, 06520, USA
| |
Collapse
|
25
|
Ramos-Lobo AM, Furigo IC, Teixeira PDS, Zampieri TT, Wasinski F, Buonfiglio DC, Donato J. Maternal metabolic adaptations are necessary for normal offspring growth and brain development. Physiol Rep 2019. [PMID: 29536670 PMCID: PMC5849578 DOI: 10.14814/phy2.13643] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Several metabolic adaptations emerge during pregnancy and continue through lactation, including increases in food intake and body weight, as well as insulin and leptin resistance. These maternal adaptations are thought to play a role in offspring viability and success. Using a model of attenuated maternal metabolic adaptations induced by ablation of the Socs3 gene in leptin receptor expressing cells (SOCS3 KO mice), our study aimed to investigate whether maternal metabolic changes are required for normal offspring development, and if their absence causes metabolic imbalances in adulthood. The litters were subjected to a cross‐fostering experimental design to distinguish the prenatal and postnatal effects caused by maternal metabolic adaptations. Males either born or raised by SOCS3 KO mice showed reduced body weight until 8 weeks of life. Both adult males and females born or raised by SOCS3 KO mice also had lower body adiposity. Despite that, no significant changes in energy expenditure, glucose tolerance or insulin resistance were observed. However, males either born or raised by SOCS3 KO mice showed reduced brain mass in adulthood. Furthermore, animals born from SOCS3 KO mice also had lower proopiomelanocortin fiber density in the paraventricular nucleus of the hypothalamus. In conclusion, these findings indicate that the commonly observed metabolic changes in pregnancy and lactation are necessary for normal offspring growth and brain development.
Collapse
Affiliation(s)
- Angela M Ramos-Lobo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Isadora C Furigo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Pryscila D S Teixeira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Thais T Zampieri
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Frederick Wasinski
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Daniella C Buonfiglio
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
26
|
González-García I, Contreras C, Estévez-Salguero Á, Ruíz-Pino F, Colsh B, Pensado I, Liñares-Pose L, Rial-Pensado E, Martínez de Morentin PB, Fernø J, Diéguez C, Nogueiras R, Le Stunff H, Magnan C, Tena-Sempere M, López M. Estradiol Regulates Energy Balance by Ameliorating Hypothalamic Ceramide-Induced ER Stress. Cell Rep 2018; 25:413-423.e5. [PMID: 30304681 PMCID: PMC6198289 DOI: 10.1016/j.celrep.2018.09.038] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/22/2018] [Accepted: 09/11/2018] [Indexed: 12/12/2022] Open
Abstract
Compelling evidence has shown that, besides its putative effect on the regulation of the gonadal axis, estradiol (E2) exerts a dichotomic effect on the hypothalamus to regulate food intake and energy expenditure. The anorectic effect of E2 is mainly mediated by its action on the arcuate nucleus (ARC), whereas its effects on brown adipose tissue (BAT) thermogenesis occur in the ventromedial nucleus (VMH). Here, we demonstrate that central E2 decreases hypothalamic ceramide levels and endoplasmic reticulum (ER) stress. Pharmacological or genetic blockade of ceramide synthesis and amelioration of ER stress selectively occurring in the VMH recapitulate the effect of E2, leading to increased BAT thermogenesis, weight loss, and metabolic improvement. These findings demonstrate that E2 regulation of ceramide-induced hypothalamic lipotoxicity and ER stress is an important determinant of energy balance, suggesting that dysregulation of this mechanism may underlie some changes in energy homeostasis seen in females.
Collapse
Affiliation(s)
- Ismael González-García
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Cristina Contreras
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Ánxela Estévez-Salguero
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Francisco Ruíz-Pino
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, 14004, Spain; Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Reina Sofía, Córdoba, 14004, Spain
| | - Benoit Colsh
- CEA-Centre d'Etude de Saclay, Laboratoire d'étude du Métabolisme des Médicaments, Gif-sur-Yvette, France
| | - Iván Pensado
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Laura Liñares-Pose
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Eva Rial-Pensado
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Pablo B Martínez de Morentin
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Johan Fernø
- Hormone Laboratory, Haukeland University Hospital, Bergen, 5021, Norway
| | - Carlos Diéguez
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Rubén Nogueiras
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Hervé Le Stunff
- Paris-Saclay Institute of Neuroscience, CNRS UMR 9197, Université Paris-Sud, University Paris Saclay, Orsay 91405 Cedex, France; Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Université Paris Diderot, Sorbonne Paris Cité, Paris, 75205, France
| | - Christophe Magnan
- Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Université Paris Diderot, Sorbonne Paris Cité, Paris, 75205, France
| | - Manuel Tena-Sempere
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, 14004, Spain; Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Reina Sofía, Córdoba, 14004, Spain; FiDiPro Program, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland
| | - Miguel López
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain.
| |
Collapse
|
27
|
Cui C, Huang C, Liu K, Xu G, Yang J, Zhou Y, Feng Y, Kararigas G, Geng B, Cui Q. Large-scale in silico identification of drugs exerting sex-specific effects in the heart. J Transl Med 2018; 16:236. [PMID: 30157868 PMCID: PMC6116388 DOI: 10.1186/s12967-018-1612-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 08/21/2018] [Indexed: 02/07/2023] Open
Abstract
Background Major differences exist between men and women in both physiology and pathophysiology. Dissecting the underlying processes and contributing mechanisms of sex differences in health and disease represents a crucial step towards precision medicine. Considering the significant differences between men and women in the response to pharmacotherapies, our aim was to develop an in silico model able to predict sex-specific drug responses in a large-scale. Methods For this purpose, we focused on cardiovascular effects because of their high morbidity and mortality. Our model predicted several drugs (including acebutolol and tacrine) with significant differences in the heart between men and women. To validate the sex-specific drug responses identified by our model, acebutolol was selected to lower blood pressure in spontaneous hypertensive rats (SHR), tacrine was used to assess cardiac injury in mice and metformin as control for a non-sex-specific response. Results As our model predicted, acebutolol exhibited a stronger decrease in heart rate and blood pressure in female than male SHRs. Tacrine lowered heart rate in male but not in female mice, induced higher plasma cTNI level and increased cardiac superoxide (DHE staining) generation in female than male mice, indicating stronger cardiac toxicity in female than male mice. To validate our model in humans, we employed two Chinese cohorts, which showed that among patients taking a beta-receptor blocker (metoprolol), women reached significantly lower diastolic blood pressure than men. Conclusions We conclude that our in silico model could be translated into clinical practice to predict sex-specific drug responses, thereby contributing towards a more appropriate medical care for both men and women. Electronic supplementary material The online version of this article (10.1186/s12967-018-1612-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Changting Cui
- Department of Physiology and Pathophysiology, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, 100191, China.,Department of Biomedical Informatics, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, 100191, China
| | - Chuanbo Huang
- Department of Physiology and Pathophysiology, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, 100191, China.,Department of Biomedical Informatics, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, 100191, China.,School of Mathematics Sciences, Huaqiao University, 269 Chenghua North Rd, Quanzhou, 362021, China
| | - Kejia Liu
- Ruike-Donghua Translational Medicine Research Center, Beijing, 100176, China
| | - Guoheng Xu
- Department of Physiology and Pathophysiology, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, 100191, China.,Department of Biomedical Informatics, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, 100191, China
| | - Jichun Yang
- Department of Physiology and Pathophysiology, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, 100191, China.,Department of Biomedical Informatics, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, 100191, China
| | - Yong Zhou
- Department of Cardiology, Beijing Institute of Heart, Lung and Blood Vascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Yingmei Feng
- Beijing Key Laboratory of Diabetes Prevention and Research, Luhe Hospital, Capital Medical University, Beijing, 101149, China.,Department of Endocrinology, Luhe Hospital, Capital Medical University, Beijing, 101149, China
| | - Georgios Kararigas
- Berlin Institute of Health, Institute of Gender in Medicine and Center for Cardiovascular Research, Charité-Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Bin Geng
- Department of Physiology and Pathophysiology, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, 100191, China. .,Department of Biomedical Informatics, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, 100191, China. .,Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, 100037, China.
| | - Qinghua Cui
- Department of Physiology and Pathophysiology, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, 100191, China. .,Department of Biomedical Informatics, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, 100191, China. .,Center of Bioinformatics, Key Laboratory for Neuro-Information of Ministry of Education, School of Life Science and Technology, Chengdu, 610054, China.
| |
Collapse
|
28
|
An integrated view of sex differences in metabolic physiology and disease. Mol Metab 2018; 15:1-2. [PMID: 30032908 PMCID: PMC6066737 DOI: 10.1016/j.molmet.2018.06.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 06/15/2018] [Indexed: 12/24/2022] Open
|
29
|
Dos Santos AC, Conley AJ, de Oliveira MF, de Assis Neto AC. Development of urogenital system in the Spix cavy: A model for studies on sexual differentiation. Differentiation 2018; 101:25-38. [PMID: 29684807 DOI: 10.1016/j.diff.2018.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 04/04/2018] [Accepted: 04/11/2018] [Indexed: 12/17/2022]
Abstract
This study documented, for the first time, the morphological patterns of differentiation of male and female genital organs of Spix cavy (Galea spixii) using histological and ultrastructural analyses, with immuno-localization of steroidogenic enzymes, cytochromes P450 aromatase (P450arom) and 17α-hydroxylase/17, 20-lyase (P450c17), involved in the synthesis of estrogens and androgens respectively throughout fetal sexual development. Undifferentiated gonads of Spix cavy develop into ovaries in females after 25 days of gestation (DG), exhibiting P450arom immunoreactivity. After 25 DG, paramesonephric ducts develop and form oviducts, uterine horns and cranial portion of the vagina. The caudal portion of the vagina originates from the urogenital sinus, and a vaginal closure membrane is present at the end of gestation. Partial channeling of the urethra into the clitoris occurs after 40 DG, but complete channeling never occurs. A preputial meatus emerges near the tip of organ. In males, undifferentiated gonads develop into testes at 25 DG and develop immunoreactivity for P450c17, which is required for androgens synthesis and likely maintenance of mesonephric ducts. Mesonephric ducts develop subsequently, forming the epididymis and ductus deferens. The pelvic urethra develops after 25 DG with channeling into the penis occurring around 30 DG. This is the first morphological study describing the process of sexual differentiation during gestation in a hystricomorph rodent and one of the most comprehensive analyses conducted in any mammal. Male genital organ development follows the general pattern described in other domestic mammals, but does not include formation of the baculum as occurs in mice and rats. In females, clitoral development includes partial canalization by the urethra and development of a preputial meatus. Further studies are required to clarify the mechanisms involved in the differentiative processes described.
Collapse
Affiliation(s)
- Amilton Cesar Dos Santos
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87 ZC, 05508-270 São Paulo-SP, Brazil
| | - Alan James Conley
- Population Health&Reproduction, School of Veterinary Medicine, University of California, Davis, USA
| | | | - Antônio Chaves de Assis Neto
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87 ZC, 05508-270 São Paulo-SP, Brazil.
| |
Collapse
|
30
|
Lohse Z, Knorr S, Bytoft B, Clausen TD, Jensen RB, Oturai P, Beck-Nielsen H, Gravholt CH, Damm P, Højlund K, Jensen DM. Differential effects of age and sex on insulin sensitivity and body composition in adolescent offspring of women with type 1 diabetes: results from the EPICOM study. Diabetologia 2018; 61:210-219. [PMID: 28971223 DOI: 10.1007/s00125-017-4458-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/15/2017] [Indexed: 12/22/2022]
Abstract
AIMS/HYPOTHESIS The aim of this study was to investigate the influence of age and sex on insulin sensitivity and insulin secretion in the adolescent offspring of women with type 1 diabetes, compared with the background population. METHODS This was a prospective nationwide follow-up study (Epigenetic, Genetic and Environmental Effects on Growth, Cognitive Functions and Metabolism in Offspring of Women with Type 1 Diabetes [EPICOM]) in a Danish population. We examined 278 offspring of women with type 1 diabetes from the Danish Diabetes Association Register born during 1993-1999 (index offspring) and 303 control offspring, identified through the Danish Central Office of Civil Registration and matched to the index offspring with respect to date of birth, sex and postal code. The offspring had an overall mean age of 16.7 years (range 13.0-20.4 years). The main outcomes were age-related changes in fasting OGTT-derived indices for insulin sensitivity (BIGTT-SI0-30-120, Matsuda index, HOMA-IR) and insulin secretion (acute insulin response [BIGTT-AIR0-0-30-120], insulinogenic index, HOMA of insulin secretory function [HOMA-β], disposition index) and physical activity (International Physical Activity Questionnaire). In addition, we determined total body fat (TBF) percentage using dual-energy x-ray absorptiometry. RESULTS We observed significantly lower insulin sensitivity in index offspring compared with control offspring, increasing with age. The differences were attenuated after adjustment for TBF percentage, but were still significant at 17 and 18 years of age. We also observed decreased disposition index and insulin secretion-sensitivity index-2 in index offspring at the same age, but we found no significant differences in other indices of insulin secretion compared with control offspring. With age, TBF percentage became increasingly more divergent between index and control offspring, and was consistently higher among female but not male index offspring. CONCLUSIONS/INTERPRETATION Differences in insulin sensitivity between the offspring of women with type 1 diabetes and control offspring increased with age. This was only partially explained by higher adiposity in the index offspring. TRIAL REGISTRATION ClinicalTrials.gov NCT01559181.
Collapse
Affiliation(s)
- Zuzana Lohse
- Department of Endocrinology, Odense University Hospital, Kløvervænget 6, 4th floor, 5000, Odense C, Denmark
- The Department of Clinical Research, Faculty of Health and Medical Sciences, University of Southern Denmark, Odense, Denmark
| | - Sine Knorr
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Birgitte Bytoft
- Center for Pregnant Women with Diabetes, Rigshospitalet, Copenhagen, Denmark
- Department of Obstetrics, Rigshospitalet, Copenhagen, Denmark
- The Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tine D Clausen
- Department of Gynecology and Obstetrics, Hilleroed Hospital, Hilleroed, Denmark
| | - Rikke B Jensen
- University Department of Growth and Reproduction, Rigshospitalet, Copenhagen, Denmark
| | - Peter Oturai
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen, Denmark
| | - Henning Beck-Nielsen
- Department of Endocrinology, Odense University Hospital, Kløvervænget 6, 4th floor, 5000, Odense C, Denmark
| | - Claus H Gravholt
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Peter Damm
- Center for Pregnant Women with Diabetes, Rigshospitalet, Copenhagen, Denmark
- Department of Obstetrics, Rigshospitalet, Copenhagen, Denmark
- The Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kurt Højlund
- Department of Endocrinology, Odense University Hospital, Kløvervænget 6, 4th floor, 5000, Odense C, Denmark
- Section of Molecular Diabetes & Metabolism, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Dorte M Jensen
- Department of Endocrinology, Odense University Hospital, Kløvervænget 6, 4th floor, 5000, Odense C, Denmark.
- The Department of Clinical Research, Faculty of Health and Medical Sciences, University of Southern Denmark, Odense, Denmark.
- Department of Gynaecology and Obstetrics, Odense University Hospital, Odense, Denmark.
| |
Collapse
|
31
|
Larson KR, Russo KA, Fang Y, Mohajerani N, Goodson ML, Ryan KK. Sex Differences in the Hormonal and Metabolic Response to Dietary Protein Dilution. Endocrinology 2017; 158:3477-3487. [PMID: 28938440 DOI: 10.1210/en.2017-00331] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 08/03/2017] [Indexed: 12/31/2022]
Abstract
Consumption of a low-protein, high-carbohydrate diet induces a striking increase in circulating fibroblast growth factor-21 (FGF21), which is associated with improved cardiometabolic health and increased longevity. Increased lifespan during this dietary protein "dilution" has been explained by resource-mediated trade-offs between reproduction and survival, such that fecundity is optimized at a greater relative intake of proteins/carbohydrates. The magnitude of this trade-off is thought to be sex-dependent. In this study, we tested the hypothesis that metabolic responses to dietary protein dilution are likewise dependent on sex. We maintained age-matched adult male and female C57BL/6J mice on isocaloric diets containing 22% fat and differing in the ratio of protein/carbohydrate. The normal protein (NP) control diet contained 18% protein and 60% carbohydrate by kcal. The protein diluted (PD) diet contained 4% protein and 74% carbohydrate. Consistent with previous reports, PD males gained less weight and less fat than did normal protein controls and exhibited both improved glucose tolerance and decreased plasma lipids. In contrast, these metabolic benefits were absent among age-matched females maintained on the same diets. Likewise, whereas circulating FGF21 was increased up to 66-fold among PD male mice, this was substantially blunted among female counterparts. Sex differences in energy balance, glucose control, and plasma FGF21 were reversed upon ovariectomy. Collectively, our findings support that female mice are relatively less sensitive to the metabolic improvements observed following dietary protein dilution. This is accompanied by blunted circulating levels of FGF21 and requires an intact female reproductive system.
Collapse
Affiliation(s)
- Karlton R Larson
- Department of Neurobiology, Physiology, and Behavior, College of Biological Sciences, University of California, Davis, Davis, California 95616
| | - Kimberly A Russo
- Department of Neurobiology, Physiology, and Behavior, College of Biological Sciences, University of California, Davis, Davis, California 95616
| | - Yanbin Fang
- Department of Neurobiology, Physiology, and Behavior, College of Biological Sciences, University of California, Davis, Davis, California 95616
| | - Niloufar Mohajerani
- Department of Neurobiology, Physiology, and Behavior, College of Biological Sciences, University of California, Davis, Davis, California 95616
| | - Michael L Goodson
- Department of Neurobiology, Physiology, and Behavior, College of Biological Sciences, University of California, Davis, Davis, California 95616
| | - Karen K Ryan
- Department of Neurobiology, Physiology, and Behavior, College of Biological Sciences, University of California, Davis, Davis, California 95616
| |
Collapse
|
32
|
Morselli E, Santos RS, Criollo A, Nelson MD, Palmer BF, Clegg DJ. The effects of oestrogens and their receptors on cardiometabolic health. Nat Rev Endocrinol 2017; 13:352-364. [PMID: 28304393 DOI: 10.1038/nrendo.2017.12] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cardiovascular disease (CVD) is one of the leading causes of mortality in developed countries. The incidence of CVD is sexually dimorphic, and research has focused on the contribution of sex steroids to the development and progression of the cardiometabolic syndrome, which is defined as a clustering of interrelated risk factors that promote the development of atherosclerosis (which can lead to CVD) and type 2 diabetes mellitus. Data are inconclusive as to how sex steroids and their respective receptors increase or suppress the risk of developing the cardiometabolic syndrome and thus CVD. In this Review, we discuss the potential role, or roles, of sex hormones in cardiometabolic health by first focusing on the influence of oestrogens and their receptors on the risk of developing cardiometabolic syndrome and CVD. We also highlight what is known about testosterone and its potential role in protecting against the development of the cardiometabolic syndrome and CVD. Given the inconclusive nature of the data regarding the direct effects of each sex hormone, we advocate and highlight the importance of studying the relative levels and the ratio of sex hormones to each other, as well as the use of cross sex hormone therapy and its effect on cardiometabolic health.
Collapse
Affiliation(s)
- Eugenia Morselli
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Roberta S Santos
- Obesity and Comorbidities Research Center, Institute of Biology, State University of Campinas, Campinas 1, 3083-864, Brazil
- Cedars-Sinai Diabetes and Obesity Research Institute, Department of Biomedical Research, Los Angeles, California 90048, USA
| | - Alfredo Criollo
- Advanced Center for Chronic Diseases (ACCDiS) and Center for Molecular Studies of the Cell (CEMC), Santiago 8380000, Chile
- Instituto de Investigación en Ciencias Odontológicas (ICOD), Facultad de Odontología, Universidad de Chile, Santiago 8380492, Chile
| | - Michael D Nelson
- Applied Physiology and Advanced Imaging Laboratory, Department of Kinesiology, University of Texas at Arlington, Texas 76019, USA
| | - Biff F Palmer
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Deborah J Clegg
- Cedars-Sinai Diabetes and Obesity Research Institute, Department of Biomedical Research, Los Angeles, California 90048, USA
| |
Collapse
|
33
|
López M, Tena-Sempere M. Estradiol effects on hypothalamic AMPK and BAT thermogenesis: A gateway for obesity treatment? Pharmacol Ther 2017; 178:109-122. [PMID: 28351720 DOI: 10.1016/j.pharmthera.2017.03.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/21/2017] [Indexed: 12/24/2022]
Abstract
In addition to their prominent roles in the control of reproduction, estrogens are important modulators of energy balance, as evident in conditions of deficiency of estrogens, which are characterized by increased feeding and decreased energy expenditure, leading to obesity. AMP-activated protein kinase (AMPK) is a ubiquitous cellular energy gauge that is activated under conditions of low energy, increasing energy production and reducing energy wasting. Centrally, the AMPK pathway is a canonical route regulating energy homeostasis, by integrating peripheral signals, such as hormones and metabolites, with neuronal networks. As a result of those actions, hypothalamic AMPK modulates feeding, as well as brown adipose tissue (BAT) thermogenesis and browning of white adipose tissue (WAT). Here, we will review the central actions of estrogens on energy balance, with particular focus on hypothalamic AMPK. The relevance of this interaction is noteworthy, because some agents with known actions on metabolic homeostasis, such as nicotine, metformin, liraglutide, olanzapine and also natural molecules, such as resveratrol and flavonoids, exert their actions by modulating AMPK. This evidence highlights the possibility that hypothalamic AMPK might be a potential target for the treatment of obesity.
Collapse
Affiliation(s)
- Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria (IDIS), 15782 Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos II, Spain.
| | - Manuel Tena-Sempere
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos II, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Spain; Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Reina Sofía, 14004 Córdoba, Spain; FiDiPro Program, Department of Physiology, University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland.
| |
Collapse
|