1
|
Ametepe S, Nyarko BYE, Wormekpor I, Kantah B, Appiah M, Kwadzokpui PK, Adejumo EN, Obirikorang C, Ibrahim A, Kwarteng BA, Nyarko EN, Osei-Yeboah J, Lokpo SY. Hypothyroidism phenotypes, clinical characteristics, and factors associated with nodular thyroid disease in patients with hypothyroidism in Southern Ghana: a 6-year retrospective study. BMC Endocr Disord 2025; 25:111. [PMID: 40264045 PMCID: PMC12013080 DOI: 10.1186/s12902-025-01934-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/14/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND This study aimed to describe the frequency of hypothyroidism phenotypes, clinical characteristics, and factors associated with nodular thyroid among hypothyroid patients at the University of Ghana Hospital in southern Ghana. METHODS This study was a 6-year hospital-based retrospective study that extracted data from 221 patients with hypothyroidism from the archival records of the University of Ghana Hospital using a checklist. These include socio-demography (age, gender, marital status, residential status, educational level), lifestyle variables (anthropometry, smoking status, alcohol intake), and co-morbidities, as well as ultrasound imaging findings of the thyroid. Serum thyroid hormone levels were used to classify hypothyroidism phenotypes. Bivariate and multivariate logistic regression analyses were performed to identify factors associated with nodular thyroid disease. RESULT The frequency of primary, subclinical, and secondary hypothyroidism was 81.4%, 16.3%, and 2.3%, respectively. Fatigue [120(54.3%)], heavy menstrual loss [54/160(33.8%)], and cold intolerance [73(33.0%)], were predominant symptoms while 54/57 (94.7%) were overweight/obese, 23/32 (71.9%) had dyslipidaemia whereas 7/34 (20.6%) had hypertension. The odds of developing nodular thyroid disease were 2.11 times higher (95% CI: 1.07-4.17; p = 0.032) in males than in females. CONCLUSION Our results provide insight into the frequency of hypothyroidism phenotypes, clinical characteristics, and factors associated with nodular thyroid, emphasizing male gender as an independent predictor of nodular thyroid disease. Our findings also emphasize the need for lifestyle adjustment as a mitigating strategy in the management of hypothyroidism. However, prospective studies are required to confirm the findings or investigate the histological characteristics of thyroid nodules in patients with hypothyroidism.
Collapse
Affiliation(s)
- Samuel Ametepe
- Department of Medical Laboratory Science, Faculty of Health and Allied Sciences, Koforidua Technical University, Koforidua, Ghana
| | - Brian Yaw Entsiey Nyarko
- Department of Medical Laboratory Sciences, School of Allied Health Sciences, University of Health and Allied Sciences, Ho, Ghana
| | - Isaac Wormekpor
- Department of Medical Laboratory Sciences, School of Allied Health Sciences, University of Health and Allied Sciences, Ho, Ghana
| | - Bright Kantah
- Department of Medical Laboratory Sciences, School of Allied Health Sciences, University of Health and Allied Sciences, Ho, Ghana
| | - Michael Appiah
- Department of Medical Laboratory Sciences, Greater Accra Region, Accra Technical University, Accra, Ghana
| | | | - Esther Ngozi Adejumo
- Department of Medical Laboratory Science, School of Public and Allied Health, Babcock University, Ilishan-Remo, Ogun State, Nigeria
| | - Christian Obirikorang
- Department of Molecular Medicine, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Abigail Ibrahim
- Nursing and Midwifery Training College, Ministry of Health, Odumasi-Krobo, Ghana
| | | | - Eric Ny Nyarko
- Department of Chemical Pathology, University of Ghana Medical School, Accra, Ghana
| | - James Osei-Yeboah
- School of Public Health, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Sylvester Yao Lokpo
- Department of Medical Laboratory Sciences, School of Allied Health Sciences, University of Health and Allied Sciences, Ho, Ghana.
| |
Collapse
|
2
|
Xie S, Zhao T, Hu C, Meng Y, Cui J, Wu X. Disruption of Ephb1 causes reduced hypothalamic CRH and TRH expression and obesity in mice. Obesity (Silver Spring) 2025. [PMID: 40207393 DOI: 10.1002/oby.24275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 01/23/2025] [Accepted: 02/04/2025] [Indexed: 04/11/2025]
Abstract
OBJECTIVE Ephrin type-B receptor 1 (EphB1) is a receptor tyrosine kinase involved in axon guidance, synaptic plasticity, and tumorigenesis. However, the role of EphB1 in metabolic regulation and obesity remains poorly understood. This study aims to uncover the role of EphB1 in energy metabolism and provide insights into the underlying mechanisms by which EphB1 regulates obesity. METHODS Two Ephb1 mutations identified from a forward genetic screen for obesity-related loci in mice were examined for their effects in gene expression, energy metabolism, and endocrine changes. The impacts of EphB1 on neuropeptide expression and signal transduction were evaluated in both hypothalamic tissues and primary cells. Potential downstream signals were modified in Ephb1 mutants to verify the interaction. RESULTS Ephb1 mutants develop obesity in adolescence and develop impaired glucose tolerance during adulthood. EphB1 deficiency caused lower body temperature, blunted cold-induced thermogenesis, and decreased locomotor activity, but it did not alter food intake. EphB1 promotes cyclic AMP-responsive element-binding protein (CREB) phosphorylation via phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling in a cell-autonomous manner. EphB1 deficiency leads to reduced expression of corticotropin-releasing hormone (CRH) and thyrotropin-releasing hormone (TRH) in the brain. Intraventricular administration of either TRH or a CRH fragment suppressed obesity in Ephb1 mutants. CONCLUSIONS EphB1 regulates hypothalamic CRH and TRH expression and promotes energy expenditure in mice.
Collapse
Affiliation(s)
- Simin Xie
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Tao Zhao
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Chengchen Hu
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Yongyong Meng
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jing Cui
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, and Clinical Genomics Program, The National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Xiaohui Wu
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Motzfeldt Jensen M, Almasi C, Florian Sørensen HC, Andersen SL, Andersen S. Thyroid response to blocking sympathetic activity in chronic cold exposed hunters in East Greenland: a case-control study. Eur Thyroid J 2024; 14:ETJ-24-0272. [PMID: 39656551 DOI: 10.1530/etj-24-0272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/10/2024] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Thyroid hormones and sympathetic stimulation are needed for activating Brown adipose tissue (BAT) during cold exposure. Studies of human cold exposure have demonstrated both increased production and raised clearance of triiodothyronine (T3). Greenlandic hunters provide a unique model for evaluating metabolic effects of cold exposure. AIM We aimed to explore the dynamics of thyroid hormones when blocking sympathetic activity in Greenlandic hunters during winter to inspire knowledge on mechanisms of BAT activation. METHODS We conducted a 7-day field study of Greenlandic hunters (n=7) in East Greenland in February. The sympathetic system was blocked using a non-selective beta blocker for seven consecutive days. A group of non-hunter Greenlanders (n = 8) from the same settlement were included for parallel sampling. All participants were healthy men. Blood samples were drawn daily for measurement of TSH, thyroid hormone levels, and thyroglobulin. RESULTS Hunters had higher serum thyroglobulin, TSH, and high fT3/fT4 ratio compared to controls. Blocking the sympathetic activity was followed by changes in serum thyroglobulin and fT3 with an initial decrease and subsequent restoration of levels, while TSH and fT4 showed a gradual increase over the course of the study. The fT3/fT4 ratio showed a continuous and marked decrease. CONCLUSION We hypothesise that when blocking the sympathetic system, TSH increases to uphold the production of T3 needed for maintaining BAT activity. Additionally, alterations of fT3/fT4-ratio support a hypothesis of adrenergic stimulation promoting T3 over T4 secretion from the thyroid via the adrenergic nerve terminals in the thyroid.
Collapse
Affiliation(s)
- Mette Motzfeldt Jensen
- M Motzfeldt Jensen, Department of Clinical Medicine, Aalborg University Hospital, Aalborg, Denmark
| | - Charlotte Almasi
- C Almasi, Department of Nuclear Medicine, Aalborg University Hospital, Aalborg, Denmark
| | | | - Stine Linding Andersen
- S Andersen, Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Stig Andersen
- S Andersen, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
4
|
Modder M, Coomans CP, Saaltink DJ, Tersteeg MMH, Hoogduin J, Scholten L, Pronk ACM, Lalai RA, Boelen A, Kalsbeek A, Rensen PCN, Vreugdenhil E, Kooijman S. Doublecortin-like knockdown in mice attenuates obesity by stimulating energy expenditure in adipose tissue. Sci Rep 2024; 14:19517. [PMID: 39174821 PMCID: PMC11341836 DOI: 10.1038/s41598-024-70639-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/20/2024] [Indexed: 08/24/2024] Open
Abstract
Crosstalk between peripheral metabolic organs and the central nervous system is essential for body weight control. At the base of the hypothalamus, β-tanycytes surround the portal capillaries and function as gatekeepers to facilitate transfer of substances from the circulation into the cerebrospinal fluid and vice versa. Here, we investigated the role of the neuroplasticity gene doublecortin-like (DCL), highly expressed by β-tanycytes, in body weight control and whole-body energy metabolism. We demonstrated that DCL-knockdown through a doxycycline-inducible shRNA expression system prevents body weight gain by reducing adiposity in mice. DCL-knockdown slightly increased whole-body energy expenditure possibly as a result of elevated circulating thyroid hormones. In white adipose tissue (WAT) triglyceride uptake was increased while the average adipocyte cell size was reduced. At histological level we observed clear signs of browning, and thus increased thermogenesis in WAT. We found no indications for stimulated thermogenesis in brown adipose tissue (BAT). Altogether, we demonstrate an important, though subtle, role of tanycytic DCL in body weight control through regulation of energy expenditure, and specifically WAT browning. Elucidating mechanisms underlying the role of DCL in regulating brain-peripheral crosstalk further might identify new treatment targets for obesity.
Collapse
Affiliation(s)
- Melanie Modder
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Claudia P Coomans
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Dirk-Jan Saaltink
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Mayke M H Tersteeg
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Janna Hoogduin
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Leonie Scholten
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Amanda C M Pronk
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Reshma A Lalai
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Anita Boelen
- Endocrine Laboratory, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Endocrine Laboratory, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Erno Vreugdenhil
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands.
- Leiden University Medical Center, Albinusdreef 2, 2333ZA, Leiden, The Netherlands.
| |
Collapse
|
5
|
Costa-E-Sousa RH, Brooks VL. The growing complexity of the control of the hypothalamic pituitary thyroid axis and brown adipose tissue by leptin. VITAMINS AND HORMONES 2024; 127:305-362. [PMID: 39864945 DOI: 10.1016/bs.vh.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
The balance between food intake and energy expenditure is precisely regulated to maintain adipose stores. Leptin, which is produced in and released from adipose in direct proportion to its size, is a major contributor to this control and initiates its homeostatic responses largely via binding to leptin receptors (LepR) in the hypothalamus. Decreases in hypothalamic LepR binding signals starvation, leading to hunger and reduced energy expenditure, whereas increases in hypothalamic LepR binding can suppress food intake and increase energy expenditure. However, large gaps persist in the specific hypothalamic sites and detailed mechanisms by which leptin increases energy expenditure, via the parallel activation of the hypothalamic pituitary thyroid (HPT) axis and brown adipose tissue (BAT). The purpose of this review is to develop a framework for the complex mechanisms and neurocircuitry. The core circuitry begins with leptin binding to receptors in the arcuate nucleus, which then sends projections to the paraventricular nucleus (to regulate the HPT axis) and the dorsomedial hypothalamus (to regulate BAT). We build on this core by layering complexities, including the intricate and unsettled regulation of arcuate proopiomelanocortin neurons by leptin and the changes that occur as the regulation of the HPT axis and BAT is engaged or modified by challenges such as starvation, hypothermia, obesity, and pregnancy.
Collapse
Affiliation(s)
- Ricardo H Costa-E-Sousa
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Virginia L Brooks
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, United States.
| |
Collapse
|
6
|
Lutterschmidt DI, Stratton K, Winters TJ, Martin S, Merlino LJ. Neural thyroid hormone metabolism integrates seasonal changes in environmental temperature with the neuroendocrine reproductive axis. Horm Behav 2024; 161:105517. [PMID: 38422864 DOI: 10.1016/j.yhbeh.2024.105517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 03/02/2024]
Abstract
We asked if environmental temperature alters thyroid hormone metabolism within the hypothalamus, thereby providing a neuroendocrine mechanism by which temperature could be integrated with photoperiod to regulate seasonal rhythms. We used immunohistochemistry to assess the effects of low-temperature winter dormancy at 4 °C or 12 °C on thyroid-stimulating hormone (TSH) within the infundibulum of the pituitary as well as deiodinase 2 (Dio2) and 3 (Dio3) within the hypothalamus of red-sided garter snakes (Thamnophis sirtalis). Both the duration and, in males, magnitude of low-temperature dormancy altered deiodinase immunoreactivity within the hypothalamus, increasing the area of Dio2-immunoreactivity in males and females and decreasing the number of Dio3-immunoreactive cells in males after 8-16 weeks. Reciprocal changes in Dio2/3 favor the accumulation of triiodothyronine within the hypothalamus. Whether TSH mediates these effects requires further study, as significant changes in TSH-immunoreactive cell number were not observed. Temporal changes in deiodinase immunoreactivity coincided with an increase in the proportion of males exhibiting courtship behavior as well as changes in the temporal pattern of courtship behavior after emergence. Our findings mirror those of previous studies, in which males require low-temperature exposure for at least 8 weeks before significant changes in gonadotropin-releasing hormone immunoreactivity and sex steroid hormones are observed. Collectively, these data provide evidence that the neuroendocrine pathway regulating the reproductive axis via thyroid hormone metabolism is capable of transducing temperature information. Because all vertebrates can potentially use temperature as a supplementary cue, these results are broadly applicable to understanding how environment-organism interactions mediate seasonally adaptive responses.
Collapse
Affiliation(s)
| | - Kalera Stratton
- Department of Biology, Portland State University, OR, United States
| | - Treven J Winters
- Department of Biology, Portland State University, OR, United States
| | - Stephanie Martin
- Department of Biology, Portland State University, OR, United States
| | - Lauren J Merlino
- Department of Biology, Portland State University, OR, United States
| |
Collapse
|
7
|
Zhang J, You S, Yu L, Zhang Y, Li Z, Zhao N, Zhang B, Kang L, Sun S. The hysteresis damage of cold exposure on tissue and transcript levels in mice. J Therm Biol 2024; 120:103823. [PMID: 38442663 DOI: 10.1016/j.jtherbio.2024.103823] [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: 06/30/2022] [Revised: 02/18/2024] [Accepted: 02/20/2024] [Indexed: 03/07/2024]
Abstract
OBJECTIVES Although cold stress-induced damage to the heart and thyroid has been reported, specific organ associations between the heart and thyroid with delayed injury mechanisms have not been investigated. In this study, we determined the damage time and transcript levels of a large number of genes in the heart and thyroid after cold exposure. Meanwhile, we analysed the relationship between heart and thyroid injury in human medical records to determine the association of delayed injury from cold exposure. METHODS Mice were exposed to cold stress and hysteresis injury. Gene changes at the transcriptional level were detected using high throughput sequencing technology. The most variable genes were verified at the protein level using Western Blotting and medical records were collected and analysed. RESULTS The damage was the most severe when the animals were allowed to recover to room temperature for 4 h after exposure to cold stress. During this process, STAT1 and ATF3 genes were acutely up-regulated. Analysis of human medical records showed the highest correlation between AST and T4 under cold stress (p = 0.0011). CONCLUSIONS Exposure to cold increases blood level of free thyroid hormone and biomarkers of myocardial injury, as well as related mRNA levels. These changes were more pronounced after return to room temperature.
Collapse
Affiliation(s)
- Jing Zhang
- Shihezi University College of Chemistry and Chemical Engineering, College of Pharmacy / Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi, 832002, Xinjiang, China; Xinjiang University of Science&Technology, School of Medicine, Korla, 841000, China
| | - Shiwan You
- Shihezi University College of Chemistry and Chemical Engineering, College of Pharmacy / Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi, 832002, Xinjiang, China
| | - Lan Yu
- Shanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest Agriculture and Forestry University, Yangling, Shaanxi, 712100, China
| | - Yuling Zhang
- Shihezi University College of Chemistry and Chemical Engineering, College of Pharmacy / Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi, 832002, Xinjiang, China
| | - Zuoping Li
- Shihezi University College of Chemistry and Chemical Engineering, College of Pharmacy / Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi, 832002, Xinjiang, China
| | - Na Zhao
- Shihezi University College of Chemistry and Chemical Engineering, College of Pharmacy / Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi, 832002, Xinjiang, China
| | - Bo Zhang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China.
| | - Lihua Kang
- Shihezi University College of Chemistry and Chemical Engineering, College of Pharmacy / Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi, 832002, Xinjiang, China.
| | - Shiguo Sun
- Shihezi University College of Chemistry and Chemical Engineering, College of Pharmacy / Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi, 832002, Xinjiang, China; Shanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest Agriculture and Forestry University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
8
|
Barany A, Fuentes J, Valderrama V, Broz-Ruiz A, Martínez-Rodríguez G, Mancera JM. Oral cortisol and dexamethasone intake: Differential physiology and transcriptional responses in the marine juvenile Sparus aurata. Gen Comp Endocrinol 2023; 344:114371. [PMID: 37640145 DOI: 10.1016/j.ygcen.2023.114371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 07/12/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
Abstract
This study approached the long-term oral administration of cortisol (F) and dexamethasone (DEX), two synthetic glucocorticoids, compared to a control group (CT) in the juveniles of a marine teleost, the gilthead seabream (Sparus aurata). Physiologically, DEX treatment impaired growth, which appears to be linked to carbohydrate allocation in muscle and liver, hepatic triglycerides depletion, and reduced hematocrit. Hypophyseal gh mRNA expression was 2-fold higher in DEX than in CT or F groups. Similarly, hypothalamic trh and hypophyseal pomcb followed this pattern. Plasma cortisol levels were significantly lower in DEX than in CT, while F presented intermediate levels. In the posterior intestine, measured short circuit-current (Isc) was more anion absorptive in CT and F compared to the DEX group, whereas Isc remained unaffected in the anterior intestine. The derived transepithelial electric resistance (TEER) significantly differed between intestinal regions in the DEX group. These results provide new insights to understand better potential targeted biomarkers indicative of the differential glucocorticoid or mineralocorticoid-receptors activation in fish.
Collapse
Affiliation(s)
- A Barany
- Department of Biology, Morrill Science Center, University of Massachusetts, 01003 Amherst, MA, USA; Departamento de Biología, Facultad de Ciencias del Mar y Ambientales, Instituto Universitario de Investigación Marina (INMAR), Campus de Excelencia Internacional del Mar (CEI·MAR), Universidad de Cádiz, E-11510 Puerto Real, Cádiz, Spain.
| | - J Fuentes
- Centro de Ciências do Mar (CCMAR), Universidade do Algarve, 8005-139 Faro, Portugal
| | - V Valderrama
- Departamento de Biología, Facultad de Ciencias del Mar y Ambientales, Instituto Universitario de Investigación Marina (INMAR), Campus de Excelencia Internacional del Mar (CEI·MAR), Universidad de Cádiz, E-11510 Puerto Real, Cádiz, Spain
| | - A Broz-Ruiz
- Departamento de Biología, Facultad de Ciencias del Mar y Ambientales, Instituto Universitario de Investigación Marina (INMAR), Campus de Excelencia Internacional del Mar (CEI·MAR), Universidad de Cádiz, E-11510 Puerto Real, Cádiz, Spain
| | - G Martínez-Rodríguez
- Instituto de Ciencias Marinas de Andalucía, Spanish National Research Council (ICMAN-CSIC), E-11510 Puerto Real, Cádiz, Spain
| | - J M Mancera
- Departamento de Biología, Facultad de Ciencias del Mar y Ambientales, Instituto Universitario de Investigación Marina (INMAR), Campus de Excelencia Internacional del Mar (CEI·MAR), Universidad de Cádiz, E-11510 Puerto Real, Cádiz, Spain
| |
Collapse
|
9
|
Singh VK, Singh S, Nandhini PB, Bhatia AK, Dixit SP, Ganguly I. Comparative genomic diversity analysis of copy number variations (CNV) in indicine and taurine cattle thriving in Europe and Indian subcontinent. Anim Biotechnol 2023; 34:3483-3494. [PMID: 36592947 DOI: 10.1080/10495398.2022.2162910] [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: 01/04/2023]
Abstract
Copy number variations (CNVs) include deletions, duplications, and insertions that are larger than 50 bp in size causing structural variation responsible for diversity, adaptation, and breed development. Indian cattle breeds are highly diverse from the taurine breeds. The pattern of CNVRs in 191 animals belonging to 39 cattle breeds (four Indicine and 35 Taurine) was studied based on Illumina 777K BovineHD chip data. The Indicine breeds revealed 2590 CNVs and 335 copy number variation regions (CNVRs) in autosomes. Out of the identified CNVs, 50 were found to be novel. Structure analysis revealed admixed nature of Siri. Neighbor joining tree from CNVR data showed that hot (Kankrej and Hallikar) and cold (Ladakhi and Siri) adapted cattle breeds clustered separately. CNVR of Indian and European breeds revealed that Balkan and Italian breeds of Podolian group are admixed with Indian cattle breeds corroborating indicine introgression (6.1-13.5%). CNVRs spanning the regions of olfactory receptors and immune system genes were identified. AMOVA revealed 9% variation among populations which is 2% greater than SNP based studies showing higher inclusion of variation by CNVR. Detailed analysis of CNVs/CNVRs in Indian cattle adapted to hot and cold climate, and their diversity among worldwide cattle is presented in this study.
Collapse
Affiliation(s)
- V K Singh
- Animal Genetics and Breeding Division, ICAR-National Dairy Research Institute, Karnal, India
| | - S Singh
- Animal Genetics Division, ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| | - P B Nandhini
- Animal Genetics and Breeding Division, ICAR-National Dairy Research Institute, Karnal, India
| | - A K Bhatia
- Animal Genetic Resources Division, ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| | - S P Dixit
- Animal Genetics Division, ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| | - I Ganguly
- Animal Genetics Division, ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| |
Collapse
|
10
|
Yoshida T, Fujitani M, Farmer S, Harada A, Shi Z, Lee JJ, Tinajero A, Singha AK, Fujikawa T. VMHdm/c SF-1 neuronal circuits regulate skeletal muscle PGC1-α via the sympathoadrenal drive. Mol Metab 2023; 77:101792. [PMID: 37633515 PMCID: PMC10491730 DOI: 10.1016/j.molmet.2023.101792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 08/28/2023] Open
Abstract
OBJECTIVE To adapt to metabolically challenging environments, the central nervous system (CNS) orchestrates metabolism of peripheral organs including skeletal muscle. The organ-communication between the CNS and skeletal muscle has been investigated, yet our understanding of the neuronal pathway from the CNS to skeletal muscle is still limited. Neurons in the dorsomedial and central parts of the ventromedial hypothalamic nucleus (VMHdm/c) expressing steroidogenic factor-1 (VMHdm/cSF-1 neurons) are key for metabolic adaptations to exercise, including increased basal metabolic rate and skeletal muscle mass in mice. However, the mechanisms by which VMHdm/cSF-1 neurons regulate skeletal muscle function remain unclear. Here, we show that VMHdm/cSF-1 neurons increase the sympathoadrenal activity and regulate skeletal muscle peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) in mice via multiple downstream nodes. METHODS Optogenetics was used to specifically manipulate VMHdm/cSF-1 neurons combined with genetically-engineered mice and surgical manipulation of the sympathoadrenal activity. RESULTS Optogenetic activation of VMHdm/cSF-1 neurons dramatically elevates mRNA levels of skeletal muscle Pgc-1α, which regulates a spectrum of skeletal muscle function including protein synthesis and metabolism. Mechanistically, the sympathoadrenal drive coupled with β2 adrenergic receptor (β2AdR) is essential for VMHdm/cSF-1 neurons-mediated increases in skeletal muscle PGC1-α. Specifically, both adrenalectomy and β2AdR knockout block augmented skeletal muscle PGC1-α by VMHdm/cSF-1 neuronal activation. Optogenetic functional mapping reveals that downstream nodes of VMHdm/cSF-1 neurons are functionally redundant to increase circulating epinephrine and skeletal muscle PGC1-α. CONCLUSIONS Collectively, we propose that VMHdm/cSF-1 neurons-skeletal muscle pathway, VMHdm/cSF-1 neurons→multiple downstream nodes→the adrenal gland→skeletal muscle β2AdR, underlies augmented skeletal muscle function for metabolic adaptations.
Collapse
Affiliation(s)
- Takuya Yoshida
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, USA; Department of Clinical Nutrition School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
| | - Mina Fujitani
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, USA; Laboratory of Nutrition Science, Department of Bioscience, Graduate School of Agriculture, Ehime University, Matsuyama, Japan
| | - Scotlynn Farmer
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, USA
| | - Ami Harada
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, USA; Nara Medical University, Nara, Japan
| | - Zhen Shi
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, USA; Department of Plastic Surgery, Hospital Zhejiang University School of Medicine, Zhejiang, China
| | - Jenny J Lee
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, USA
| | - Arely Tinajero
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, USA
| | - Ashish K Singha
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, USA
| | - Teppei Fujikawa
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, USA; Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, USA.
| |
Collapse
|
11
|
Peng YY, Lu XM, Li S, Tang C, Ding Y, Wang HY, Yang C, Wang YT. Effects and mechanisms of extremely cold environment on body response after trauma. J Therm Biol 2023; 114:103570. [PMID: 37344028 DOI: 10.1016/j.jtherbio.2023.103570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 04/07/2023] [Accepted: 04/07/2023] [Indexed: 06/23/2023]
Abstract
With the outbreak of the Ukrainian crisis, extremely cold environment warfare has once again become the focus of international attention. People exposed to extremely cold environments may suffer from cold damage, further aggravate trauma, trigger high disability and mortality rates, and even cause serious sequelae. To declare the effects and mechanisms of the extremely cold environment on the body after trauma, this paper reviews, firstly, physiological reaction of human body in an extremely cold environment. Then, the post-traumatic body response in an extremely cold environment was introduced, and finally, the sequelae of trauma in extremely cold environment was further summarized in the paper. The results indicated that extremely cold environment can cause a series of damage to the body, especially the body after trauma. The extremely cold factor is a double-edged sword, showing a favorable and unfavorable side in different aspects. Moreover, in addition to the trauma suffered by the body, the subsequent sequelae such as cognitive dysfunction, anxiety, depression and even post-traumatic stress disorder may also be induced. The paper summarizes the human body's physiological response in an extremely cold environment, and declares the effects and mechanisms of the extremely cold environment on the body after trauma, which may provide a theoretical basis for effectively improving the level of combat trauma treatment in extremely cold regions.
Collapse
Affiliation(s)
- Yu-Yuan Peng
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China; College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Xiu-Min Lu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Sen Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Can Tang
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Yang Ding
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Hai-Yan Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Ce Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yong-Tang Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
12
|
Bahar MR, Tekin S, Beytur A, Onalan EE, Ozyalin F, Colak C, Sandal S. Effects of intracerebroventricular MOTS-c infusion on thyroid hormones and uncoupling proteins. Biol Futur 2023:10.1007/s42977-023-00163-6. [PMID: 37067760 DOI: 10.1007/s42977-023-00163-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/06/2023] [Indexed: 04/18/2023]
Abstract
This study was conducted to determine the possible effects of intracerebroventricular MOTS-c infusion on thyroid hormones and uncoupling proteins (UCPs) in rats. Forty male Wistar Albino rats were divided into 4 groups with 10 animals in each group: control, sham, 10 and 100 µM MOTS-c. Hypothalamus, blood, muscle, adipose tissues samples were collected for thyrotropin-releasing hormone (TRH), UCP1 and UCP3 levels were determined by the RT-PCR and western blot analysis. Serum thyroid hormone levels were determined by the ELISA assays. MOTS-c infusion was found to increase food consumption but it did not cause any changes in the body weight. MOTS-c decreased serum TSH, T3, and T4 hormone levels. On the other hand, it was also found that MOTS-c administration increased UCP1 and UCP3 levels in peripheral tissues. The findings obtained in the study show that central MOTS-c infusion is a directly effective agent in energy metabolism.
Collapse
Affiliation(s)
- Mehmet Refik Bahar
- Department of Physiology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Suat Tekin
- Department of Physiology, Faculty of Medicine, Inonu University, Malatya, Turkey.
| | - Asiye Beytur
- Department of Physiology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Ebru Etem Onalan
- Department of Medical Biology, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Fatma Ozyalin
- Laboratory and Veterinary Health Program, Akcadag Vocational School, Malatya Turgut Ozal University, Malatya, Turkey
| | - Cemil Colak
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Süleyman Sandal
- Department of Physiology, Faculty of Medicine, Inonu University, Malatya, Turkey
| |
Collapse
|
13
|
Xue S, Lee D, Berry DC. Thermogenic adipose tissue in energy regulation and metabolic health. Front Endocrinol (Lausanne) 2023; 14:1150059. [PMID: 37020585 PMCID: PMC10067564 DOI: 10.3389/fendo.2023.1150059] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/07/2023] [Indexed: 04/07/2023] Open
Abstract
The ability to generate thermogenic fat could be a targeted therapy to thwart obesity and improve metabolic health. Brown and beige adipocytes are two types of thermogenic fat cells that regulate energy balance. Both adipocytes share common morphological, biochemical, and thermogenic properties. Yet, recent evidence suggests unique features exist between brown and beige adipocytes, such as their cellular origin and thermogenic regulatory processes. Beige adipocytes also appear highly plastic, responding to environmental stimuli and interconverting between beige and white adipocyte states. Additionally, beige adipocytes appear to be metabolically heterogenic and have substrate specificity. Nevertheless, obese and aged individuals cannot develop beige adipocytes in response to thermogenic fat-inducers, creating a key clinical hurdle to their therapeutic promise. Thus, elucidating the underlying developmental, molecular, and functional mechanisms that govern thermogenic fat cells will improve our understanding of systemic energy regulation and strive for new targeted therapies to generate thermogenic fat. This review will examine the recent advances in thermogenic fat biogenesis, molecular regulation, and the potential mechanisms for their failure.
Collapse
Affiliation(s)
| | | | - Daniel C. Berry
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| |
Collapse
|
14
|
Zhao X, Zhang J, Wang H, Li H, Qu C, Wen J, Zhang X, Zhu T, Nie C, Li X, Muhatai G, Wang L, Lv X, Yang W, Zhao C, Bao H, Li J, Zhu B, Cao G, Xiong W, Ning Z, Qu L. Genomic and transcriptomic analyses reveal genetic adaptation to cold conditions in the chickens. Genomics 2022; 114:110485. [PMID: 36126832 DOI: 10.1016/j.ygeno.2022.110485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/27/2022] [Accepted: 09/16/2022] [Indexed: 01/14/2023]
Abstract
Under the pressure of natural and artificial selection, domestic animals, including chickens, have evolved unique mechanisms of genetic adaptations such as high-altitude adaptation, hot and arid climate adaptation, and desert adaptation. Here, we investigated the genetic basis of cold tolerance in chicken by integrating whole-genome and transcriptome sequencing technologies. Genome-wide comparative analyses of 118 chickens living in different latitudes showed 46 genes and several pathways that may be involved in cold adaptation. The results of the functional enrichment analysis of differentially expressed genes proved the important role of metabolic pathways and immune-related pathways in cold tolerance in chickens. The subsequent integration of whole genome and transcriptome sequencing technology further identified six genes - dnah5 (dynein axonemal heavy chain 5), ptgs2 (prostaglandin-endoperoxide synthase 2), inhba (inhibin beta A subunit), irx2 (iroquois homeobox 2), ensgalg00000054917, and ensgalg00000046652 - requiring more detailed studies. In addition, we also discovered different allele frequency distributions of five SNPs (single nucleotide polymorphisms) within ptgs2 and nine SNPs within dnah5 in chickens in different latitudes, suggesting strong selective pressure of these two genes in chickens. We provide a novel insight into the genetic adaptation in chickens to cold environments, and provide a reference for evaluating and developing adaptive chicken breeds in cold environments.
Collapse
Affiliation(s)
- Xiurong Zhao
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Jinxin Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Huie Wang
- Xinjiang Production & Construction Corps Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, Alar, Xinjiang 843300, China.
| | - Haiying Li
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830000, China.
| | - Changqing Qu
- Engineering Technology Research Center of Anti-aging Chinese Herbal Medicine of Anhui Province, Fuyang Normal University, Fuyang, Anhui 236037, China.
| | - Junhui Wen
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Xinye Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Tao Zhu
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Changsheng Nie
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Xinghua Li
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Gemingguli Muhatai
- Xinjiang Production & Construction Corps Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, Alar, Xinjiang 843300, China.
| | - Liang Wang
- Beijing Municipal General Station of Animal Science, Beijing 100107, China.
| | - XueZe Lv
- Beijing Municipal General Station of Animal Science, Beijing 100107, China.
| | - Weifang Yang
- Beijing Municipal General Station of Animal Science, Beijing 100107, China.
| | - Chunjiang Zhao
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Haigang Bao
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Junying Li
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Bo Zhu
- Animal Health Supervision Institute of Zhuozhou, Hebei Province 072750, China.
| | - Guomin Cao
- Animal husbandry station of Fangchenggang, Guangxi Province 538001, China.
| | - Wenjie Xiong
- Animal Disease Prevention and Control Center of Fangchenggang, Guangxi Province 538001, China.
| | - Zhonghua Ning
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Lujiang Qu
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
15
|
Heim M, Nixon IJ, Emmerson E, Callanan A. From hormone replacement therapy to regenerative scaffolds: A review of current and novel primary hypothyroidism therapeutics. Front Endocrinol (Lausanne) 2022; 13:997288. [PMID: 36277721 PMCID: PMC9581390 DOI: 10.3389/fendo.2022.997288] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Primary hypothyroidism severely impacts the quality of life of patients through a decrease in the production of the thyroid hormones T3 and T4, leading to symptoms affecting cardiovascular, neurological, cognitive, and metabolic function. The incidence rate of primary hypothyroidism is expected to increase in the near future, partially due to increasing survival of patients that have undergone radiotherapy for head and neck cancer, which induces this disease in over half of those treated. The current standard of care encompasses thyroid hormone replacement therapy, traditionally in the form of synthetic T4. However, there is mounting evidence that this is unable to restore thyroid hormone signaling in all tissues due to often persistent symptoms. Additional complications are also present in the form of dosage difficulties, extensive drug interactions and poor patience compliance. The alternative therapeutic approach employed in the past is combination therapy, which consists of administration of both T3 and T4, either synthetic or in the form of desiccated thyroid extract. Here, issues are present regarding the lack of regulation concerning formulation and lack of data regarding safety and efficacy of these treatment methods. Tissue engineering and regenerative medicine have been applied in conjunction with each other to restore function of various tissues. Recently, these techniques have been adapted for thyroid tissue, primarily through the fabrication of regenerative scaffolds. Those currently under investigation are composed of either biopolymers or native decellularized extracellular matrix (dECM) in conjunction with either primary thyrocytes or stem cells which have undergone directed thyroid differentiation. Multiple of these scaffolds have successfully restored an athyroid phenotype in vivo. However, further work is needed until clinical translation can be achieved. This is proposed in the form of exploration and combination of materials used to fabricate these scaffolds, the addition of peptides which can aid restoration of tissue homeostasis and additional in vivo experimentation providing data on safety and efficacy of these implants.
Collapse
Affiliation(s)
- Maria Heim
- Institute for Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
| | - Ian J. Nixon
- Department of ENT, Head and Neck Surgery, NHS Lothian, Edinburgh, United Kingdom
| | - Elaine Emmerson
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
| | - Anthony Callanan
- Institute for Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
16
|
Grassi D, Marraudino M, Garcia-Segura LM, Panzica GC. The hypothalamic paraventricular nucleus as a central hub for the estrogenic modulation of neuroendocrine function and behavior. Front Neuroendocrinol 2022; 65:100974. [PMID: 34995643 DOI: 10.1016/j.yfrne.2021.100974] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/17/2022]
Abstract
Estradiol and hypothalamic paraventricular nucleus (PVN) help coordinate reproduction with body physiology, growth and metabolism. PVN integrates hormonal and neural signals originating in the periphery, generating an output mediated both by its long-distance neuronal projections, and by a variety of neurohormones produced by its magnocellular and parvocellular neurosecretory cells. Here we review the cyto-and chemo-architecture, the connectivity and function of PVN and the sex-specific regulation exerted by estradiol on PVN neurons and on the expression of neurotransmitters, neuromodulators, neuropeptides and neurohormones in PVN. Classical and non-classical estrogen receptors (ERs) are expressed in neuronal afferents to PVN and in specific PVN interneurons, projecting neurons, neurosecretory neurons and glial cells that are involved in the input-output integration and coordination of neurohormonal signals. Indeed, PVN ERs are known to modulate body homeostatic processes such as autonomic functions, stress response, reproduction, and metabolic control. Finally, the functional implications of the estrogenic modulation of the PVN for body homeostasis are discussed.
Collapse
Affiliation(s)
- D Grassi
- Department of Anatomy, Histology and Neuroscience, Universidad Autonoma de Madrid, Madrid, Spain
| | - M Marraudino
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy
| | - L M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - G C Panzica
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy; Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy.
| |
Collapse
|
17
|
Watts AG, Kanoski SE, Sanchez-Watts G, Langhans W. The physiological control of eating: signals, neurons, and networks. Physiol Rev 2022; 102:689-813. [PMID: 34486393 PMCID: PMC8759974 DOI: 10.1152/physrev.00028.2020] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Graciela Sanchez-Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Eidgenössische Technische Hochschule-Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
18
|
Tran LT, Park S, Kim SK, Lee JS, Kim KW, Kwon O. Hypothalamic control of energy expenditure and thermogenesis. Exp Mol Med 2022; 54:358-369. [PMID: 35301430 PMCID: PMC9076616 DOI: 10.1038/s12276-022-00741-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 12/05/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
Energy expenditure and energy intake need to be balanced to maintain proper energy homeostasis. Energy homeostasis is tightly regulated by the central nervous system, and the hypothalamus is the primary center for the regulation of energy balance. The hypothalamus exerts its effect through both humoral and neuronal mechanisms, and each hypothalamic area has a distinct role in the regulation of energy expenditure. Recent studies have advanced the understanding of the molecular regulation of energy expenditure and thermogenesis in the hypothalamus with targeted manipulation techniques of the mouse genome and neuronal function. In this review, we elucidate recent progress in understanding the mechanism of how the hypothalamus affects basal metabolism, modulates physical activity, and adapts to environmental temperature and food intake changes. The hypothalamus is a key regulator of metabolism, controlling resting metabolism, activity levels, and responses to external temperature and food intake. The balance between energy intake and expenditure must be tightly controlled, with imbalances resulting in metabolic disorders such as obesity or diabetes. Obin Kwon at Seoul National University College of Medicine and Ki Woo Kim at Yonsei University College of Dentistry, Seoul, both in South Korea, and coworkers reviewed how metabolism is regulated by the hypothalamus, a small hormone-producing brain region. They report that hormonal and neuronal signals from the hypothalamus influence the ratio of lean to fatty tissue, gender-based differences in metabolism, activity levels, and weight gain in response to food intake. They note that further studies to untangle cause-and-effect relationships and other genetic factors will improve our understanding of metabolic regulation.
Collapse
Affiliation(s)
- Le Trung Tran
- Departments of Oral Biology and Applied Biological Science, BK21 Four, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Sohee Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.,Departments of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Seul Ki Kim
- Departments of Oral Biology and Applied Biological Science, BK21 Four, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Jin Sun Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.,Departments of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Ki Woo Kim
- Departments of Oral Biology and Applied Biological Science, BK21 Four, Yonsei University College of Dentistry, Seoul, 03722, Korea.
| | - Obin Kwon
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea. .,Departments of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 03080, Korea.
| |
Collapse
|
19
|
Effects of meteorin-like hormone on endocrine function of hypothalamo-hypophysial system and peripheral uncoupling proteins in rats. Mol Biol Rep 2022; 49:5919-5925. [PMID: 35332411 DOI: 10.1007/s11033-022-07374-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/15/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Meteorin-like hormone (Metrnl) is a peptide secreted from the adipose tissue and modulates the whole-body energy metabolism. Metrnl release into the circulation is influenced by obesity, cold exposure, and exercise. Thyroid hormones also exert many of their effects on metabolism through uncoupling proteins (UCPs). This study aimed to determine effect of Metrnl on hypothalamo-hypophysier-thyroid axis and energy metabolism and reveal the possible involvement of UCPs in this process. METHODS AND RESULTS Fourty male Sprague-Dawley rats were divided into 4 groups with 10 animals in each group: control, sham, 10 and 100 nM Metrnl. Hypothalamus, muscle, white adipose tissue (WAT) and brown adipose tissue (BAT) samples were collected to detect thyrotropin-releasing hormone (TRH), and UCP1 and UCP3 protein levels by western blot analysis. Serum thyroid-stimulating hormone (TSH), triiodothyronine (T3) and thyroxine (T4) hormone levels were determined by enzyme-linked immunosorbent assay. Central infusion of Metrnl caused significant increase in serum TSH, T3 and T4 levels compared to control (p < 0.05). After Metrnl treatment, there were significant increases in TRH in hypothalamus tissue, UCP1 in WAT and BAT; and UCP3 protein in the muscle tissue (p < 0.05). CONCLUSIONS The findings that Metrnl induced increases in the peripheral UCPs and hypothalamus-pituitary-thyroid axis hormones implicate a role for this hormone in body energy homeostasis through UCP-mediated mechanisms.
Collapse
|
20
|
Role of TRPM8 in cold avoidance behaviors and brain activation during innocuous and nocuous cold stimuli. Physiol Behav 2022; 248:113729. [DOI: 10.1016/j.physbeh.2022.113729] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/03/2022] [Accepted: 02/03/2022] [Indexed: 11/22/2022]
|
21
|
Nabi G, Xing D, Sun Y, Zhang Q, Li M, Jiang C, Ahmad IM, Wingfield JC, Wu Y, Li D. Coping with extremes: High-altitude sparrows enhance metabolic and thermogenic capacities in the pectoralis muscle and suppress in the liver relative to their lowland counterparts. Gen Comp Endocrinol 2021; 313:113890. [PMID: 34453929 DOI: 10.1016/j.ygcen.2021.113890] [Citation(s) in RCA: 5] [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: 06/05/2021] [Revised: 08/01/2021] [Accepted: 08/18/2021] [Indexed: 11/26/2022]
Abstract
Animals living at high altitudes are challenged by the extreme environmental conditions of cold temperature and hypobaric hypoxia. It is not well understood how high-altitude birds enhance the capacity of metabolic thermogenesis and allocate metabolic capacity in different organs to maximize survival in extreme conditions of a cold winter. The Qinghai-Tibet Plateau (QTP) is the largest and highest plateau globally, offering a natural laboratory for investigating coping mechanisms of organisms inhabiting extreme environments. To understand the adaptive strategies in the morphology and physiology of small songbirds on the QTP, we compared plasma triiodothyronine (T3), pectoralis muscle mitochondrial cytochrome c oxidase (COX) and state IV capacities, the expression of peroxisome proliferator-activated receptor γ coactivator α (PGC-1α), adenine nucleotide translocase (ANT), uncoupling protein (UCP), and adenosine monophosphate-dependent kinase (AMPK) α1 mRNA in the pectoralis and liver of Eurasian tree sparrows (Passer montanus) from high-altitude (3,230 m), medium-altitude (1400 m), and low-altitude (80 m) regions. Our results showed that high-altitude sparrows had greater body masses, longer wings and tarsometatarsi, but comparable bill lengths relative to medium- and low-altitude individuals. High-altitude sparrows had higher plasma T3 levels and pectoralis muscle mitochondrial COX capacities than their lowland counterparts. They also upregulated the pectoralis muscle mRNA expression of UCP, PGC-1α, and ANT proteins relative to low-altitude sparrows. Unlike pectoralis, high-altitude sparrows significantly down-regulated hepatic AMPKα1 and ANT protein expression as compared with their lowland counterparts. Our results contribute to understanding the morphological, biochemical, and molecular adaptations in free-living birds to cope with the cold seasons in the extreme environment of the QTP.
Collapse
Affiliation(s)
- Ghulam Nabi
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Danning Xing
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Yanfeng Sun
- Ocean College, Hebei Agricultural University, Qinhuangdao, China
| | - Qian Zhang
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Mo Li
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Chuan Jiang
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Ibrahim M Ahmad
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - John C Wingfield
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, CA, USA
| | - Yuefeng Wu
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Dongming Li
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, China.
| |
Collapse
|
22
|
Weiner J, Roth L, Kranz M, Brust P, Boelen A, Klöting N, Heiker JT, Blüher M, Tönjes A, Pfluger PT, Stumvoll M, Mittag J, Krause K. Leptin counteracts hypothermia in hypothyroidism through its pyrexic effects and by stabilizing serum thyroid hormone levels. Mol Metab 2021; 54:101348. [PMID: 34610354 PMCID: PMC8556519 DOI: 10.1016/j.molmet.2021.101348] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 11/30/2022] Open
Abstract
Objective Thyroid hormones (TH) are essential for the homeostatic control of energy metabolism and the regulation of body temperature. The hypothalamic–pituitary–thyroid (HPT) axis is regulated by negative feedback mechanisms, ensuring that TH levels are maintained at a constant level. However, the feedback mechanisms underlying the resetting of the HPT axis regulation in the control of body temperature are still not fully understood. Here, we aimed to determine the thermoregulatory response in hypothyroid mice to different environmental temperatures and the underlying mechanisms. Methods Distinct thermogenic challenges were induced in hypothyroid female C57BL/6N and leptin-deficient ob/ob mice through housing at either room temperature or thermoneutrality. The thermogenic and metabolic effects were analyzed through metabolic chambers, 18F-FDG-PET/MRI, infrared thermography, metabolic profiling, histology, gene expression and Western blot analysis. Results In hypothyroid mice maintained at room temperature, high leptin serum levels induce a pyrexic effect leading to the stabilization of body temperature through brown adipose tissue thermogenesis and white adipose tissue browning. Housing at thermoneutrality leads to the normalization of leptin levels and a reduction of the central temperature set point, resulting in decreased thermogenesis in brown and white adipose tissue and skeletal muscle and a significant decline in body temperature. Furthermore, anapyrexia in hypothyroid leptin-deficient ob/ob mice indicates that besides its pyrexic actions, leptin exerts a stimulatory effect on the HPT axis to stabilize the remaining TH serum levels in hypothyroid mice. Conclusion This study led to the identification of a previously unknown endocrine loop in which leptin acts in concert with the HPT axis to stabilize body temperature in hypothyroid mice. Thyroid hormones are essential for the regulation of body temperature. Thyroid hormone-deficient (hypothyroid) mice show distinct leptin serum concentrations in response to changes in ambient housing temperature. High leptin serum levels confer a stimulatory effect on the hypothalamic-pituitary-thyroid axis. High leptin serum level prevents fall in body temperature in hypothyroid mice at room temperature through its pyrexic effects.
Collapse
Affiliation(s)
- Juliane Weiner
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Lisa Roth
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Mathias Kranz
- University Hospital of North Norway, Tromsø, Norway; Helmholtz-Zentrum Dresden-Rossendorf, Department of Neuroradiopharmaceuticals, Leipzig, Germany
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf, Department of Neuroradiopharmaceuticals, Leipzig, Germany
| | - Anita Boelen
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Nora Klöting
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany; Helmholtz Zentrum München, Helmholtz Institute for Metabolic, Obesity and Vascular Research, Leipzig, Germany
| | - John T Heiker
- Helmholtz Zentrum München, Helmholtz Institute for Metabolic, Obesity and Vascular Research, Leipzig, Germany
| | - Matthias Blüher
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany; Helmholtz Zentrum München, Helmholtz Institute for Metabolic, Obesity and Vascular Research, Leipzig, Germany
| | - Anke Tönjes
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Paul T Pfluger
- Helmholtz Zentrum München, Research Unit NeuroBiology of Diabetes, Neuherberg, Germany; Technical University of Munich (TUM), TUM School of Medicine, NeuroBiology of Diabetes, Munich, Germany
| | - Michael Stumvoll
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany; Helmholtz Zentrum München, Helmholtz Institute for Metabolic, Obesity and Vascular Research, Leipzig, Germany
| | - Jens Mittag
- Institute for Endocrinology & Diabetes/CBBM, University of Lübeck, Lübeck, Germany
| | - Kerstin Krause
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany.
| |
Collapse
|
23
|
Walczak K, Sieminska L. Obesity and Thyroid Axis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18189434. [PMID: 34574358 PMCID: PMC8467528 DOI: 10.3390/ijerph18189434] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/28/2021] [Accepted: 09/03/2021] [Indexed: 12/19/2022]
Abstract
Development of obesity is primarily the result of imbalance between energy intake and energy expenditure. Thyroid hormones influence energy expenditure by regulating cellular respiration and thermogenesis and by determining resting metabolic rate. Triiodothyronine influences lipid turnover in adipocytes and impacts appetite regulation through the central nervous system, mainly the hypothalamus. Thyroid-stimulating hormone may also influence thermogenesis, suppress appetite and regulate lipid storage through lipolysis and lipogenesis control. Subclinical hypothyroidism may induce changes in basal metabolic rate with subsequent increase in BMI, but obesity can also affect thyroid function via several mechanisms such as lipotoxicity and changes in adipokines and inflammatory cytokine secretion. The present study investigated the complex and mutual relationships between the thyroid axis and adiposity.
Collapse
Affiliation(s)
- Krzysztof Walczak
- Department of Thoracic Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland;
| | - Lucyna Sieminska
- Department of Pathophysiology and Endocrinology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland
- Correspondence:
| |
Collapse
|
24
|
Correa‐da‐Silva F, Fliers E, Swaab DF, Yi C. Hypothalamic neuropeptides and neurocircuitries in Prader Willi syndrome. J Neuroendocrinol 2021; 33:e12994. [PMID: 34156126 PMCID: PMC8365683 DOI: 10.1111/jne.12994] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/19/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
Prader-Willi Syndrome (PWS) is a rare and incurable congenital neurodevelopmental disorder, resulting from the absence of expression of a group of genes on the paternally acquired chromosome 15q11-q13. Phenotypical characteristics of PWS include infantile hypotonia, short stature, incomplete pubertal development, hyperphagia and morbid obesity. Hypothalamic dysfunction in controlling body weight and food intake is a hallmark of PWS. Neuroimaging studies have demonstrated that PWS subjects have abnormal neurocircuitry engaged in the hedonic and physiological control of feeding behavior. This is translated into diminished production of hypothalamic effector peptides which are responsible for the coordination of energy homeostasis and satiety. So far, studies with animal models for PWS and with human post-mortem hypothalamic specimens demonstrated changes particularly in the infundibular and the paraventricular nuclei of the hypothalamus, both in orexigenic and anorexigenic neural populations. Moreover, many PWS patients have a severe endocrine dysfunction, e.g. central hypogonadism and/or growth hormone deficiency, which may contribute to the development of increased fat mass, especially if left untreated. Additionally, the role of non-neuronal cells, such as astrocytes and microglia in the hypothalamic dysregulation in PWS is yet to be determined. Notably, microglial activation is persistently present in non-genetic obesity. To what extent microglia, and other glial cells, are affected in PWS is poorly understood. The elucidation of the hypothalamic dysfunction in PWS could prove to be a key feature of rational therapeutic management in this syndrome. This review aims to examine the evidence for hypothalamic dysfunction, both at the neuropeptidergic and circuitry levels, and its correlation with the pathophysiology of PWS.
Collapse
Affiliation(s)
- Felipe Correa‐da‐Silva
- Department of Endocrinology and MetabolismAmsterdam Gastroenterology Endocrinology and MetabolismAmsterdam University Medical Center (UMC)University of AmsterdamAmsterdamThe Netherlands
- Laboratory of EndocrinologyAmsterdam University Medical Center (UMC)University of AmsterdamAmsterdamThe Netherlands
- Department of Neuropsychiatric DisordersNetherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and SciencesAmsterdamThe Netherlands
| | - Eric Fliers
- Department of Endocrinology and MetabolismAmsterdam Gastroenterology Endocrinology and MetabolismAmsterdam University Medical Center (UMC)University of AmsterdamAmsterdamThe Netherlands
| | - Dick F. Swaab
- Department of Neuropsychiatric DisordersNetherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and SciencesAmsterdamThe Netherlands
| | - Chun‐Xia Yi
- Department of Endocrinology and MetabolismAmsterdam Gastroenterology Endocrinology and MetabolismAmsterdam University Medical Center (UMC)University of AmsterdamAmsterdamThe Netherlands
- Laboratory of EndocrinologyAmsterdam University Medical Center (UMC)University of AmsterdamAmsterdamThe Netherlands
- Department of Neuropsychiatric DisordersNetherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and SciencesAmsterdamThe Netherlands
| |
Collapse
|
25
|
Santocildes G, Viscor G, Pagès T, Ramos-Romero S, Torres JL, Torrella JR. Physiological Effects of Intermittent Passive Exposure to Hypobaric Hypoxia and Cold in Rats. Front Physiol 2021; 12:673095. [PMID: 34135770 PMCID: PMC8201611 DOI: 10.3389/fphys.2021.673095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/15/2021] [Indexed: 11/25/2022] Open
Abstract
The benefits of intermittent hypobaric hypoxia (IHH) exposure for health and its potential use as a training tool are well-documented. However, since hypobaric hypoxia and cold are environmental factors always strongly associated in the biosphere, additive or synergistic adaptations could have evolved in animals' genomes. For that reason, the aim of the present study was to investigate body composition and hematological and muscle morphofunctional responses to simultaneous intermittent exposure to hypoxia and cold. Adult male rats were randomly divided into four groups: (1) control, maintained in normoxia at 25°C (CTRL); (2) IHH exposed 4 h/day at 4,500 m (HYPO); (3) intermittent cold exposed 4 h/day at 4°C (COLD); and (4) simultaneously cold and hypoxia exposed (COHY). At the end of 9 and 21 days of exposure, blood was withdrawn and gastrocnemius (GAS) and tibialis anterior muscles, perigonadal and brown adipose tissue, diaphragm, and heart were excised. GAS transversal sections were stained for myofibrillar ATPase and succinate dehydrogenase for fiber typing and for endothelial ATPase to assess capillarization. Hypoxia-inducible factor 1α (HIF-1α), vascular endothelial growth factor (VEGF), and glucose transporter 1 (GLUT1) from GAS samples were semi-quantified by Western blotting. COLD and HYPO underwent physiological adjustments such as higher brown adipose tissue weight and increase in blood-related oxygen transport parameters, while avoiding some negative effects of chronic exposure to cold and hypoxia, such as body weight and muscle mass loss. COHY presented an additive erythropoietic response and was prevented from right ventricle hypertrophy. Intermittent cold exposure induced muscle angiogenesis, and IHH seems to indicate better muscle oxygenation through fiber area reduction.
Collapse
Affiliation(s)
- Garoa Santocildes
- Secció de Fisiologia, Departament de Biologia Cellular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Ginés Viscor
- Secció de Fisiologia, Departament de Biologia Cellular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Teresa Pagès
- Secció de Fisiologia, Departament de Biologia Cellular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Sara Ramos-Romero
- Secció de Fisiologia, Departament de Biologia Cellular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Departament de Química Biològica, Institut de Química Avançada de Catalunya (IQAC-CSIC), Barcelona, Spain
| | - Josep Lluís Torres
- Departament de Química Biològica, Institut de Química Avançada de Catalunya (IQAC-CSIC), Barcelona, Spain
| | - Joan Ramon Torrella
- Secció de Fisiologia, Departament de Biologia Cellular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
26
|
Bacillus subtilis-Based Probiotic Improves Skeletal Health and Immunity in Broiler Chickens Exposed to Heat Stress. Animals (Basel) 2021; 11:ani11061494. [PMID: 34064126 PMCID: PMC8224346 DOI: 10.3390/ani11061494] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary High ambient temperature is a major environmental stressor affecting the physiological and behavioral status of animals, increasing stress susceptibility and immunosuppression, and consequently increasing intestinal permeability (leaky gut) and related neuroinflammation. Probiotics, as well as prebiotics and synbiotics, have been used to prevent or decrease stress-associated detrimental effects on physiological and behavioral homeostasis in humans and various animals. The current data indicate that a dietary probiotic supplement, Bacillus subtilis, reduces heat stress-induced abnormal behaviors and negative effects on skeletal health in broilers through a variety of cellular responses, regulating the functioning of the microbiota–gut–brain axis and/or microbiota-modulated immunity during bone remodeling under thermoneutral and heat-stressed conditions. Abstract The elevation of ambient temperature beyond the thermoneutral zone leads to heat stress, which is a growing health and welfare issue for homeothermic animals aiming to maintain relatively constant reproducibility and survivability. Particularly, global warming over the past decades has resulted in more hot days with more intense, frequent, and long-lasting heat waves, resulting in a global surge in animals suffering from heat stress. Heat stress causes pathophysiological changes in animals, increasing stress sensitivity and immunosuppression, consequently leading to increased intestinal permeability (leaky gut) and related neuroinflammation. Probiotics, as well as prebiotics and synbiotics, have been used to prevent or reduce stress-induced negative effects on physiological and behavioral homeostasis in humans and various animals. The current data indicate dietary supplementation with a Bacillus subtilis-based probiotic has similar functions in poultry. This review highlights the recent findings on the effects of the probiotic Bacillus subtilis on skeletal health of broiler chickens exposed to heat stress. It provides insights to aid in the development of practical strategies for improving health and performance in poultry.
Collapse
|
27
|
Ouchi Y, Chowdhury VS, Cockrem JF, Bungo T. Av-UCP single nucleotide polymorphism affects heat production during cold exposure in chicks. J Therm Biol 2021; 98:102909. [PMID: 34016336 DOI: 10.1016/j.jtherbio.2021.102909] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/11/2021] [Accepted: 03/11/2021] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Uncoupling protein one (UCP1) is involved in thermogenesis, especially in non-shivering heat production. In chickens, a single nucleotide polymorphism (SNP) of the av-UCP (avian UCP) gene has been reported to be associated with body weight gain and increased abdominal fat. The purpose of this study was to examine the relationship between the av-UCP gene SNP and heat production in chicks. METHODS C/C and T/T male chicks (Rhode Island Red) of av-UCP gene SNP (g. 1270, C > T) were exposed to a low temperature environment (16 °C for 15 min) and their physiological responses were compared. RESULTS After cold exposure, mean rectal temperatures of C/C chicks were higher than those of T/T chicks. In pectoral muscle, genes expression of av-UCP and carnitine palmitoyltransferase-1 were higher in C/C chicks than T/T chicks. Hypothalamic expression levels of thyrotropin-releasing hormone and proopiomelanocortin genes were higher in C/C chicks than T/T chicks. Expression of hypothalamic corticotropin-releasing hormone, arginine vasotocin, brain-derived neurotrophic factor and neuropeptide Y genes did not differ between C/C and T/T chicks. In addition, plasma free fatty acid levels in C/C chicks were lower than those of T/T chicks. CONCLUSION These results suggest that the av-UCP gene SNP affects non-shivering heat production via the hypothalamo-pituitary-thyroid axis and fatty acid metabolism in the chicken.
Collapse
Affiliation(s)
- Yoshimitsu Ouchi
- Laboratory of Animal Behavior and Physiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, 739-8528, Japan
| | - Vishwajit S Chowdhury
- Graduate School of Bioresource and Bioenvironmental Sciences, Faculty of Arts and Science, Kyushu University, Fukuoka, 819-0395, Japan
| | - John F Cockrem
- School of Veterinary Science, Massey University, Palmerston North, 4442, New Zealand
| | - Takashi Bungo
- Laboratory of Animal Behavior and Physiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, 739-8528, Japan.
| |
Collapse
|
28
|
Dardente H, Migaud M. Thyroid hormone and hypothalamic stem cells in seasonal functions. VITAMINS AND HORMONES 2021; 116:91-131. [PMID: 33752829 DOI: 10.1016/bs.vh.2021.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Seasonal rhythms are a pervasive feature of most living organisms, which underlie yearly timeliness in breeding, migration, hibernation or weight gain and loss. To achieve this, organisms have developed inner timing devices (circannual clocks) that endow them with the ability to predict then anticipate changes to come, usually using daylength as the proximate cue. In Vertebrates, daylength interpretation involves photoperiodic control of TSH production by the pars tuberalis (PT) of the pituitary, which governs a seasonal switch in thyroid hormone (TH) availability in the neighboring hypothalamus. Tanycytes, specialized glial cells lining the third ventricle (3V), are responsible for this TH output through the opposite, PT-TSH-driven, seasonal control of deiodinases 2/3 (Dio 2/3). Tanycytes comprise a photoperiod-sensitive stem cell niche and TH is known to play major roles in cell proliferation and differentiation, which suggests that seasonal control of tanycyte proliferation may be involved in the photoperiodic synchronization of seasonal rhythms. Here we review our current knowledge of the molecular and neuroendocrine pathway linking photoperiodic information to seasonal changes in physiological functions and discuss the potential implication of tanycytes, TH and cell proliferation in seasonal timing.
Collapse
Affiliation(s)
- Hugues Dardente
- PRC, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France.
| | - Martine Migaud
- PRC, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| |
Collapse
|
29
|
Schredelseker T, Veit F, Dorsky RI, Driever W. Bsx Is Essential for Differentiation of Multiple Neuromodulatory Cell Populations in the Secondary Prosencephalon. Front Neurosci 2020; 14:525. [PMID: 32581684 PMCID: PMC7290237 DOI: 10.3389/fnins.2020.00525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/28/2020] [Indexed: 01/17/2023] Open
Abstract
The hypothalamus is characterized by great neuronal diversity, with many neuropeptides and other neuromodulators being expressed within its multiple anatomical domains. The regulatory networks directing hypothalamic development have been studied in detail, but, for many neuron types, control of differentiation is still not understood. The highly conserved Brain-specific homeobox (Bsx) transcription factor has previously been described in regulating Agrp and Npy expression in the hypothalamic arcuate nucleus (ARC) in mice. While Bsx is expressed in many more subregions of both tuberal and mamillary hypothalamus, the functions therein are not known. Using genetic analyses in zebrafish, we show that most bsx expression domains are dependent on Nkx2.1 and Nkx2.4 homeodomain transcription factors, while a subset depends on Otp. We show that the anatomical pattern of the ventral forebrain appears normal in bsx mutants, but that Bsx is necessary for the expression of many neuropeptide encoding genes, including agrp, penka, vip, trh, npb, and nts, in distinct hypothalamic anatomical domains. We also found Bsx to be critical for normal expression of two Crh family members, crhb and uts1, as well as crhbp, in the hypothalamus and the telencephalic septal region. Furthermore, we demonstrate a crucial role for Bsx in serotonergic, histaminergic and nitrergic neuron development in the hypothalamus. We conclude that Bsx is critical for the terminal differentiation of multiple neuromodulatory cell types in the forebrain.
Collapse
Affiliation(s)
- Theresa Schredelseker
- Developmental Biology, Institute Biology 1, Faculty of Biology, Albert Ludwig University of Freiburg, Freiburg im Breisgau, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg im Breisgau, Germany.,CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg im Breisgau, Germany
| | - Florian Veit
- Developmental Biology, Institute Biology 1, Faculty of Biology, Albert Ludwig University of Freiburg, Freiburg im Breisgau, Germany
| | - Richard I Dorsky
- Department of Neurobiology & Anatomy, University of Utah, Salt Lake City, UT, United States
| | - Wolfgang Driever
- Developmental Biology, Institute Biology 1, Faculty of Biology, Albert Ludwig University of Freiburg, Freiburg im Breisgau, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg im Breisgau, Germany.,CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
30
|
Baranowska-Bik A, Bik W. The Association of Obesity with Autoimmune Thyroiditis and Thyroid Function-Possible Mechanisms of Bilateral Interaction. Int J Endocrinol 2020; 2020:8894792. [PMID: 33381173 PMCID: PMC7755496 DOI: 10.1155/2020/8894792] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 11/28/2020] [Accepted: 12/04/2020] [Indexed: 12/17/2022] Open
Abstract
A growing number of patients suffer from autoimmune diseases, including autoimmune thyroid disease. There has simultaneously been a significant increase in the prevalence of obesity worldwide. It is still an open question whether adiposity can directly influence activation of inflammatory processes affecting the thyroid in genetically predisposed individuals. Adipokines, biologically active substances derived from the adipocytes, belong to a heterogenic group of compounds involved in numerous physiological functions, including the maintenance of metabolism, hormonal balance, and immune response. Notably, the presence of obesity worsens the course of selected autoimmune diseases and impairs response to treatment. Moreover, the excess of body fat may result in the progression of autoimmune diseases. Nutritional status, body weight, and energy expenditure may influence thyroid hormone secretion. Interestingly, thyroid hormones might influence the activity of adipose tissue as metabolic alterations related to fat tissue are observed under pathological conditions in which there are deficits or overproduction of thyroid hormones. Functioning TSH receptors are expressed on adipocytes. Thermogenesis may presumably be stimulated by TSH binding to its receptor on brown adipocytes. There could be a bilateral interaction between the thyroid and adipose. Obesity may influence the onset and course of autoimmune disease.
Collapse
Affiliation(s)
- Agnieszka Baranowska-Bik
- Department of Endocrinology, Centre of Postgraduate Medical Education, Ceglowska 80, Warsaw 01-809, Poland
| | - Wojciech Bik
- Department of Neuroendocrinology, Centre of Postgraduate Medical Education, Marymoncka 99/103, Warsaw 01-813, Poland
| |
Collapse
|
31
|
Napolitano G, Fasciolo G, Di Meo S, Venditti P. Vitamin E Supplementation and Mitochondria in Experimental and Functional Hyperthyroidism: A Mini-Review. Nutrients 2019; 11:nu11122900. [PMID: 31805673 PMCID: PMC6950234 DOI: 10.3390/nu11122900] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 02/06/2023] Open
Abstract
Mitochondria are both the main sites of production and the main target of reactive oxygen species (ROS). This can lead to mitochondrial dysfunction with harmful consequences for the cells and the whole organism, resulting in metabolic and neurodegenerative disorders such as type 2 diabetes, obesity, dementia, and aging. To protect themselves from ROS, mitochondria are equipped with an efficient antioxidant system, which includes low-molecular-mass molecules and enzymes able to scavenge ROS or repair the oxidative damage. In the mitochondrial membranes, a major role is played by the lipid-soluble antioxidant vitamin E, which reacts with the peroxyl radicals faster than the molecules of polyunsaturated fatty acids, and in doing so, protects membranes from excessive oxidative damage. In the present review, we summarize the available data concerning the capacity of vitamin E supplementation to protect mitochondria from oxidative damage in hyperthyroidism, a condition that leads to increased mitochondrial ROS production and oxidative damage. Vitamin E supplementation to hyperthyroid animals limits the thyroid hormone-induced increases in mitochondrial ROS and oxidative damage. Moreover, it prevents the reduction of the high functionality components of the mitochondrial population induced by hyperthyroidism, thus preserving cell function.
Collapse
Affiliation(s)
- Gaetana Napolitano
- Dipartimento di Scienze e Tecnologie, Università degli Studi di Napoli Parthenope, via Acton n. 38, I-0133 Napoli, Italy;
| | - Gianluca Fasciolo
- Dipartimento di Biologia, Università di Napoli Federico II, Complesso Universitario Monte Sant’Angelo, Via Cinthia, I-80126 Napoli, Italy; (G.F.); (S.D.M.)
| | - Sergio Di Meo
- Dipartimento di Biologia, Università di Napoli Federico II, Complesso Universitario Monte Sant’Angelo, Via Cinthia, I-80126 Napoli, Italy; (G.F.); (S.D.M.)
| | - Paola Venditti
- Dipartimento di Biologia, Università di Napoli Federico II, Complesso Universitario Monte Sant’Angelo, Via Cinthia, I-80126 Napoli, Italy; (G.F.); (S.D.M.)
- Correspondence: ; Tel.: +39-081-2535080; Fax: +39-081-679233
| |
Collapse
|
32
|
Thyroid Disorders in Homozygous β-Thalassemia: Current Knowledge, Emerging Issues and Open Problems. Mediterr J Hematol Infect Dis 2019; 11:e2019029. [PMID: 31205633 PMCID: PMC6548211 DOI: 10.4084/mjhid.2019.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 03/08/2019] [Indexed: 12/19/2022] Open
Abstract
Changes in thyroid function and thyroid function tests occur in patients with β-thalassemia major (TM). The frequency of hypothyroidism in TM patients ranges from 4% to 29 % in different reports. The wide variation has been attributed to several factors such as patients’ genotype, age, ethnic heterogeneity, treatment protocols of transfusions and chelation, and varying compliance to treatment. Hypothyroidism is the result of primary gland failure or insufficient thyroid gland stimulation by the hypothalamus or pituitary gland. The main laboratory parameters of thyroid function are the assessments of serum thyroid-stimulating hor-mone (TSH) and serum free thyroxine (FT4). It is of primary importance to interpret these measurements within the context of the laboratory-specific normative range for each test. An elevated serum TSH level with a standard range of serum FT4 level is consistent with subclinical hypothyroidism. A low serum FT4 level with a low, or inappropriately normal, serum TSH level is consistent with secondary hypothyroidism. Doctors caring for TM patients most commonly encounter subjects with subclinical primary hypothyroidism in the second decade of life. Several aspects remain to be elucidated as the frequency of thyroid cancer and the possible existence of a relationship between thyroid dysfunction, on one hand, cardiovascular diseases, components of metabolic syndrome (insulin resistance) and hypercoagulable state, on the other hand. Further studies are needed to explain these emerging issues. Following a brief description of thyroid hormone regulation, production and actions, this article is conceptually divided into two parts; the first reports the spectrum of thyroid disease occurring in patients with TM, and the second part focuses on the emerging issues and the open problems in TM patients with thyroid disorders.
Collapse
|