1
|
Soares De Oliveira L, Kaserman JE, Van Der Spek AH, Lee NJ, Undeutsch HJ, Werder RB, Wilson AA, Hollenberg AN. Thyroid hormone receptor beta (THRβ1) is the major regulator of T3 action in human iPSC-derived hepatocytes. Mol Metab 2024:102057. [PMID: 39481850 DOI: 10.1016/j.molmet.2024.102057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/08/2024] [Accepted: 10/24/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVE Thyroid hormone (TH) action is mediated by thyroid hormone receptor (THR) isoforms. While THRβ1 is likely the main isoform expressed in liver, its role in human hepatocytes is not fully understood. METHODS To elucidate the role of THRβ1 action in human hepatocytes we used CRISPR/Cas9 editing to knock out THRβ1 in induced pluripotent stem cells (iPSC). Following directed differentiation to the hepatic lineage, iPSC-derived hepatocytes were then interrogated to determine the role of THRβ1 in ligand-independent and -dependent functions. RESULTS We found that the loss of THRβ1 promoted alterations in proliferation rate and metabolic pathways regulated by T3, including gluconeogenesis, lipid oxidation, fatty acid synthesis, and fatty acid uptake. We observed that key genes involved in liver metabolism are regulated through both T3 ligand-dependent and -independent THRβ1 signaling mechanisms. Finally, we demonstrate that following THRβ1 knockout, several key metabolic genes remain T3 responsive suggesting they are THRα targets. CONCLUSIONS These results highlight that iPSC-derived hepatocytes are an effective platform to study mechanisms regulating TH signaling in human hepatocytes.
Collapse
Affiliation(s)
- Lorraine Soares De Oliveira
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Cornell Medicine, New York, NY 10021, USA; Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, USA; Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Joseph E Kaserman
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, USA; Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Anne H Van Der Spek
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Cornell Medicine, New York, NY 10021, USA; Department of Endocrinology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam 1081 HV, Netherlands
| | - Nora J Lee
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, USA; Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Hendrik J Undeutsch
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Cornell Medicine, New York, NY 10021, USA; Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, USA; Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Rhiannon B Werder
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, USA; Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Andrew A Wilson
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, USA; Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA.
| | - Anthony N Hollenberg
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Cornell Medicine, New York, NY 10021, USA; Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, USA; Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA.
| |
Collapse
|
2
|
Borisov V, Shkil F. Effects and phenotypic consequences of transient thyrotoxicosis and hypothyroidism at different stages of zebrafish Danio rerio (Teleostei; Cyprinidae) skeleton development. Anat Rec (Hoboken) 2024. [PMID: 39431292 DOI: 10.1002/ar.25592] [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: 06/27/2024] [Revised: 09/21/2024] [Accepted: 10/04/2024] [Indexed: 10/22/2024]
Abstract
The effects and consequences of changes in thyroid hormones (THs) level are among the actively studied topics in teleost developmental and evolutionary biology. In most of the experimental models used, the altered hormonal status (either hypo- or hyperthyroidism) is a stable characteristic of the developing organism, and the observed phenotypic outcomes are the cumulative consequences of multiple TH-induced developmental changes. Meanwhile, the influence of the transient fluctuations of TH content on skeleton development has been much less studied. Here, we present experimental data on the developmental effects and phenotypic consequences of transient, pharmacologically induced thyrotoxicosis and hypothyroidism at different stages of ossified skeleton patterning in zebrafish. According to the results, the skeleton structures differed in TH sensitivity. Some showed a notable shift in the developmental timing and rate, while other demonstrated little or no response to changes in TH content. The developmental stages also differed in TH sensitivity. We identified a relatively short developmental period, during which changes in TH level significantly increased the developmental instability and plasticity, leading to phenotypic consequences comparable to those in fish with a persistent hypo- or hyperthyroidism. These findings allow this period to be considered as a critical developmental window.
Collapse
Affiliation(s)
- Vasily Borisov
- A.N. Severtsov Institute of Ecology and Evolution, RAS, Moscow, Russia
| | - Fedor Shkil
- A.N. Severtsov Institute of Ecology and Evolution, RAS, Moscow, Russia
- N.K. Koltzov Institute of Developmental Biology, RAS, Moscow, Russia
| |
Collapse
|
3
|
Raterman ST, Wagener FADTG, Zethof J, Cuijpers V, Klaren PHM, Metz JR, Von den Hoff JW. foxe1 mutant zebrafish show indications of a hypothyroid phenotype and increased sensitivity to ethanol for craniofacial malformations. Dev Dyn 2024. [PMID: 39360443 DOI: 10.1002/dvdy.745] [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: 02/15/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND FOXE1 mutations in humans are associated with cleft palate and hypothyroidism. We previously developed a foxe1 mutant zebrafish demonstrating mineralization defects in larvae. In the present study, we investigate the thyroid status and skeletal phenotype of adult foxe1 mutants. RESULTS Mutant fish have increased expression of tshβ in the pituitary, and of hepatic dio1 and dio2. In plasma, we found higher Mg levels. Together these findings are indicative of hypothyroidism. We further observed mineralization defects in scales due to enhanced osteoclast activity as measured by increased expression levels of tracp, ctsk, and rankl. Gene-environment interactions in the etiology of FOXE1-related craniofacial abnormalities remain elusive, which prompts the need for models to investigate genotype-phenotype associations. We here investigated whether ethanol exposure increases the risk of developing craniofacial malformations in foxe1 mutant larvae that we compared to wild types. We found in ethanol-exposed mutants an increased incidence of developmental malformations and marked changes in gene expression patterns of cartilage markers (sox9a), apoptotic markers (casp3b), retinoic acid metabolism (cyp26c1), and tissue hypoxia markers (hifaa, hifab). CONCLUSION Taken together, this study shows that the foxe1 mutant zebrafish recapitulates phenotypes associated with FOXE1 mutations in human patients and a clear foxe1-ethanol interaction.
Collapse
Affiliation(s)
- Sophie T Raterman
- Department of Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Plant & Animal Biology, Radboud Institute for Biological and Environmental Sciences (RIBES), Radboud University, Nijmegen, The Netherlands
| | - Frank A D T G Wagener
- Department of Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan Zethof
- Department of Plant & Animal Biology, Radboud Institute for Biological and Environmental Sciences (RIBES), Radboud University, Nijmegen, The Netherlands
| | - Vincent Cuijpers
- Department of Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter H M Klaren
- Department of Plant & Animal Biology, Radboud Institute for Biological and Environmental Sciences (RIBES), Radboud University, Nijmegen, The Netherlands
| | - Juriaan R Metz
- Department of Plant & Animal Biology, Radboud Institute for Biological and Environmental Sciences (RIBES), Radboud University, Nijmegen, The Netherlands
| | - Johannes W Von den Hoff
- Department of Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
4
|
Yang L, Fu MF, Wang HY, Sun H. Research Advancements in the Interplay between T3 and Macrophages. Curr Med Sci 2024; 44:883-889. [PMID: 39446284 DOI: 10.1007/s11596-024-2935-6] [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: 12/14/2023] [Accepted: 09/03/2024] [Indexed: 10/25/2024]
Abstract
3,3',5-Triiodo-L-thyronine (T3) is a key endocrine hormone in the human body that plays crucial roles in growth, development, metabolism, and immune function. Macrophages, the key regulatory cells within the immune system, exhibit marked "heterogeneity" and "plasticity", with their phenotype and function subject to modulation by local environmental signals. The interplay between the endocrine and immune systems is well documented. Numerous studies have shown that T3 significantly target macrophages, highlighting them as key cellular components in this interaction. Through the regulation of macrophage function and phenotype, T3 influences immune function and tissue repair in the body. This review comprehensively summarizes the regulatory actions and mechanisms of T3 on macrophages, offering valuable insights into further research of the immunoregulatory effects of T3.
Collapse
Affiliation(s)
- Liu Yang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, 430022, China
| | - Meng-Fei Fu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, 430022, China
| | - Han-Yu Wang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, 430022, China
| | - Hui Sun
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, 430022, China.
| |
Collapse
|
5
|
Yilmaz U, Tanbek K. Intracerebroventricular prokineticin 2 infusion may play a role on the hypothalamus-pituitary-thyroid axis and energy metabolism. Physiol Behav 2024; 283:114601. [PMID: 38838800 DOI: 10.1016/j.physbeh.2024.114601] [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: 03/30/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 06/07/2024]
Abstract
AIM The hypothesis of this study is to determine the effects of intracerebroventricular (icv) prokineticin 2 infusion on food consumption and body weight and to elucidate whether it has effects on energy expenditure via the hypothalamus-pituitary-thyroid (HPT) axis in adipose tissue. MATERIAL AND METHODS A total of 40 rats were used in the study and 4 groups were established: Control, Sham, Prokineticin 1.5 and Prokineticin 4.5 (n=10). Except for the Control group, rats were treated intracerebroventricularly via osmotic minipumps, the Sham group was infused with aCSF (vehicle), and the Prokineticin 1.5 and Prokineticin 4.5 groups were infused with 1.5 nMol and 4.5 nMol prokineticin 2, respectively. Food and water consumption and body weight were monitored during 7-day infusion in all groups. At the end of the infusion, the rats were decapitated and serum TSH, fT4 and fT3 levels were determined by ELISA. In addition, PGC-1α and UCP1 gene expression levels in white adipose tissue (WAT) and brown adipose tissue (BAT), TRH from rat hypothalamic tissue were determined by real-time PCR. RESULTS Icv prokineticin 2 (4.5 nMol) infusion had no effect on water consumption but reduced daily food consumption and body weight (p<0.05). Icv prokineticin 2 (4.5 nMol) infusion significantly increased serum TSH, fT4 and fT3 levels when compared to Control and Sham groups (p<0.05). Also, icv prokineticin 2 (4.5 nMol) infusion increased the expression of TRH in the hypothalamus tissue and expression of PGC-1α UCP1 in the WAT and BAT (p<0.05). CONCLUSION Icv prokineticin 2 (4.5 nMol) infusion may suppress food consumption via its receptors in the hypothalamus and reduce body weight by stimulating energy expenditure and thermogenesis in adipose tissue through the HPT axis.
Collapse
Affiliation(s)
- Umit Yilmaz
- Department of Physiology, Faculty of Medicine, Karabuk University, Karabuk, Turkey.
| | - Kevser Tanbek
- Department of Physiology, Faculty of Medicine, Inonu University, Malatya, Turkey
| |
Collapse
|
6
|
Chu J, Zhao M, Hu X, Wang Q, Li X, Cui R, Wang L. Soluble Guanylate Cyclase α1 Gene Influences Egg-Laying Amount and Hatching Rate in Bombyx mori. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2024; 117:e22153. [PMID: 39323098 DOI: 10.1002/arch.22153] [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] [Received: 06/03/2024] [Revised: 07/26/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
Soluble guanylate cyclase (sGC) serves as a receptor of nitric oxide (NO) and is the core metalloenzyme in the NO signal transduction pathway. sGC plays a key role in the NO-cGMP signal transduction pathway and participates in various physiological processes, including cell differentiation, neuron transmission, and internal environment homeostasis. sGC consists of two subunits, α and β, each subunit containing multiple isoforms. In this study, we cloned and analyzed the sGC-α1 gene in the silkworm Bombyx mori (BmsGC-α1). The BmsGC-α1 gene was expressed highest at the pupal stages. The highest BmsGC-α1 mRNA expression was observed in the head of fifth instar larvae and in fat body during the wandering stage of B. mori. Furthermore, we observed that feeding fifth instar larvae with thyroid hormone and nitroglycerin induced the expression of the BmsGC-α1 gene. Injection of BmsGC-α1 siRNA into silkworms at the prepupal stage resulted in a significant decrease in BmsGC-α1 expression levels at 48 and 72 h postinjection. After silencing BmsGC-α1, both the egg-laying amount and hatching rate of silkworm eggs were significantly reduced compared to the control group. These results suggest that BmsGC-α1 plays an important role in regulating the reproductive system of silkworms. This finding enhances our understanding of the functional diversity of sGC in insects.
Collapse
Affiliation(s)
- Jianghong Chu
- Anhui Key Laboratory of Resource Insect Biology and Innovative Utilization, School of Life Science, Anhui Agricultural University, Hefei, China
| | - Mengting Zhao
- Anhui Key Laboratory of Resource Insect Biology and Innovative Utilization, School of Life Science, Anhui Agricultural University, Hefei, China
| | - Xiaoxuan Hu
- Anhui Key Laboratory of Resource Insect Biology and Innovative Utilization, School of Life Science, Anhui Agricultural University, Hefei, China
| | - Qing Wang
- Anhui Key Laboratory of Resource Insect Biology and Innovative Utilization, School of Life Science, Anhui Agricultural University, Hefei, China
| | - Xudong Li
- Anhui Key Laboratory of Resource Insect Biology and Innovative Utilization, School of Life Science, Anhui Agricultural University, Hefei, China
| | - Ruirui Cui
- Anhui Key Laboratory of Resource Insect Biology and Innovative Utilization, School of Life Science, Anhui Agricultural University, Hefei, China
| | - Lei Wang
- Anhui Key Laboratory of Resource Insect Biology and Innovative Utilization, School of Life Science, Anhui Agricultural University, Hefei, China
| |
Collapse
|
7
|
Yang Y, Valdés-Rives SA, Liu Q, Gao T, Burudpakdee C, Li Y, Tan J, Tan Y, Koch CA, Rong Y, Houser SR, Wei S, Cai KQ, Wu J, Cheng SY, Wechsler-Reya R, Yang ZJ. Thyroid hormone suppresses medulloblastoma progression through promoting terminal differentiation of tumor cells. Cancer Cell 2024; 42:1434-1449.e5. [PMID: 39137728 DOI: 10.1016/j.ccell.2024.07.008] [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: 11/17/2023] [Revised: 06/10/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024]
Abstract
Hypothyroidism is commonly detected in patients with medulloblastoma (MB). However, whether thyroid hormone (TH) contributes to MB pathogenicity remains undetermined. Here, we find that TH plays a critical role in promoting tumor cell differentiation. Reduction in TH levels frees the TH receptor, TRα1, to bind to EZH2 and repress expression of NeuroD1, a transcription factor that drives tumor cell differentiation. Increased TH reverses EZH2-mediated repression of NeuroD1 by abrogating the binding of EZH2 and TRα1, thereby stimulating tumor cell differentiation and reducing MB growth. Importantly, TH-induced differentiation of tumor cells is not restricted by the molecular subgroup of MB, suggesting that TH can be used to broadly treat MB subgroups. These findings establish an unprecedented association between TH signaling and MB pathogenicity, providing solid evidence for TH as a promising modality for MB treatment.
Collapse
Affiliation(s)
- Yijun Yang
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA; Cancer Epigenetic Institute, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA
| | - Silvia Anahi Valdés-Rives
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA; Cancer Epigenetic Institute, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tong Gao
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA
| | - Chakkapong Burudpakdee
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA; Cancer Epigenetic Institute, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA
| | - Yuzhe Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jun Tan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yinfei Tan
- Department of Pathology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA
| | - Christian A Koch
- Department of Medicine, Division of Endocrinology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA
| | - Yuan Rong
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University Health System, Philadelphia, PA 19140, USA
| | - Steven R Houser
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University Health System, Philadelphia, PA 19140, USA
| | - Shuanzeng Wei
- Department of Pathology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA
| | - Kathy Q Cai
- Histopathology Facility, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA
| | - Jinhua Wu
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA
| | - Sheue-Yann Cheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Robert Wechsler-Reya
- Brain Tumor Research, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - Zeng-Jie Yang
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA; Cancer Epigenetic Institute, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA.
| |
Collapse
|
8
|
Muralidharan A, Gomez GA, Kesavan C, Pourteymoor S, Larkin D, Tambunan W, Sechriest VF, Mohan S. Sex-Specific Effects of THRβ Signaling on Metabolic Responses to High Fat Diet in Mice. Endocrinology 2024; 165:bqae075. [PMID: 38935021 PMCID: PMC11237353 DOI: 10.1210/endocr/bqae075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/11/2024] [Accepted: 06/21/2024] [Indexed: 06/28/2024]
Abstract
Thyroid hormone (TH) plays a crucial role in regulating the functions of both bone and adipose tissue. Given that TH exerts its cholesterol-lowering effects in hepatic tissue through the TH receptor-β (TRβ), we hypothesized that TRβ agonist therapy using MGL3196 (MGL) would be effective in treating increased adiposity and bone loss in response to a 12-week high-fat diet (HFD) in adult C57BL/6J mice. Transcriptional and serum profiling revealed that HFD-induced leptin promoted weight gain in both males and females, but MGL only suppressed leptin induction and weight gain in males. In vitro studies suggest that estrogen suppresses MGL activity in adipocytes, indicating that estrogen might interfere with MGL-TRβ function. Compared to systemic adiposity, HFD reduced bone mass in male but not female mice. Paradoxically, MGL treatment reversed macroscopic bone mineral density loss in appendicular bones, but micro-CT revealed that MGL exacerbated HFD-induced trabecular bone loss, and reduced bone strength. In studies on the mechanisms for HFD effects on bone, we found that HFD induced Rankl expression in male femurs that was blocked by MGL. By ex vivo assays, we found that RANKL indirectly represses osteoblast lineage allocation of osteoprogenitors by induction of inflammatory cytokines TNFα, IL-1β, and CCL2. Finally, we found that MGL functions in both systemic adiposity and bone by nongenomic TRβ signaling, as HFD-mediated phenotypes were not rescued in TRβ147F knockout mice with normal genomic but defective nongenomic TRβ signaling. Our findings demonstrate that the negative effects of HFD on body fat and bone phenotypes are impacted by MGL in a gender-specific manner.
Collapse
Affiliation(s)
- Aruljothi Muralidharan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
| | - Gustavo A Gomez
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
| | - Chandrasekhar Kesavan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
| | - Sheila Pourteymoor
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
| | - Destiney Larkin
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
| | - William Tambunan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
| | - V Franklin Sechriest
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
| | - Subburaman Mohan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Department of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA
- Department of Orthopedic Surgery, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
9
|
Simpson JB, Walker ME, Sekela JJ, Ivey SM, Jariwala PB, Storch CM, Kowalewski ME, Graboski AL, Lietzan AD, Walton WG, Davis KA, Cloer EW, Borlandelli V, Hsiao YC, Roberts LR, Perlman DH, Liang X, Overkleeft HS, Bhatt AP, Lu K, Redinbo MR. Gut microbial β-glucuronidases influence endobiotic homeostasis and are modulated by diverse therapeutics. Cell Host Microbe 2024; 32:925-944.e10. [PMID: 38754417 PMCID: PMC11176022 DOI: 10.1016/j.chom.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/18/2024] [Accepted: 04/24/2024] [Indexed: 05/18/2024]
Abstract
Hormones and neurotransmitters are essential to homeostasis, and their disruptions are connected to diseases ranging from cancer to anxiety. The differential reactivation of endobiotic glucuronides by gut microbial β-glucuronidase (GUS) enzymes may influence interindividual differences in the onset and treatment of disease. Using multi-omic, in vitro, and in vivo approaches, we show that germ-free mice have reduced levels of active endobiotics and that distinct gut microbial Loop 1 and FMN GUS enzymes drive hormone and neurotransmitter reactivation. We demonstrate that a range of FDA-approved drugs prevent this reactivation by intercepting the catalytic cycle of the enzymes in a conserved fashion. Finally, we find that inhibiting GUS in conventional mice reduces free serotonin and increases its inactive glucuronide in the serum and intestines. Our results illuminate the indispensability of gut microbial enzymes in sustaining endobiotic homeostasis and indicate that therapeutic disruptions of this metabolism promote interindividual response variabilities.
Collapse
Affiliation(s)
- Joshua B Simpson
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Morgan E Walker
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Joshua J Sekela
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Samantha M Ivey
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Parth B Jariwala
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Cameron M Storch
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Mark E Kowalewski
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA
| | - Amanda L Graboski
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Adam D Lietzan
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William G Walton
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Kacey A Davis
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA
| | - Erica W Cloer
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Valentina Borlandelli
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Yun-Chung Hsiao
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lee R Roberts
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA
| | - David H Perlman
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA
| | - Xue Liang
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA
| | - Hermen S Overkleeft
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Aadra P Bhatt
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kun Lu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew R Redinbo
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA; Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
10
|
Castañeda-Cortés DC, Lefebvre-Raine M, Triffault-Bouchet G, Langlois VS. Toxicogenomics of Five Cytostatics in Fathead Minnow (Pimephales promelas) Larvae. BULLETIN OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2024; 112:66. [PMID: 38643435 DOI: 10.1007/s00128-024-03896-9] [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] [Received: 02/01/2024] [Accepted: 04/09/2024] [Indexed: 04/22/2024]
Abstract
In this study, the toxicogenomic effects of five cytostatics (tamoxifen, methotrexate, capecitabine, cyclophosphamide, and ifosfamide) on fathead minnow (Pimephales promelas) larvae were evaluated. Post-fertilization eggs were exposed to increasing concentrations of the drugs for six days. The expression levels of two genetic biomarkers for toxicity and four thyroid hormone-related gene pathways were measured. Interestingly, the results showed that all concentrations of the five cytostatics affect the transcription levels of both toxicity biomarker genes. Additionally, the thyroid hormone-related genes had different expression levels than the control, with the most significant changes observed in those larvae exposed to cyclophosphamide and ifosfamide. While a previous study found no effects on fish morphology, this study suggests that the five cytostatics modify subtle molecular responses of P. promelas, highlighting the importance of assessing multibiological level endpoints throughout the lifecycle of animals to understand the full portrait of potential effects of cytostatics and other contaminants.
Collapse
Affiliation(s)
- D C Castañeda-Cortés
- Institut National de La Recherche Scientifique (INRS), Centre Eau Terre Environnement (ETE), Quebec City, QC, Canada
| | - M Lefebvre-Raine
- Institut National de La Recherche Scientifique (INRS), Centre Eau Terre Environnement (ETE), Quebec City, QC, Canada
| | - G Triffault-Bouchet
- Ministère de l'Environnement, de la Lutte Contre les Changements Climatiques, de la Faune et des Parcs (MELCCFP), Centre d'expertise en analyse environnementale du Québec (CEAEQ), Quebec city, QC, Canada
| | - V S Langlois
- Institut National de La Recherche Scientifique (INRS), Centre Eau Terre Environnement (ETE), Quebec City, QC, Canada.
| |
Collapse
|
11
|
Yang Y, Valdés-Rives SA, Liu Q, Li Y, Tan J, Tan Y, Koch CA, Rong Y, Houser SR, Wei S, Cai KQ, Cheng SY, Curran T, Wechsler-Reya R, Yang ZJ. Thyroid Hormone Suppresses Medulloblastoma Progression Through Promoting Terminal Differentiation of Tumor Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.580111. [PMID: 38405864 PMCID: PMC10888774 DOI: 10.1101/2024.02.13.580111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Hypothyroidism is commonly detected in patients with medulloblastoma (MB). A possible link between thyroid hormone (TH) signaling and MB pathogenicity has not been reported. Here, we find that TH plays a critical role in promoting tumor cell differentiation. Reduction in TH levels frees the TH receptor, TRα1, to bind to EZH2 and repress expression of NeuroD1, a transcription factor that drives tumor cell differentiation. Increased TH reverses EZH2-mediated repression of NeuroD1 by abrogating the binding of EZH2 and TRα1, thereby stimulating tumor cell differentiation and reducing MB growth. Importantly, TH-induced differentiation of tumor cells is not restricted by the molecular subgroup of MB. These findings establish an unprecedented association between TH signaling and MB pathogenicity, providing solid evidence for TH as a promising modality for MB treatment.
Collapse
|
12
|
Cho YW, Fu Y, Huang CCJ, Wu X, Ng L, Kelley KA, Vella KR, Berg AH, Hollenberg AN, Liu H, Forrest D. Thyroid hormone-regulated chromatin landscape and transcriptional sensitivity of the pituitary gland. Commun Biol 2023; 6:1253. [PMID: 38081939 PMCID: PMC10713718 DOI: 10.1038/s42003-023-05546-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 11/03/2023] [Indexed: 12/18/2023] Open
Abstract
Thyroid hormone (3,5,3'-triiodothyronine, T3) is a key regulator of pituitary gland function. The response to T3 is thought to hinge crucially on interactions of nuclear T3 receptors with enhancers but these sites in pituitary chromatin remain surprisingly obscure. Here, we investigate genome-wide receptor binding in mice using tagged endogenous thyroid hormone receptor β (TRβ) and analyze T3-regulated open chromatin using an anterior pituitary-specific Cre driver (Thrbb2Cre). Strikingly, T3 regulates histone modifications and chromatin opening primarily at sites that maintain TRβ binding regardless of T3 levels rather than at sites where T3 abolishes or induces de novo binding. These sites associate more frequently with T3-activated than T3-suppressed genes. TRβ-deficiency blunts T3-regulated gene expression, indicating that TRβ confers transcriptional sensitivity. We propose a model of gene activation in which poised receptor-enhancer complexes facilitate adjustable responses to T3 fluctuations, suggesting a genomic basis for T3-dependent pituitary function or pituitary dysfunction in thyroid disorders.
Collapse
Affiliation(s)
- Young-Wook Cho
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yulong Fu
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Chen-Che Jeff Huang
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xuefeng Wu
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lily Ng
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kevin A Kelley
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Kristen R Vella
- Division of Endocrinology, Diabetes and Metabolism, Weill Department of Medicine Weill Cornell Medicine, New York, New York, 10065, USA
| | - Anders H Berg
- Department of Pathology, Cedars Sinai Medical Center, Los Angeles, California, 90048, USA
| | - Anthony N Hollenberg
- Division of Endocrinology, Diabetes and Metabolism, Weill Department of Medicine Weill Cornell Medicine, New York, New York, 10065, USA
| | - Hong Liu
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Douglas Forrest
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
13
|
Lin WH, Zhou SJ, Chen XH, Cao H, Chen Q. Changes of serum TSH, FT3, and FT4 levels in infants received surgical correction of congenital heart disease under cardiopulmonary bypass. BMC Cardiovasc Disord 2023; 23:562. [PMID: 37974091 PMCID: PMC10655303 DOI: 10.1186/s12872-023-03590-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023] Open
Abstract
OBJECTIVE This study aimed to explore the fluctuations and clinical relevance of serum thyrotropin (TSH), free triiodothyronine (FT3), and free thyroxine (FT4) levels in infants undergoing surgical correction for congenital heart disease (CHD) using cardiopulmonary bypass (CPB). METHODS In a retrospective design, 58 infants who underwent CHD surgical correction under CPB between January 2021 and January 2022 at our institution were incorporated. These infants were categorized into two groups: simple CHD (n = 34) and complex CHD (n = 24). TSH, FT3, and FT4 serum concentrations were assessed at four intervals: 24 h pre-surgery (T0) and 24 h (T1), 48 h (T2), and 72 h (T3) post-surgery. RESULTS The simple CHD group displayed a significantly reduced CPB duration compared to the complex CHD group (P < 0.001). Both groups exhibited a notable decline in serum thyroid hormone concentrations at T1 compared to T0. However, from T1 to T3, an upward trend in hormone levels was observed. By T3, though the levels in both groups had risen notably from T1, they remained significantly diminished from T0 (P < 0.01). In both the simple and complex CHD cohorts, significant fluctuations in thyroid hormone levels (TSH, FT3, FT4) were noted across the different timepoints (T0, T1, T3) (P < 0.01). While no significant disparities were found between the two groups' hormone concentrations at T0 and T1 (P > 0.05), at T2 and T3, the simple CHD group manifested higher TSH, FT3, and FT4 levels compared to the complex CHD group (P < 0.05). CONCLUSIONS Infants undergoing CHD surgical correction under CPB experience significant declines in TSH, FT3, and FT4 serum levels. The post-surgery thyroid hormone recovery was more pronounced in infants with simple CHD compared to those with complex CHD. As such, vigilant monitoring of thyroid hormone levels during the perioperative phase is imperative, and timely intervention measures should be employed when necessary.
Collapse
Affiliation(s)
- Wen-Hao Lin
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Si-Jia Zhou
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Xiu-Hua Chen
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Hua Cao
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China.
| | - Qiang Chen
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
14
|
Esin EV, Shulgina EV, Shkil FN. Rapid hyperthyroidism-induced adaptation of salmonid fish in response to environmental pollution. J Evol Biol 2023; 36:1471-1483. [PMID: 37731226 DOI: 10.1111/jeb.14220] [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: 05/04/2023] [Revised: 08/02/2023] [Accepted: 08/25/2023] [Indexed: 09/22/2023]
Abstract
The streams draining volcanic landscapes are often characterized by a complex series of factors that negatively affect hydrobionts and lead to declines in their populations. However, in a number of cases, a range of rapid adaptive changes ensure the resilience of hydrobiont populations. Here, we present both field and experimental data shedding light on the physiological basis of adaptation to heavy metal contamination in populations of Dolly Varden charr (Salvelinus malma) differing in duration of isolation in volcanic streams. The study reveals that isolated populations have a physiological phenotype that distinguishes them from populations inhabiting clean waters. They are characterized by a hyperthyroid status accompanied by an increased metabolic rate, elevated activity of antioxidant enzymes, decreased ionic conductivity of tissues and reduced stored energy reserves. Our experimental data reveal that hyperthyroidism is an adaptive characteristic enhancing the resistance to heavy metal contamination and shaping the evolution of these populations. The similarity of physiological, developmental and morphological changes in isolated populations suggests a common source and mechanisms underpinning this case of 'evolutionary rescue'. Thus, populations of S. malma trapped in volcanic streams represent a genuine case of rapid endocrine-driven adaptation to changing environmental stimuli.
Collapse
Affiliation(s)
- Evgeny V Esin
- A.N. Severtsov Institute of Ecology and Evolution RAS, Moscow, Russian Federation
| | - Elena V Shulgina
- Russian Federal Research Institute of Fisheries and Oceanography, Moscow, Russian Federation
| | - Fedor N Shkil
- A.N. Severtsov Institute of Ecology and Evolution RAS, Moscow, Russian Federation
- N.K. Koltzov Institute of Developmental Biology, Moscow, Russian Federation
| |
Collapse
|
15
|
He Y, Hou J, Qiu Y, Ouyang K, Li D, Li L. Microcystin-LR immersion caused sequential endocrine disruption and growth inhibition in zebrafish (Danio rerio) from fertilization to sexual differentiation completion. Toxicology 2023:153569. [PMID: 37295766 DOI: 10.1016/j.tox.2023.153569] [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: 04/10/2023] [Revised: 06/03/2023] [Accepted: 06/06/2023] [Indexed: 06/12/2023]
Abstract
Microcystin-LR (MC-LR) is a highly toxic congener and is also one of the most commonly found. Recent studies have demonstrated that MC-LR can disrupt growth and endocrine in fish, but how it works at the stage of the sex differentiation period had not been determined to date. In this study, zebrafish (Danio rerio) embryos were exposed to MC-LR (0 and 10μg/L), and sampled at 14, 28, and 42 days post fertilization (dpf), respectively. The results demonstrated that MC-LR caused the growth inhibition of zebrafish at 42 dpf. The expression levels of genes related to the growth hormone/insulin-like growth factor (GH/IGF) and hypothalamic-pituitary-thyroid (HPT) axes, as well as the levels of hormone 3,5,3'- Triiodothyronine (T3) and thyroxine (T4), were significantly decreased at all time points. A Significant decrease in the ratio of testosterone and estradiol (T/E2) were detected at 28 and 42 dpf in MC-LR group along with changes in genes related to the hypothalamic-pituitary-gonadal (HPG) axis. The result of sex ratio showed that the percentage of females was up to 61.84%, indicating a estrogenic effect induced by MC-LR. The significant changes on hormone levels and gene transcripts occurred mainly in the stage of sex differentiation. The correlation analysis further suggested that key cross-talks among three endocrine axes may be the growth hormone releasing hormone (GHRH), Transthyretin (TTR) and gonadotropin releasing hormone (GnRH) signaling molecules. Overall, our findings provide a new insight for understanding the mechanisms by which MC-LR affects fish growth and reproduction during gonadal development.
Collapse
Affiliation(s)
- Ya He
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Jie Hou
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Yuming Qiu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Kang Ouyang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Dapeng Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, P.R. China; Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, P.R. China; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, P.R. China; Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, P.R. China
| | - Li Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, P.R. China; Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, P.R. China; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, P.R. China; Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, P.R. China.
| |
Collapse
|
16
|
Yang H, Fang B, Wang Z, Chen Y, Dong Y. The Timing Sequence and Mechanism of Aging in Endocrine Organs. Cells 2023; 12:cells12070982. [PMID: 37048056 PMCID: PMC10093290 DOI: 10.3390/cells12070982] [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: 01/25/2023] [Revised: 03/15/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
The world is increasingly aging, and there is an urgent need to find a safe and effective way to delay the aging of the body. It is well known that the endocrine glands are one of the most important organs in the context of aging. Failure of the endocrine glands lead to an abnormal hormonal environment, which in turn leads to many age-related diseases. The aging of endocrine glands is closely linked to oxidative stress, cellular autophagy, genetic damage, and hormone secretion. The first endocrine organ to undergo aging is the pineal gland, at around 6 years old. This is followed in order by the hypothalamus, pituitary gland, adrenal glands, gonads, pancreatic islets, and thyroid gland. This paper summarises the endocrine gland aging-related genes and pathways by bioinformatics analysis. In addition, it systematically summarises the changes in the structure and function of aging endocrine glands as well as the mechanisms of aging. This study will advance research in the field of aging and help in the intervention of age-related diseases.
Collapse
Affiliation(s)
- He Yang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, Ministry of Education, China Agricultural University, Beijing 100193, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, Ministry of Education, China Agricultural University, Beijing 100193, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Zixu Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yaoxing Chen
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yulan Dong
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, Ministry of Education, China Agricultural University, Beijing 100193, China
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
17
|
Tanizaki Y, Bao L, Shi YB. Steroid-receptor coactivator complexes in thyroid hormone-regulation of Xenopus metamorphosis. VITAMINS AND HORMONES 2023; 123:483-502. [PMID: 37717995 PMCID: PMC11274430 DOI: 10.1016/bs.vh.2023.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Anuran metamorphosis is perhaps the most drastic developmental change regulated by thyroid hormone (T3) in vertebrate. It mimics the postembryonic development in mammals when many organs/tissues mature into adult forms and plasma T3 level peaks. T3 functions by regulating target gene transcription through T3 receptors (TRs), which can recruit corepressor or coactivator complexes to target genes in the absence or presence of T3, respectively. By using molecular and genetic approaches, we and others have investigated the role of corepressor or coactivator complexes in TR function during the development of two highly related anuran species, the pseudo-tetraploid Xenopus laevis and diploid Xenopus tropicalis. Here we will review some of these studies that demonstrate a critical role of coactivator complexes, particularly those containing steroid receptor coactivator (SRC) 3, in regulating metamorphic rate and ensuring the completion of metamorphosis.
Collapse
Affiliation(s)
- Yuta Tanizaki
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Lingyu Bao
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, United States.
| |
Collapse
|
18
|
Self-Similar Patterns from Abiotic Decarboxylation Metabolism through Chemically Oscillating Reactions: A Prebiotic Model for the Origin of Life. Life (Basel) 2023; 13:life13020551. [PMID: 36836908 PMCID: PMC9960873 DOI: 10.3390/life13020551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 02/03/2023] [Accepted: 02/11/2023] [Indexed: 02/18/2023] Open
Abstract
The origin of life must have included an abiotic stage of carbon redox reactions that involved electron transport chains and the production of lifelike patterns. Chemically oscillating reactions (COR) are abiotic, spontaneous, out-of-equilibrium, and redox reactions that involve the decarboxylation of carboxylic acids with strong oxidants and strong acids to produce CO2 and characteristic self-similar patterns. Those patterns have circular concentricity, radial geometries, characteristic circular twins, colour gradients, cavity structures, and branching to parallel alignment. We propose that COR played a role during the prebiotic cycling of carboxylic acids, furthering the new model for geology where COR can also explain the patterns of diagenetic spheroids in sediments. The patterns of COR in Petri dishes are first considered and compared to those observed in some eukaryotic lifeforms. The molecular structures and functions of reactants in COR are then compared to key biological metabolic processes. We conclude that the newly recognised similarities in compositions and patterns warrant future research to better investigate the role of halogens in biochemistry; COR in life-forms, including in humans; and the COR-stage of prebiotic carbon cycling on other planets, such as Mars.
Collapse
|
19
|
Tanizaki Y, Zhang H, Shibata Y, Shi YB. Organ-specific effects on target binding due to knockout of thyroid hormone receptor α during Xenopus metamorphosis. Dev Growth Differ 2023; 65:23-28. [PMID: 36397722 DOI: 10.1111/dgd.12825] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 11/02/2022] [Accepted: 11/02/2022] [Indexed: 11/19/2022]
Abstract
Thyroid hormone (T3) is essential for normal development and metabolism, especially during postembryonic development, a period around birth in mammals when plasma T3 levels reach their peak. T3 functions through two T3 receptors, TRα and TRβ. However, little is known about the tissue-specific functions of TRs during postembryonic development because of maternal influence and difficulty in manipulation of mammalian models. We have studied Xenopus tropicalis metamorphosis as a model for human postembryonic development. By using TRα knockout (Xtr·thratmshi ) tadpoles, we have previously shown that TRα is important for T3-dependent intestinal remodeling and hindlimb development but not tail resorption during metamorphosis. Here, we have identified genes bound by TR in premetamorphic wild-type and Xtr·thratmshi tails with or without T3 treatment by using chromatin immunoprecipitation-sequencing and compared them with those in the intestine and hindlimb. Compared to other organs, the tail has much fewer genes bound by TR or affected by TRα knockout. Bioinformatic analyses revealed that among the genes bound by TR in wild-type but not Xtr·thratmshi organs, fewer gene ontology (GO) terms or biological pathways related to metamorphosis were enriched in the tail compared to those in the intestine and hindlimb. This difference likely underlies the drastic effects of TRα knockout on the metamorphosis of the intestine and hindlimb but not the tail. Thus, TRα has tissue-specific roles in regulating T3-dependent anuran metamorphosis by directly targeting the pathways and GO terms important for metamorphosis.
Collapse
Affiliation(s)
- Yuta Tanizaki
- Section on Molecular Morphogenesis, Cell Regulation and Development Affinity Group, Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Hongen Zhang
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Yuki Shibata
- Section on Molecular Morphogenesis, Cell Regulation and Development Affinity Group, Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, Cell Regulation and Development Affinity Group, Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
20
|
Kotsopoulou I, Vyas AK, Cory MJ, Chan CS, Jagarapu J, Gill S, Mudduluru M, Angelis D. Developmental changes of the fetal and neonatal thyroid gland and functional consequences on the cardiovascular system. J Perinatol 2022; 42:1576-1586. [PMID: 36376450 DOI: 10.1038/s41372-022-01559-3] [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: 08/11/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022]
Abstract
Thyroid hormones play an important role in the development and function of the cardiac myocyte. Dysregulation of the thyroid hormone milieu affects the fetal cardiac cells via complex molecular mechanisms, either by altering gene expression or directly by affecting post-translational processes. This review offers a comprehensive summary of the effects of thyroid hormones on the developing cardiovascular system and its adaptation. Furthermore, we will highlight the gaps in knowledge and provide suggestions for future research.
Collapse
Affiliation(s)
- Ioanna Kotsopoulou
- Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Arpita K Vyas
- Division of Pediatrics and Endocrinology, College of Medicine, California Northstate University, Elk Grove, CA, USA
| | - Melinda J Cory
- Division of Cardiology, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christina S Chan
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jawahar Jagarapu
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shamaila Gill
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Manjula Mudduluru
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dimitrios Angelis
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
21
|
Zuñiga LFF, Muñoz YS, Pustovrh MC. Thyroid hormones: Metabolism and transportation in the fetoplacental unit. Mol Reprod Dev 2022; 89:526-539. [PMID: 36208482 DOI: 10.1002/mrd.23647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 09/20/2022] [Accepted: 09/25/2022] [Indexed: 12/25/2022]
Abstract
The thyroid hormones (THs), thyroxine (T4) and triiodothyronine (T3), are of vital importance for fetal development. The concentration of THs in fetal circulation varies throughout gestation and differs from the concentration in the maternal serum, indicating the presence of maternal-fetal thyroid homeostasis regulatory mechanisms in the placenta. The passage of THs from maternal circulation to fetal circulation is modulated by plasma membrane transporters, enzymes, and carrier proteins. Monocarboxylate transporter 8, iodothyronine deiodinases (DIO2 and DIO3), and transthyretin are especially involved in this maternal-fetal thyroid modulation, shown by a greater expression in the placenta. THs also play a role in placental development and as expected, abnormal variations in TH levels are associated with pregnancy complications and can result in damage to the fetus. Although new evidence regarding TH regulation during pregnancy and its effects in the mother, placenta, and fetus has been published, many aspects of these interactions are still poorly understood. The objective of this review is to provide an evidence-based update, drawn from current data, on the metabolism and transport of THs in the placenta and their vital role in the maternal-fetal relationship.
Collapse
Affiliation(s)
- Luis Felipe Falla Zuñiga
- Department of Morphology, College of Basic Sciences, Faculty of Health, Universidad del Valle, Cali, Colombia
| | - Yhoiss Smiht Muñoz
- Department of Morphology, College of Basic Sciences, Faculty of Health, Universidad del Valle, Cali, Colombia
| | - Maria Carolina Pustovrh
- Department of Morphology, College of Basic Sciences, Faculty of Health, Universidad del Valle, Cali, Colombia
| |
Collapse
|
22
|
Association of TSHR gene single nucleotide intronic polymorphism with the risk of hypothyroid and hyperthyroid disorders in Yazd province. Sci Rep 2022; 12:15745. [PMID: 36130976 PMCID: PMC9492782 DOI: 10.1038/s41598-022-19822-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
The present study was carried out, for the first time, to evaluate the association of rs2268458 polymorphism, biochemical and environmental factors on hypothyroid and hyperthyroid disorders in thyroid patients and healthy individuals in Yazd province, Iran. In this study, blood samples were collected from a total of 100 cases, including 60 hypothyroid, 20 hyperthyroid and 20 normal individuals. DNA was extracted from blood samples and the rs2268458 single nucleotide intronic polymorphism was evaluated using Restriction Fragment Length Polymorphism PCR (RFLP-PCR). The results have shown that 59 individuals were homozygote (TT), 40 cases were heterozygote (TC) and one homozygote (CC) case. Of 59 TT homozygote cases, 25 cases were hypothyroid females and 7 hypothyroid male patients. While, heterozygote TC group consisted of 20 hypothyroid females and 7 hypothyroid male cases. Furthermore, only 1 (CC) homozygote male hypothyroid patient was observed in this study. The hyperthyroid population consisted of 7 (TT) homozygote hyperthyroid female cases, 8 (TC) heterozygote hyperthyroid female cases, 3 (TT) homozygote hyperthyroid male cases and 2 (TC) heterozygote hyperthyroid male cases. According to our study, heterozygote cases (TC) showed less severe symptoms, while homozygote cases (TT) showed no serious symptoms and the (CC) homozygote case showed severe thyroid abnormalities. So, it can be concluded that the TSHR-related rs2268458 polymorphism is associated with hypothyroidism and hyperthyroidism in the male and female populations of Yazd Province, Iran and C allele can be a risk factor for some physio-biochemical and hormonal imbalance in the thyroid disorder patients.
Collapse
|
23
|
Chartoumpekis DV, Ziros PG, Habeos IG, Sykiotis GP. Emerging roles of Keap1/Nrf2 signaling in the thyroid gland and perspectives for bench-to-bedside translation. Free Radic Biol Med 2022; 190:276-283. [PMID: 35988853 DOI: 10.1016/j.freeradbiomed.2022.08.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/11/2022] [Accepted: 08/14/2022] [Indexed: 12/14/2022]
Abstract
The signaling pathway centered on the transcription factor nuclear erythroid factor 2-like 2 (Nrf2) has emerged during the last 15 years as a target for the prevention and treatment of diseases broadly related with oxidative stress such as cancer, neurodegenerative and metabolic diseases. The roles of Nrf2 are expanding beyond general cytoprotection, and they encompass its crosstalk with other pathways as well as tissue-specific functions. The thyroid gland relies on reactive oxygen species for its main physiological function, the synthesis and secretion of thyroid hormones. A few years ago, Nrf2 was characterized as a central regulator of the antioxidant response in the thyroid, as well as of the transcription and processing of thyroglobulin, the major thyroidal protein that serves as the substrate for thyroid hormone synthesis. Herein, we summarize the current knowledge about the roles of Nrf2 in thyroid physiology, pathophysiology and disease. We focus specifically on the most recent publications in the field, and we discuss the implications for the preclinical and clinical use of Nrf2 modulators.
Collapse
Affiliation(s)
- Dionysios V Chartoumpekis
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, CH-1011, Lausanne, Switzerland
| | - Panos G Ziros
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, CH-1011, Lausanne, Switzerland
| | - Ioannis G Habeos
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, GR-26504, Patras, Greece
| | - Gerasimos P Sykiotis
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, CH-1011, Lausanne, Switzerland.
| |
Collapse
|
24
|
Endocrine Diagnostics for Exotic Animals. Vet Clin North Am Exot Anim Pract 2022; 25:631-661. [PMID: 36122944 DOI: 10.1016/j.cvex.2022.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endocrine disease in exotic species is less common than in small animals. Nevertheless, the diagnostic principles used in small animals can be adapted to evaluate endocrine disease in many of the exotic species although species-specific aspects need to be considered. This article covers important diseases such as thyroid dysfunction in reptiles and birds, hyperthyroidism in guinea pigs, and hyperadrenocorticism in ferrets. Glucose metabolism in neoplasms affecting normal physiology, such as insulinoma in ferrets and gastric neuroendocrine carcinoma in bearded dragons, is discussed. Calcium abnormalities, including metabolic bone disease in reptiles and hypocalcemia in birds, are also covered.
Collapse
|
25
|
Zorrilla Veloz RI, McKenzie T, Palacios BE, Hu J. Nuclear hormone receptors in demyelinating diseases. J Neuroendocrinol 2022; 34:e13171. [PMID: 35734821 PMCID: PMC9339486 DOI: 10.1111/jne.13171] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/20/2022] [Accepted: 05/27/2022] [Indexed: 11/28/2022]
Abstract
Demyelination results from the pathological loss of myelin and is a hallmark of many neurodegenerative diseases. Despite the prevalence of demyelinating diseases, there are no disease modifying therapies that prevent the loss of myelin or promote remyelination. This review aims to summarize studies in the field that highlight the importance of nuclear hormone receptors in the promotion and maintenance of myelination and the relevance of nuclear hormone receptors as potential therapeutic targets for demyelinating diseases. These nuclear hormone receptors include the estrogen receptor, progesterone receptor, androgen receptor, vitamin D receptor, thyroid hormone receptor, peroxisome proliferator-activated receptor, liver X receptor, and retinoid X receptor. Pre-clinical studies in well-established animal models of demyelination have shown a prominent role of these nuclear hormone receptors in myelination through their promotion of oligodendrocyte maturation and development. The activation of the nuclear hormone receptors by their ligands also promotes the synthesis of myelin proteins and lipids in mouse models of demyelination. There are limited clinical studies that focus on how the activation of these nuclear hormone receptors could alleviate demyelination in patients with diseases such as multiple sclerosis (MS). However, the completed clinical trials have reported improved clinical outcome in MS patients treated with the ligands of some of these nuclear hormone receptors. Together, the positive results from both clinical and pre-clinical studies point to nuclear hormone receptors as promising therapeutic targets to counter demyelination.
Collapse
Affiliation(s)
- Rocío I Zorrilla Veloz
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Takese McKenzie
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Bridgitte E Palacios
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Jian Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
26
|
Petrovic Jurcevic J, Jurcevic M, Jagic M, Jazbec A, Mandic K, Juri Mandic J. “Influence of Clinically Active Graves’ Ophthalmopathy on Spherical Equivalent and Visual Acuity”. Clin Ophthalmol 2022; 16:2353-2361. [PMID: 35924183 PMCID: PMC9342880 DOI: 10.2147/opth.s369677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Clinical experience regarding the fluctuations of the refractive error of the eye during the different stages of Graves’ ophthalmopathy observed through outpatient clinic frequent check-ups points towards an underestimated and often overlooked problem. Published data about it are sparse. The clinical manifestations of Graves’ ophthalmopathy can be understood from the perspective of “compartment syndrome” and literature implies how such changes can affect the refractive error and consequently, the visual acuity. The purpose of the study was to explore how the clinical activity score of Graves’ ophthalmopathy affects refractive error and visual acuity. Patients and Methods The study was prospective and observational, including 60 eyes of 30 patients with clinically active Graves’ ophthalmopathy. All the patients were monitored and evaluated over a period of 36 months by the clinical activity score, spherical equivalent and visual acuity. All the observed parameters were statistically analyzed. Results The mean values of spherical equivalent and visual acuity throughout the observed period showed continuous fluctuation. Repeated measure analysis of variance showed statistically significant differences in visual acuity and spherical equivalent over the observed period. There was a statistically significant positive correlation between visual acuity and clinical activity score. The correlation between spherical equivalent and clinical activity score was also positive but not statistically significant. Conclusion A decrease in the clinical activity score is either the result of a spontaneously resolving course of Graves’ ophthalmopathy or a consequence of treatment, so lowering in fluctuation of refractive error and improved visual acuity may be associated with a reduction in orbital inflammation.
Collapse
Affiliation(s)
- Jasenka Petrovic Jurcevic
- Medical School University of Zagreb, PhD Candidate on the Programme “Biomedicine and Health”, Zagreb, Croatia
- Correspondence: Jasenka Petrovic Jurcevic, Medical School University of Zagreb, Šalata 4a, Zagreb, Croatia, Email
| | - Marko Jurcevic
- University of Zagreb Faculty of Electrical Engineering and Computing, Department of Electrical Engineering Fundamentals and Measurements, Zagreb, Croatia
| | - Mateja Jagic
- Special Hospital „Svjetlost“, Department of Refractive Surgery, Zagreb, Croatia
| | - Anamarija Jazbec
- University of Zagreb Faculty of Forestry and Wood Technology, Department for Forest Inventory and Management, Zagreb, HR-10000, Croatia
| | - Kresimir Mandic
- Clinical Department of Ophthalmology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Jelena Juri Mandic
- Clinical Department of Ophthalmology, University Hospital Center Zagreb, Zagreb, Croatia
| |
Collapse
|
27
|
Li M, Lan F, Li C, Li N, Chen X, Zhong Y, Yang Y, Shao Y, Kong Y, Li X, Wu D, Zhang J, Chen W, Li Z, Zhu X. Expression and Regulation Network of HDAC3 in Acute Myeloid Leukemia and the Implication for Targeted Therapy Based on Multidataset Data Mining. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:4703524. [PMID: 35371279 PMCID: PMC8966751 DOI: 10.1155/2022/4703524] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/15/2022] [Indexed: 02/07/2023]
Abstract
Background Histone deacetylase 3 (HDAC3) plays an important role in the development and progression of a variety of cancers, but its regulatory mechanism in acute myeloid leukemia (LAML) is not entirely understood. Methods We analyzed the expression of HDAC3 in normal and cancerous tissues using Oncomine, UALCAN, and GEO databases. Changes of the HDAC3 gene were analyzed by cBioPortal. The genes coexpressed with HDAC3 were analyzed by WebGestalt, and the predicted signaling pathways in KEGG were discussed. Results We discovered that the expression of HDAC3 was elevated in some types of acute myeloid leukemia. The HDAC3 gene has a strong positive correlation with SLC25A5, NDUFA2, Cox4I1, and EIF3K, which regulate cell growth and development. HDAC3 transcription is higher in patients with FLT3 mutation than in healthy people. HDAC3 can be directly involved in regulating the thyroid hormone signaling pathway. MEF2D is directly involved in the cGMP-PKG signaling pathway, and the HDAC3 gene has a strong synergistic relationship with MEF2D. HDAC3 is indirectly involved in the cGMP-PKG signaling pathway, thereby indirectly regulating the expression levels of p53 and p21 genes in patients with LAML. Genomics of Drug Sensitivity in Cancer (GDSC) database analysis revealed that the application of the HDAC3 inhibitor can inhibit the proliferation of leukemia cells. Conclusions Therefore, our data suggest that HDAC3 may be a possible therapeutic target for acute myeloid leukemia.
Collapse
Affiliation(s)
- Minhua Li
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, China
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Feifei Lan
- Medical Genetics Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Chen Li
- Department of Biology, Chemistry, Pharmacy, Free University of Berlin, 14195 Berlin, Germany
| | - Ning Li
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Xiaojie Chen
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Yueyuan Zhong
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Yue Yang
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Yingqi Shao
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Yi Kong
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Xinming Li
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Danny Wu
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Jingyu Zhang
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Wenqing Chen
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Zesong Li
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China
| | - Xiao Zhu
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, China
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
28
|
Das D, Banerjee A, Jena AB, Duttaroy AK, Pathak S. Essentiality, relevance, and efficacy of adjuvant/combinational therapy in the management of thyroid dysfunctions. Biomed Pharmacother 2022; 146:112613. [PMID: 35062076 DOI: 10.1016/j.biopha.2022.112613] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/14/2021] [Accepted: 01/02/2022] [Indexed: 11/02/2022] Open
Abstract
Thyroid dysfunction is the most prevalent endocrine disorder worldwide having an epidemiology of 11% in Indians, 4.6% in the United Kingdom, and 2% in the United States of America among the overall population. The common thyroid disorders include hypothyroidism, hyperthyroidism, Hashimoto's thyroiditis, and thyroid cancer. This review briefly elaborates the molecular regulation and mechanism of thyroid hormone, and its associated thyroid disorders. The thyroid hormones regulate critical biochemical functions in brain development and function. Hypothyroidism is mainly associated with dysregulation of cytokines, increased ROS production, and altered signal transduction in major regions of the brain. In addition, it is associated with reduced antioxidant capacity and increased oxidative stress in humans. Though 70% of thyroid disorders are caused by heredity, environmental factors have a significant influence in developing autoimmune thyroid disorders in people who are predisposed to them. This drives us to understand the relationship between environmental factors and thyroid dysregulated disorders. The treatment option for the thyroid disorder includes antithyroid medications, receiving radioactive iodine therapy, or surgery at a critical stage. However, antithyroid drugs are not typically used long-term in thyroid disease due to the high recurrence rate. Adjuvant treatment of antioxidants can produce better outcomes with anti-thyroid drug treatment. Thus, Adjuvant therapy has been proven as an effective strategy for managing thyroid dysfunction, herbal remedies can be used to treat thyroid dysfunction in the future, which in turn can reduce the prevalence of thyroid disorders.
Collapse
Affiliation(s)
- Diptimayee Das
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | | | - Asim K Duttaroy
- Department of Nutrition, Institute of Medical Sciences, Faculty of Medicine, University of Oslo, Norway.
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India.
| |
Collapse
|
29
|
Carvalho DP, Dias AF, Sferruzzi-Perri AN, Ortiga-Carvalho TM. Gaps in the knowledge of thyroid hormones and placental biology. Biol Reprod 2022; 106:1033-1048. [DOI: 10.1093/biolre/ioac006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
Thyroid hormones (THs) are required for the growth and development of the foetus, stimulating anabolism and oxygen consumption from the early stages of pregnancy to the period of foetal differentiation close to delivery. Maternal changes in the hypothalamic–pituitary thyroid axis are also well known. In contrast, several open questions remain regarding the relationships between the placenta and the maternal and foetal TH systems. The exact mechanism by which the placenta participates in regulating the TH concentration in the foetus and mother and the role of TH in the placenta are still poorly studied. In this review, we aim to summarize the available data in the area and highlight significant gaps in our understanding of the ontogeny and cell-specific localization of TH transporters, TH receptors and TH metabolic enzymes in the placenta in both human and rodent models. Significant deficiencies also exist in knowledge of the contribution of genomic and nongenomic effects of TH on the placenta and finally how the placenta reacts during pregnancy when the mother has thyroid disease. By addressing these key knowledge gaps, improved pregnancy outcomes and management of women with thyroid alterations may be possible.
Collapse
Affiliation(s)
- Daniela Pereira Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Endocrinologia Translacional, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ariane Fontes Dias
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Endocrinologia Translacional, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amanda Nancy Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Tania Maria Ortiga-Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Endocrinologia Translacional, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
30
|
Tanizaki Y, Shibata Y, Zhang H, Shi YB. Thyroid Hormone Receptor α Controls the Hind Limb Metamorphosis by Regulating Cell Proliferation and Wnt Signaling Pathways in Xenopus tropicalis. Int J Mol Sci 2022; 23:ijms23031223. [PMID: 35163147 PMCID: PMC8835992 DOI: 10.3390/ijms23031223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/17/2022] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
Thyroid hormone (T3) receptors (TRs) mediate T3 effects on vertebrate development. We have studied Xenopus tropicalis metamorphosis as a model for postembryonic human development and demonstrated that TRα knockout induces precocious hind limb development. To reveal the molecular pathways regulated by TRα during limb development, we performed chromatin immunoprecipitation- and RNA-sequencing on the hind limb of premetamorphic wild type and TRα knockout tadpoles, and identified over 700 TR-bound genes upregulated by T3 treatment in wild type but not TRα knockout tadpoles. Interestingly, most of these genes were expressed at higher levels in the hind limb of premetamorphic TRα knockout tadpoles than stage-matched wild-type tadpoles, suggesting their derepression upon TRα knockout. Bioinformatic analyses revealed that these genes were highly enriched with cell cycle and Wingless/Integrated (Wnt) signaling-related genes. Furthermore, cell cycle and Wnt signaling pathways were also highly enriched among genes bound by TR in wild type but not TRα knockout hind limb. These findings suggest that direct binding of TRα to target genes related to cell cycle and Wnt pathways is important for limb development: first preventing precocious hind limb formation by repressing these pathways as unliganded TR before metamorphosis and later promoting hind limb development during metamorphosis by mediating T3 activation of these pathways.
Collapse
Affiliation(s)
- Yuta Tanizaki
- Section on Molecular Morphogenesis, Cell Regulation and Development Affinity Group, Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA; (Y.T.); (Y.S.)
| | - Yuki Shibata
- Section on Molecular Morphogenesis, Cell Regulation and Development Affinity Group, Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA; (Y.T.); (Y.S.)
- Center for the Development of New Model Organisms, National Institute for Basic Biology, National Institute of Natural Sciences, Okazaki 444-8585, Aichi, Japan
| | - Hongen Zhang
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA;
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, Cell Regulation and Development Affinity Group, Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA; (Y.T.); (Y.S.)
- Correspondence:
| |
Collapse
|
31
|
Lee KW, Shin Y, Lee S, Lee S. Inherited Disorders of Thyroid Hormone Metabolism Defect Caused by the Dysregulation of Selenoprotein Expression. Front Endocrinol (Lausanne) 2022; 12:803024. [PMID: 35126314 PMCID: PMC8807339 DOI: 10.3389/fendo.2021.803024] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/20/2021] [Indexed: 11/25/2022] Open
Abstract
Consistent activation and functioning of thyroid hormones are essential to the human body as a whole, especially in controlling the metabolic rate of all organs and systems. Impaired sensitivity to thyroid hormones describes any process that interferes with the effectiveness of thyroid hormones. The genetic origin of inherited thyroid hormone defects and the investigation of genetic defects upon the processing of thyroid hormones are of utmost importance. Impaired sensitivity to thyroid hormone can be categorized into three conditions: thyroid hormone cell membrane transport defect (THCMTD), thyroid hormone metabolism defect (THMD), and thyroid hormone action defect (THAD). THMD is caused by defects in the synthesis and processing of deiodinases that convert the prohormone thyroxine (T4) to the active hormone triiodothyronine (T3). Deiodinase, a selenoprotein, requires unique translation machinery that is collectively composed of the selenocysteine (Sec) insertion sequence (SECIS) elements, Sec-insertion sequence-binding protein 2 (SECISBP2), Sec-specific eukaryotic elongation factor (EEFSEC), and Sec-specific tRNA (TRU-TCA1-1), which leads to the recognition of the UGA codon as a Sec codon for translation into the growing polypeptide. In addition, THMD could be expanded to the defects of enzymes that are involved in thyroid hormone conjugation, such as glucuronidation and sulphation. Paucity of inherited disorders in this category leaves them beyond the scope of this review. This review attempts to specifically explore the genomic causes and effects that result in a significant deficiency of T3 hormones due to inadequate function of deiodinases. Moreover, along with SECISBP2, TRU-TCA1-1, and deiodinase type-1 (DIO1) mutations, this review describes the variants in DIO2 single nucleotide polymorphism (SNP) and thyroid stimulating hormone receptor (TSHR) that result in the reduced activity of DIO2 and subsequent abnormal conversion of T3 from T4. Finally, this review provides additional insight into the general functionality of selenium supplementation and T3/T4 combination treatment in patients with hypothyroidism, suggesting the steps that need to be taken in the future.
Collapse
Affiliation(s)
- Kyu Won Lee
- Department of Food Science and Engineering, Ewha Womans University, Seoul, South Korea
| | - Yoochan Shin
- Laboratory of Genomics and Translational Medicine, Department of Internal Medicine, Gachon University College of Medicine, Incheon, South Korea
| | - Sungahn Lee
- Laboratory of Genomics and Translational Medicine, Department of Internal Medicine, Gachon University College of Medicine, Incheon, South Korea
| | - Sihoon Lee
- Laboratory of Genomics and Translational Medicine, Department of Internal Medicine, Gachon University College of Medicine, Incheon, South Korea
| |
Collapse
|
32
|
Kővári D, Penksza V, Szilvásy-Szabó A, Sinkó R, Gereben B, Mackie K, Fekete C. Tanycyte specific ablation of diacylglycerol lipase alpha stimulates the hypothalamic-pituitary-thyroid axis by decreasing the endocannabinoid mediated inhibition of TRH release. J Neuroendocrinol 2022; 34:e13079. [PMID: 34970803 DOI: 10.1111/jne.13079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/31/2022]
Abstract
In addition to the hypophysiotropic thyrotropin-releasing hormone (TRH)-synthesizing neurons, a glial cell type, the tanycytes, also play a role in the regulation of the hypothalamic-pituitary-thyroid (HPT) axis. Tanycytes modulate the feedback regulation of the axis by regulating the local thyroid hormone availability in the median eminence where the hypophysiotropic axons terminate. Recently, we showed that tanycytes produce diacylglycerol lipase alpha (DAGLα), the synthesizing enzyme of the endocannabinoid 2-arachidonoylglycerol (2-AG) that inhibits the release of TRH from the hypophysiotropic terminals in median eminence explants. To determine the importance of the endocannabinoid production of tanycytes, adult male Rax-CreERT2//DAGLαfl/fl mice were treated with tamoxifen to induce a tanycyte specific decrease of DAGLα expression (T-DAGLα KO). The effect of this genetic manipulation on the activity of the HPT axis was determined. Tanycyte specific decrease of DAGLα expression resulted in an approximately 2-fold increase of TSHβ mRNA level that was accompanied by increased levels of circulating free T4. The TRH mRNA level was, however, not influenced by the genetic manipulation. In addition to the effects on the HPT axis, the T-DAGLα KO mice showed increased fat mass ratio and decreased blood glucose levels. These data indicate that when endocannabinoid release of tanycytes is decreased, the disinhibition of the TRH release induces increased TSH synthesis and higher circulating T4 levels. Thus it suggests that in wild-type mice, tanycytes exert a tonic inhibitory effect on the TRH release of hypophysiotropic axons. Furthermore, the endocannabinoid release of tanycytes also influences glucose homeostasis and fat deposition.
Collapse
Affiliation(s)
- Dóra Kővári
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Neuroendocrinology Program, Semmelweis University, Budapest, Hungary
| | - Veronika Penksza
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Budapest, Hungary
| | - Anett Szilvásy-Szabó
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Budapest, Hungary
| | - Richárd Sinkó
- János Szentágothai Doctoral School of Neurosciences, Neuroendocrinology Program, Semmelweis University, Budapest, Hungary
- Laboratory of Molecular Cell Metabolism, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Gereben
- Laboratory of Molecular Cell Metabolism, Institute of Experimental Medicine, Budapest, Hungary
| | - Ken Mackie
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA
| | - Csaba Fekete
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
33
|
Girgis J, Yang D, Chakroun I, Liu Y, Blais A. Six1 promotes skeletal muscle thyroid hormone response through regulation of the MCT10 transporter. Skelet Muscle 2021; 11:26. [PMID: 34809717 PMCID: PMC8607597 DOI: 10.1186/s13395-021-00281-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/29/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The Six1 transcription factor is implicated in controlling the development of several tissue types, notably skeletal muscle. Six1 also contributes to muscle metabolism and its activity is associated with the fast-twitch, glycolytic phenotype. Six1 regulates the expression of certain genes of the fast muscle program by directly stimulating their transcription or indirectly acting through a long non-coding RNA. We hypothesized that additional mechanisms of action of Six1 might be at play. METHODS A combined analysis of gene expression profiling and genome-wide location analysis data was performed. Results were validated using in vivo RNA interference loss-of-function assays followed by measurement of gene expression by RT-PCR and transcriptional reporter assays. RESULTS The Slc16a10 gene, encoding the thyroid hormone transmembrane transporter MCT10, was identified as a gene with a transcriptional enhancer directly bound by Six1 and requiring Six1 activity for full expression in adult mouse tibialis anterior, a predominantly fast-twitch muscle. Of the various thyroid hormone transporters, MCT10 mRNA was found to be the most abundant in skeletal muscle, and to have a stronger expression in fast-twitch compared to slow-twitch muscle groups. Loss-of-function of MCT10 in the tibialis anterior recapitulated the effect of Six1 on the expression of fast-twitch muscle genes and led to lower activity of a thyroid hormone receptor-dependent reporter gene. CONCLUSIONS These results shed light on the molecular mechanisms controlling the tissue expression profile of MCT10 and identify modulation of the thyroid hormone signaling pathway as an additional mechanism by which Six1 influences skeletal muscle metabolism.
Collapse
Affiliation(s)
- John Girgis
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, K1H 8M5, Canada.,Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada.,Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Dabo Yang
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, K1H 8M5, Canada.,Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada
| | - Imane Chakroun
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, K1H 8M5, Canada.,Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada
| | - Yubing Liu
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, K1H 8M5, Canada.,Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada
| | - Alexandre Blais
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, K1H 8M5, Canada. .,Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada. .,University of Ottawa Centre for Inflammation, Immunity and Infection (CI3), Ottawa, Ontario, Canada.
| |
Collapse
|
34
|
Corinti D, Chiavarino B, Spano M, Tintaru A, Fornarini S, Crestoni ME. Molecular Basis for the Remarkably Different Gas-Phase Behavior of Deprotonated Thyroid Hormones Triiodothyronine (T3) and Reverse Triiodothyronine (rT3): A Clue for Their Discrimination? Anal Chem 2021; 93:14869-14877. [PMID: 34714056 PMCID: PMC8581966 DOI: 10.1021/acs.analchem.1c03892] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Thyroid hormones
are biologically active small molecules responsible
for growth and development regulation, basal metabolic rate, and lipid
and carbohydrate metabolism. Liquid chromatography mass spectrometry
(LC–MS) can be used to quantify thyroid hormones blood level
with high speed and selectivity, aiming to improve the diagnosis and
treatment of the severe pathological conditions in which they are
implicated, i.e., hypo- and hyperthyroidism. In this work, the gas-phase
behavior of the isomeric thyroid hormones triiodothyronine (T3) and
reverse triiodothyronine (rT3) in their deprotonated form was studied
at a molecular level using MS-based techniques. Previously reported
collision-induced dissociation experiments yielded distinct spectra
despite the high structural similarity of the two compounds, suggesting
different charge sites to be responsible. Infrared multiple photon
dissociation spectroscopy on [T3-H]− and [rT3-H]− was performed, and the results were interpreted using
DFT and MP2 calculations, assessing the prevalence of T3 in the carboxylate
form and rT3 as a phenolate isomer. The different deprotonation sites
of the two isomers were also found to drive their ion-mobility behavior.
In fact, [T3-H]− and [rT3-H]− were
successfully separated. Drift times were correlated with collisional
cross section values of 209 and 215 Å2 for [T3-H]− and [rT3-H]−, respectively. Calculations
suggested the charge site to be the main parameter involved in the
different mobilities of the two anions. Finally, bare [T3-H]− and [rT3-H]− were made to react with neutral acetylacetone
and trifluoroacetic acid, confirming rT3 to be more acidic than T3
in agreement with the calculated gas-phase acidities of T3 and rT3
equal to 1345 and 1326 kJ mol–1, respectively.
Collapse
Affiliation(s)
- Davide Corinti
- Dipartimento di Chimica e Tecnologie del Farmaco, Università di Roma "La Sapienza", Roma I-00185, Italy
| | - Barbara Chiavarino
- Dipartimento di Chimica e Tecnologie del Farmaco, Università di Roma "La Sapienza", Roma I-00185, Italy
| | - Mattia Spano
- Dipartimento di Chimica e Tecnologie del Farmaco, Università di Roma "La Sapienza", Roma I-00185, Italy
| | - Aura Tintaru
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire, UMR 7273, Marseille 13397, France
| | - Simonetta Fornarini
- Dipartimento di Chimica e Tecnologie del Farmaco, Università di Roma "La Sapienza", Roma I-00185, Italy
| | - Maria Elisa Crestoni
- Dipartimento di Chimica e Tecnologie del Farmaco, Università di Roma "La Sapienza", Roma I-00185, Italy
| |
Collapse
|
35
|
Suzuki Y, Toh L. Constraints and Opportunities for the Evolution of Metamorphic Organisms in a Changing Climate. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.734031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We argue that developmental hormones facilitate the evolution of novel phenotypic innovations and timing of life history events by genetic accommodation. Within an individual’s life cycle, metamorphic hormones respond readily to environmental conditions and alter adult phenotypes. Across generations, the many effects of hormones can bias and at times constrain the evolution of traits during metamorphosis; yet, hormonal systems can overcome constraints through shifts in timing of, and acquisition of tissue specific responses to, endocrine regulation. Because of these actions of hormones, metamorphic hormones can shape the evolution of metamorphic organisms. We present a model called a developmental goblet, which provides a visual representation of how metamorphic organisms might evolve. In addition, because developmental hormones often respond to environmental changes, we discuss how endocrine regulation of postembryonic development may impact how organisms evolve in response to climate change. Thus, we propose that developmental hormones may provide a mechanistic link between climate change and organismal adaptation.
Collapse
|
36
|
Xue L, Bao L, Roediger J, Su Y, Shi B, Shi YB. Protein arginine methyltransferase 1 regulates cell proliferation and differentiation in adult mouse adult intestine. Cell Biosci 2021; 11:113. [PMID: 34158114 PMCID: PMC8220849 DOI: 10.1186/s13578-021-00627-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/10/2021] [Indexed: 01/03/2023] Open
Abstract
Background Adult stem cells play an essential role in adult organ physiology and tissue repair and regeneration. While much has been learnt about the property and function of various adult stem cells, the mechanisms of their development remain poorly understood in mammals. Earlier studies suggest that the formation of adult mouse intestinal stem cells takes place during the first few weeks after birth, the postembryonic period when plasma thyroid hormone (T3) levels are high. Furthermore, deficiency in T3 signaling leads to defects in adult mouse intestine, including reduced cell proliferation in the intestinal crypts, where stem cells reside. Our earlier studies have shown that protein arginine methyltransferase 1 (PRMT1), a T3 receptor coactivator, is highly expressed during intestinal maturation in mouse. Methods We have analyzed the expression of PRMT1 by immunohistochemistry and studied the effect of tissue-specific knockout of PRMT1 in the intestinal epithelium. Results We show that PRMT1 is expressed highly in the proliferating transit amplifying cells and crypt base stem cells. By using a conditional knockout mouse line, we have demonstrated that the expression of PRMT1 in the intestinal epithelium is critical for the development of the adult mouse intestine. Specific removal of PRMT1 in the intestinal epithelium results in, surprisingly, more elongated adult intestinal crypts with increased cell proliferation. In addition, epithelial cell migration along the crypt-villus axis and cell death on the villus are also increased. Furthermore, there are increased Goblet cells and reduced Paneth cells in the crypt while the number of crypt base stem cells remains unchanged. Conclusions Our finding that PRMT1 knockout increases cell proliferation is surprising considering the role of PRMT1 in T3-signaling and the importance of T3 for intestinal development, and suggests that PRMT1 likely regulates pathways in addition to T3-signaling to affect intestinal development and/or homeostasis, thus affecting cell proliferating and epithelial turn over in the adult. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00627-z.
Collapse
Affiliation(s)
- Lu Xue
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, 182 Minyuan Road, Hongshan District, Wuhan, 430074, China.,Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Lingyu Bao
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, No. 277, Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China.,Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Julia Roediger
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Yijun Su
- Laboratory of High Resolution Optical Imaging and Advanced Imaging and Microscopy Resource, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, No. 277, Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA.
| |
Collapse
|
37
|
Wibowo A, Hastuti P, Susanti V. Polymorphism of Thyroid Hormones Receptor, Angiotensin-Converting Enzyme, and High Blood Pressure in Childbearing Age Women with Hyperthyroidism. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
AIM: This study aimed to investigate the association between two polymorphisms of thyroid hormone receptor and renin-angiotensin system (THRA C/T and angiotensin-converting enzyme (ACE) I/D) genes with hypertension in childbearing age women with hypertension.
METHODS: This was a case–control study including 35 cases and 40 matched control subjects. The case group was hyperthyroid women with hypertension while the control group was hyperthyroid women with normotension. The polymorphisms were identified by a classical polymerase chain reaction.
RESULTS: The THRA C/T gene and ACE I/D polymorphisms were not associated with the hypertension while the genotype frequencies in hyperthyroid women with hypertension were as follows: CC genotype was 25.71%, CT genotype was 54.29%, and TT genotype was 20.00%. The ACE I/D genotype frequencies in hyperthyroid women with hypertension were as follows: II genotype was 23.53%, whereas in hyperthyroid women without hypertension, the I/D genotype frequencies were CC: 7.50%, CT: 72.50%, and TT: 20.00%. There were no differences in age, thyroid-stimulating hormone, FT4, systolic blood pressure (SBP), diastolic BP between subgroups in ACE I/D, and at THRA rs-939348. There was a significant difference in the levels of FT4 concentrations in THRA rs-939348. Hypertension groups have higher SBP than controls. Genotype II had higher SBP but it was not statistically significant, while individuals with allele I had SBP higher than D allele.
CONCLUSIONS: The I allele of the ACE gene is involved in susceptibility to hypertension and polymorphism in THRA increasing concentration of FT4. There was no statistically significant difference in blood pressure between hyperthyroid women with or without hypertension.
Collapse
|
38
|
Lino CA, de Bortoli Teixeira L, Capelupe Simões S, de Oliveira Silva T, Diniz GP, da Costa-Neto CM, Barreto-Chaves MLM. Beta-arrestin 2 mediates cardiac hypertrophy induced by thyroid hormones via AT1R. J Cell Physiol 2021; 236:4640-4654. [PMID: 33345322 DOI: 10.1002/jcp.30187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 12/30/2022]
Abstract
We have previously reported that angiotensin II receptor type 1 (AT1R) contributes to the hypertrophic effects of thyroid hormones (TH) in cardiac cells. Even though evidence indicates crosstalks between TH and AT1R, the underlying mechanisms are poorly understood. Beta-arrestin (ARRB) signaling has been described as noncanonical signal transduction pathway that exerts important effects in the cardiovascular system through G-protein-coupled receptors, as AT1R. Herein, we investigated the contribution of ARRB signaling in TH-induced cardiomyocyte hypertrophy. Primary cardiomyocyte cultures were treated with Triiodothyronine (T3) to induce cell hypertrophy. T3 rapidly activates extracellular signal-regulated kinase 1/2 (ERK1/2) signaling, which was partially inhibited by AT1R blockade. Also, ERK1/2 inhibition attenuated the hypertrophic effects of T3. ARRB2 was upregulated by T3, and small interfering RNA assays revealed the role of ARRB2-but not ARRB1-on ERK1/2 activation and cardiomyocyte hypertrophy. Corroborating these findings, the ARRB2-overexpressed cells showed increased expression of hypertrophic markers, which were attenuated by ERK1/2 inhibition. Immunocytochemistry and immunoprecipitation assays revealed the increased expression of nuclear AT1R after T3 stimulation and the increased interaction of AT1R/ARRB2. The inhibition of endocytosis also attenuated the T3 effects on cardiac cells. Our results evidence the contribution of ARRB2 on ERK1/2 activation and cardiomyocyte hypertrophy induced by T3 via AT1R.
Collapse
Affiliation(s)
- Caroline Antunes Lino
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Larissa de Bortoli Teixeira
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Sarah Capelupe Simões
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Gabriela Placoná Diniz
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Claudio Miguel da Costa-Neto
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | | |
Collapse
|
39
|
Huang HB, Siao CY, Lo YTC, Shih SF, Lu CH, Huang PC. Mediation effects of thyroid function in the associations between phthalate exposure and glucose metabolism in adults. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 278:116799. [PMID: 33743268 DOI: 10.1016/j.envpol.2021.116799] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/04/2021] [Accepted: 02/18/2021] [Indexed: 06/12/2023]
Abstract
The mediating influence of thyroid function on the association of phthalate exposure with glucose metabolism, including insulin resistance, remains unclear. We explored the mediating influence of thyroid hormone levels on the phthalate exposure-insulin resistance association. This cross-sectional study of 217 Taiwanese adults assessed insulin resistance (Homeostatic Model Assessment for Insulin Resistance, HOMA-IR scores) and the levels of 11 urinary phthalate metabolites and 5 thyroid hormones. Multiple regression models were used to analyze the associations among serum thyroid hormone levels, urinary phthalate metabolite levels, and HOMA-IR scores. The mediation analysis assessed the influence of thyroid function on the phthalate exposure-HOMA-IR association. Our data indicated urinary mono-ethylhexyl phthalate (MEHP) levels was negatively associated with free thyroxine (T4) (β = -0.018; 95% confidence interval [CI]: -0.031, -0.005) and positively associated with HOMA-IR scores (β = 0.051, 95% CI: 0.012, 0.090). The study also revealed urinary mono-(2-ethyl-5-oxohexyl) phthalate (MEOHP) levels was negatively associated with free T4 (β = -0.036, 95% CI: -0.056, -0.017) and HOMA-IR (β = 0.070, 95% CI: 0.013, 0.126). Free T4 and HOMA-IR had a negative association (β = -0.757, 95% CI: -1.122, -0.392). In the mediation analysis, free T4 mediated 24% and 35% of the associations of urinary MEHP and MEOHP with HOMA-IR, respectively. Our findings revealed the mediating role of thyroid function in the phthalate exposure-glucose metabolism association in adults.
Collapse
Affiliation(s)
- Han-Bin Huang
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Chi-Ying Siao
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Yuan-Ting C Lo
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Fang Shih
- Department of Health Administration, College of Health Professions, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Chieh-Hua Lu
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Po-Chin Huang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan; Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Safety, Health and Environmental Engineering, National United University, Miaoli, Taiwan; National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan.
| |
Collapse
|
40
|
Estradiol Regulates mRNA Levels of Estrogen Receptor Beta 4 and Beta 5 Isoforms and Modulates Human Granulosa Cell Apoptosis. Int J Mol Sci 2021; 22:ijms22095046. [PMID: 34068748 PMCID: PMC8126246 DOI: 10.3390/ijms22095046] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 11/17/2022] Open
Abstract
Estrogen receptor beta (ERβ) plays a critical role in granulosa cell (GC) functions. The existence of four human ERβ splice isoforms in the ovary suggests their differential implication in 17β-estradiol (E2) actions on GC apoptosis causing follicular atresia. In this study, we investigated whether E2 can regulate ERβ isoforms expression to fine tune its apoptotic activities in human GC. For this purpose, we measured by RT-qPCR the expression of ERβ isoforms in primary culture of human granulosa cells (hGCs) collected from patients undergoing in vitro fertilization, before and after E2 exposure. Besides, we assessed the potential role of ERβ isoforms on cell growth and apoptosis after their overexpression in a human GC line (HGrC1 cells). We confirmed that ERβ1, ERβ2, ERβ4, and ERβ5 isoform mRNAs were predominant over that of ERα in hGCs, and found that E2 selectively regulates mRNA levels of ERβ4 and ERβ5 isoforms in these cells. In addition, we demonstrated that overexpression of ERβ1 and ERβ4 in HGrC1 cells increased cell apoptosis by 225% while ERβ5 or ERβ2 had no effect. Altogether, our study revealed that E2 may influence GC fate by specifically regulating the relative abundance of ERβ isoforms mRNA to modulate the balance between pro-apoptotic and non-apoptotic ERβ isoforms.
Collapse
|
41
|
Changes in the Ocular Parameters of Patients with Graves' Disease after Antithyroid Drug Treatment. ACTA ACUST UNITED AC 2021; 57:medicina57050414. [PMID: 33922887 PMCID: PMC8146600 DOI: 10.3390/medicina57050414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 11/24/2022]
Abstract
Background and Objectives: To find the differences in ocular axial length, keratometric measurements, and intraocular lens (IOL) power in patients with Graves’ disease (GD) after treatment with a thionamide antithyroid drug (ATD), methimazole. Materials and Methods: The medical charts of 28 patients (4 males and 24 females; mean age: 47.2 ± 21.2 years) were studied. Each patient was examined twice using an IOL Master Device and keratometry at the first visit (before ATD treatment) and after 1 month of ATD treatment. The IOL power was calculated for each patient using the Hoffer Q, SRK-2, and SRK/T formulas according to axial length. Results: After 1 month, the axial length increased (right and left eyes: p < 0.001 and p = 0.05, respectively). Based on keratometry, changes in the horizontal and vertical optical power [in diopters (D)] were not statistically significant. However, the IOL power changed after 1 month of ATD treatment in 64.3% of the patients. In 14 patients (50%), there was a 0.5–1.0 D IOL power decrease in single eyes; in two patients (7.1%), an IOL power decrease of 0.5–1.0 D in both eyes; and in two patients (7.1%), a 0.5 D IOL power increase in single eyes. The calculated IOL power values were lower after ATD treatment (right and left eyes, p = 0.010 and p = 0.018, respectively). Conclusions: The IOL power changed in 64.3% of GD patients after ATD treatment. Therefore, avoiding cataract surgery at the early stage of ATD treatment would be appropriate for selecting a more accurate IOL power.
Collapse
|
42
|
Liu S, Li D, Shi D, Zhang G, Luo X, Xu Q, Zhao L, Guo J, Yan G. Construction of a room-temperature phosphorescent quantum dot probe and quantitative detection of thyroxine and carbamazepine. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129582] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
43
|
Liang Y, Zhao D, Wang R, Dang P, Xi Y, Zhang D, Wang W, Shan Z, Teng X, Teng W. Generation and Characterization of a New Resistance to Thyroid Hormone Mouse Model with Thyroid Hormone Receptor Alpha Gene Mutation. Thyroid 2021; 31:678-691. [PMID: 32924834 DOI: 10.1089/thy.2019.0733] [Citation(s) in RCA: 4] [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] [Indexed: 12/12/2022]
Abstract
Background: In humans, resistance to thyroid hormone (RTH) caused by mutations in the thyroid hormone receptor alpha (THRA) gene, RTHα, manifests as tissue-specific hypothyroidism and circulating thyroid hormone levels exhibit hypothyroid-like clinical features. Before the identification of patients with RTHα, several Thrα1 knock-in mouse models were generated to clarify the function of TRα1. However, the phenotypes of these mice were not consistent with the clinical presentation of RTHα in humans. For the present study, we generated an RTHα mouse model that carries the Thra1E403X mutation found in human RTHα patients. Here, we report the gross phenotypes of this mouse RTHα model. Methods: Traditional homologous recombination gene targeting techniques were used to introduce a mutation (Thra1E403X) in the mouse Thra gene. The phenotypes of the resulting mice were studied and compared with clinical features observed for RTHα with THRAE403X. Results: Thrα1E403X/E403X homozygous mice exhibited severe neurological phenotypes, such as spasticity and motor ataxia, which were similar to those observed in endemic cretinism. Thrα1E403X/+ heterozygous mice reproduced most clinical manifestations of patient with RTHα, such as a normal survival rate and male fertility, as well as delayed postnatal growth and development, neurological and motor coordination deficits, and anemia. The mice had typical thyroid function with a modest increase in serum triiodothyronine (T3) levels, a low thyroxine (T4)/T3 ratio, and low reverse T3 (rT3) levels. Conclusions: The Thrα1E403X/+ mice faithfully recapitulate the clinical features of human RTHα and thus can provide a useful tool to dissect the role of TRα1 in development and to determine the pathological mechanisms of RTHα.
Collapse
Affiliation(s)
- Yue Liang
- Department of Endocrinology and Metabolism, Endocrine Institute, and Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, China
| | - Defa Zhao
- Department of Endocrinology and Metabolism, Endocrine Institute, and Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, China
| | - Ranran Wang
- Department of Endocrinology and Metabolism, Endocrine Institute, and Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, China
| | - Pingping Dang
- Department of Endocrinology and Metabolism, Endocrine Institute, and Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, China
| | - Yue Xi
- Department of Endocrinology and Metabolism, Endocrine Institute, and Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, China
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Dan Zhang
- Department of Endocrinology and Metabolism, Endocrine Institute, and Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, China
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Wei Wang
- Department of Endocrinology and Metabolism, Endocrine Institute, and Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, China
| | - Zhongyan Shan
- Department of Endocrinology and Metabolism, Endocrine Institute, and Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, China
| | - Xiaochun Teng
- Department of Endocrinology and Metabolism, Endocrine Institute, and Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, China
| | - Weiping Teng
- Department of Endocrinology and Metabolism, Endocrine Institute, and Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
44
|
Abstract
Thyroid hormone (T3) is critical not only for organ function and metabolism in the adult but also for animal development. This is particularly true during the neonatal period when T3 levels are high in mammals. Many processes during this postembryonic developmental period resemble those during amphibian metamorphosis. Anuran metamorphosis is perhaps the most dramatic developmental process controlled by T3 and affects essentially all organs/tissues, often in an organ autonomous manner. This offers a unique opportunity to study how T3 regulates vertebrate development. Earlier transgenic studies in the pseudo-tetraploid anuran Xenopus laevis revealed that T3 receptors (TRs) are necessary and sufficient for mediating the effects of T3 during metamorphosis. Recent gene knockout studies with gene-editing technologies in the highly related diploid anuran Xenopus tropicalis showed, surprisingly, that TRs are not required for most metamorphic transformations, although tadpoles lacking TRs are stalled at the climax of metamorphosis and eventually die. Analyses of the changes in different organs suggest that removal of TRs enables premature development of many adult tissues, likely due to de-repression of T3-inducible genes, while preventing the degeneration of tadpole-specific tissues, which is possibly responsible for the eventual lethality. Comparison with findings in TR knockout mice suggests both conservation and divergence in TR functions, with the latter likely due to the greatly reduced need, if any, to remove embryo/prenatal-specific tissues during mammalian postembryonic development.
Collapse
Affiliation(s)
- Yun-Bo Shi
- Section on Molecular Morphogenesis, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
- Correspondence: Yun-Bo Shi, Section on Molecular Morphogenesis, National Institute of Child Health and Human Development, National Institutes of Health, 49 Convent Drive, Building 49, Room 6A82, MSC 4480, Bethesda, MD 20892, USA.
| |
Collapse
|
45
|
Breton JM, Long KLP, Barraza MK, Perloff OS, Kaufer D. Hormonal Regulation of Oligodendrogenesis II: Implications for Myelin Repair. Biomolecules 2021; 11:290. [PMID: 33669242 PMCID: PMC7919830 DOI: 10.3390/biom11020290] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/10/2021] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Alterations in myelin, the protective and insulating sheath surrounding axons, affect brain function, as is evident in demyelinating diseases where the loss of myelin leads to cognitive and motor dysfunction. Recent evidence suggests that changes in myelination, including both hyper- and hypo-myelination, may also play a role in numerous neurological and psychiatric diseases. Protecting myelin and promoting remyelination is thus crucial for a wide range of disorders. Oligodendrocytes (OLs) are the cells that generate myelin, and oligodendrogenesis, the creation of new OLs, continues throughout life and is necessary for myelin plasticity and remyelination. Understanding the regulation of oligodendrogenesis and myelin plasticity within disease contexts is, therefore, critical for the development of novel therapeutic targets. In our companion manuscript, we review literature demonstrating that multiple hormone classes are involved in the regulation of oligodendrogenesis under physiological conditions. The majority of hormones enhance oligodendrogenesis, increasing oligodendrocyte precursor cell differentiation and inducing maturation and myelin production in OLs. Thus, hormonal treatments present a promising route to promote remyelination. Here, we review the literature on hormonal regulation of oligodendrogenesis within the context of disorders. We focus on steroid hormones, including glucocorticoids and sex hormones, peptide hormones such as insulin-like growth factor 1, and thyroid hormones. For each hormone, we describe whether they aid in OL survival, differentiation, or remyelination, and we discuss their mechanisms of action, if known. Several of these hormones have yielded promising results in both animal models and in human conditions; however, a better understanding of hormonal effects, interactions, and their mechanisms will ultimately lead to more targeted therapeutics for myelin repair.
Collapse
Affiliation(s)
- Jocelyn M Breton
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Kimberly L P Long
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Matthew K Barraza
- Molecular and Cellular Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Olga S Perloff
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
- Integrative Biology, University of California Berkeley, Berkeley, CA 94720, USA
- Canadian Institute for Advanced Research, Toronto, ON M5G1M1, Canada
| |
Collapse
|
46
|
Long KLP, Breton JM, Barraza MK, Perloff OS, Kaufer D. Hormonal Regulation of Oligodendrogenesis I: Effects across the Lifespan. Biomolecules 2021; 11:biom11020283. [PMID: 33672939 PMCID: PMC7918364 DOI: 10.3390/biom11020283] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 02/07/2023] Open
Abstract
The brain’s capacity to respond to changing environments via hormonal signaling is critical to fine-tuned function. An emerging body of literature highlights a role for myelin plasticity as a prominent type of experience-dependent plasticity in the adult brain. Myelin plasticity is driven by oligodendrocytes (OLs) and their precursor cells (OPCs). OPC differentiation regulates the trajectory of myelin production throughout development, and importantly, OPCs maintain the ability to proliferate and generate new OLs throughout adulthood. The process of oligodendrogenesis, the creation of new OLs, can be dramatically influenced during early development and in adulthood by internal and environmental conditions such as hormones. Here, we review the current literature describing hormonal regulation of oligodendrogenesis within physiological conditions, focusing on several classes of hormones: steroid, peptide, and thyroid hormones. We discuss hormonal regulation at each stage of oligodendrogenesis and describe mechanisms of action, where known. Overall, the majority of hormones enhance oligodendrogenesis, increasing OPC differentiation and inducing maturation and myelin production in OLs. The mechanisms underlying these processes vary for each hormone but may ultimately converge upon common signaling pathways, mediated by specific receptors expressed across the OL lineage. However, not all of the mechanisms have been fully elucidated, and here, we note the remaining gaps in the literature, including the complex interactions between hormonal systems and with the immune system. In the companion manuscript in this issue, we discuss the implications of hormonal regulation of oligodendrogenesis for neurological and psychiatric disorders characterized by white matter loss. Ultimately, a better understanding of the fundamental mechanisms of hormonal regulation of oligodendrogenesis across the entire lifespan, especially in vivo, will progress both basic and translational research.
Collapse
Affiliation(s)
- Kimberly L. P. Long
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA; (J.M.B.); (D.K.)
- Correspondence:
| | - Jocelyn M. Breton
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA; (J.M.B.); (D.K.)
| | - Matthew K. Barraza
- Department of Molecular and Cellular Biology, University of California, Berkeley, CA 94720, USA;
| | - Olga S. Perloff
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA 94143, USA;
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA; (J.M.B.); (D.K.)
- Department of Integrative Biology, University of California, Berkeley, CA 94720, USA
- Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
| |
Collapse
|
47
|
Tanizaki Y, Shibata Y, Zhang H, Shi YB. Analysis of Thyroid Hormone Receptor α-Knockout Tadpoles Reveals That the Activation of Cell Cycle Program Is Involved in Thyroid Hormone-Induced Larval Epithelial Cell Death and Adult Intestinal Stem Cell Development During Xenopus tropicalis Metamorphosis. Thyroid 2021; 31:128-142. [PMID: 32515287 PMCID: PMC7840310 DOI: 10.1089/thy.2020.0022] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: There are two highly conserved thyroid hormone (triiodothyronine [T3]) receptor (TR) genes, TRα and TRβ, in all vertebrates, and the expression of TRα but not TRβ is activated earlier than T3 synthesis during development. In human, high levels of T3 are present during the several months around birth, and T3 deficiency during this period causes severe developmental abnormalities including skeletal and intestinal defects. It is, however, difficult to study this period in mammals as the embryos and neonates depend on maternal supply of nutrients for survival. However, Xenopus tropicalis undergoes a T3-dependent metamorphosis, which drastically changes essentially every organ in a tadpole. Of interest is intestinal remodeling, which involves near complete degeneration of the larval epithelium through apoptosis. Concurrently, adult intestinal stem cells are formed de novo and subsequently give rise to the self-renewing adult epithelial system, resembling intestinal maturation around birth in mammals. We have previously demonstrated that T3 signaling is essential for the formation of adult intestinal stem cells during metamorphosis. Methods: We studied the function of endogenous TRα in the tadpole intestine by using knockout animals and RNA-seq analysis. Results: We observed that removing endogenous TRα caused defects in intestinal remodeling, including drastically reduced larval epithelial cell death and adult intestinal stem cell proliferation. Using RNA-seq on intestinal RNA from premetamorphic wild-type and TRα-knockout tadpoles treated with or without T3 for one day, before any detectable T3-induced cell death and stem cell formation in the tadpole intestine, we identified more than 1500 genes, which were regulated by T3 treatment of the wild-type but not TRα-knockout tadpoles. Gene Ontology and biological pathway analyses revealed that surprisingly, these TRα-regulated genes were highly enriched with cell cycle-related genes, in addition to genes related to stem cells and apoptosis. Conclusions: Our findings suggest that TRα-mediated T3 activation of the cell cycle program is involved in larval epithelial cell death and adult epithelial stem cell development during intestinal remodeling.
Collapse
Affiliation(s)
- Yuta Tanizaki
- Section on Molecular Morphogenesis, Cell Regulation and Development Affinity Group, Division of Molecular and Cellular Biology, and Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Yuki Shibata
- Section on Molecular Morphogenesis, Cell Regulation and Development Affinity Group, Division of Molecular and Cellular Biology, and Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Hongen Zhang
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, Cell Regulation and Development Affinity Group, Division of Molecular and Cellular Biology, and Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
- Address correspondence to: Yun-Bo Shi, PhD, Section on Molecular Morphogenesis, Cell Regulation and Development Affinity Group, Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 49 Room 6A82, Bethesda, MD 20814, USA
| |
Collapse
|
48
|
Sanoh S, Hanada H, Kashiwagi K, Mori T, Goto-Inoue N, Suzuki KIT, Mori J, Nakamura N, Yamamoto T, Kitamura S, Kotake Y, Sugihara K, Ohta S, Kashiwagi A. Amiodarone bioconcentration and suppression of metamorphosis in Xenopus. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2020; 228:105623. [PMID: 32956954 DOI: 10.1016/j.aquatox.2020.105623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 06/11/2023]
Abstract
Trace concentrations of a number of pharmaceutically active compounds have been detected in the aquatic environment in many countries, where they are thought to have the potential to exert adverse effects on non-target organisms. Amiodarone (AMD) is one such high-risk compound commonly used in general hospitals. AMD is known to alter normal thyroid hormone (TH) function, although little information is available regarding the specific mechanism by which this disruption occurs. Anuran tadpole metamorphosis is a TH-controlled developmental process and has proven to be useful as a screening tool for environmental pollutants suspected of disrupting TH functions. In the present study, our objective was to clarify the effects of AMD on Xenopus metamorphosis as well as to assess the bioconcentration of this pharmaceutical in the liver. We found that AMD suppressed spontaneous metamorphosis, including tail regression and hindlimb elongation in pro-metamorphic stage tadpoles, which is controlled by endogenous circulating TH, indicating that AMD is a TH antagonist. In transgenic X. laevis tadpoles carrying plasmid DNA containing TH-responsive element (TRE) and a 5'-upstream promoter region of the TH receptor (TR) βA1 gene linked to a green fluorescent protein (EGFP) gene, triiodothyronine (T3) exposure induced a strong EGFP expression in the hind limbs, whereas the addition of AMD to T3 suppressed EGFP expression, suggesting that this drug interferes with the binding of T3 to TR, leading to the inhibition of TR-mediated gene expression. We also found AMD to be highly bioconcentrated in the liver of pro-metamorphic X. tropicalis tadpoles, and we monitored hepatic accumulation of this drug using mass spectrometry imaging (MSI). Our findings suggest that AMD imposes potential risk to aquatic wildlife by disrupting TH homeostasis, with further possibility of accumulating in organisms higher up in the food chain.
Collapse
Affiliation(s)
- Seigo Sanoh
- Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8553, Japan.
| | - Hideki Hanada
- Amphibian Research Center, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan.
| | - Keiko Kashiwagi
- Amphibian Research Center, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan.
| | - Tsukasa Mori
- Department of Marine Science and Resources, Nihon University College of Bioresource Sciences, Kameino 1866, Fujisawa 252-0880, Japan.
| | - Naoko Goto-Inoue
- Department of Marine Science and Resources, Nihon University College of Bioresource Sciences, Kameino 1866, Fujisawa 252-0880, Japan.
| | - Ken-Ichi T Suzuki
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan.
| | - Junpei Mori
- Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8553, Japan.
| | - Naoki Nakamura
- Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8553, Japan.
| | - Takashi Yamamoto
- Program of Mathematical and Life Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan.
| | - Shigeyuki Kitamura
- Nihon Pharmaceutical University, Komuro 10281, Ina-machi, Kitaadachi-gun, Saitama, 362-0806, Japan.
| | - Yaichiro Kotake
- Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8553, Japan.
| | - Kazumi Sugihara
- Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshinkai, Kure City, Hiroshima 737-0112, Japan.
| | - Shigeru Ohta
- Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8553, Japan; Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 641-8509, Japan.
| | - Akihiko Kashiwagi
- Amphibian Research Center, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan.
| |
Collapse
|
49
|
Yang Y, Zhan F, Wang YC, Wang B, Shi MX, Guo C, Xu DX, Meng XH. Pubertal fenvalerate exposure impairs cognitive and behavioral development partially through down-regulating hippocampal thyroid hormone receptor signaling. Toxicol Lett 2020; 332:192-201. [PMID: 32693020 DOI: 10.1016/j.toxlet.2020.07.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 07/10/2020] [Accepted: 07/17/2020] [Indexed: 12/18/2022]
Abstract
Fenvalerate, a synthetic pyrethroid insecticide, is an environmental endocrine disruptor and neurodevelopmental toxicant. An early report found that pubertal exposure to high-dose fenvalerate impaired cognitive and behavioral development. Here, we aimed to further investigate the effect of pubertal exposure to low-dose fenvalerate on cognitive and behavioral development. Mice were orally administered with fenvalerate (0.2, 1.0 and 5.0 mg/kg) daily from postnatal day (PND) 28 to PND56. Learning and memory were assessed by Morris water maze. Anxiety-related activities were detected by open-field and elevated plus-maze. Increased anxiety activities were observed only in females exposed to fenvalerate. Spatial learning and memory were damaged only in females exposed to fenvalerate. Histopathology observed numerous scattered shrinking neurons and nuclear pyknosis in hippocampal CA1 region. Neuronal density was reduced in hippocampal CA1 region of fenvalerate-exposed mice. Mechanistically, hippocampal thyroid hormone receptor (TR)β1 was down-regulated in a dose-dependent manner in females. In addition, TRα1 was declined only in females exposed to 5.0 mg/kg fenvalerate. Taken together, these suggests that pubertal exposure to low-dose fenvalerate impairs cognitive and behavioral development in a gender-dependent manner. Hippocampal TR signaling may be, at least partially, involved in fenvalerate-induced impairment of cognitive and behavioral development.
Collapse
Affiliation(s)
- Yang Yang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People's Republic of China; Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei, Anhui, China
| | - Feng Zhan
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People's Republic of China; Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei, Anhui, China
| | - Ye-Cheng Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People's Republic of China; Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei, Anhui, China
| | - Bo Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People's Republic of China; Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei, Anhui, China
| | - Meng-Xing Shi
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People's Republic of China; Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei, Anhui, China
| | - Ce Guo
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People's Republic of China; Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei, Anhui, China
| | - De-Xiang Xu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People's Republic of China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, China.
| | - Xiu-Hong Meng
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People's Republic of China; Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei, Anhui, China.
| |
Collapse
|
50
|
Ditsch N, Heublein S, Jeschke U, Sattler C, Kuhn C, Hester A, Czogalla B, Trillsch F, Mahner S, Engel J, Mayr D, Schmoeckel E. Cytoplasmic versus nuclear THR alpha expression determines survival of ovarian cancer patients. J Cancer Res Clin Oncol 2020; 146:1923-1932. [PMID: 32533406 PMCID: PMC7324415 DOI: 10.1007/s00432-020-03241-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 05/01/2020] [Indexed: 12/16/2022]
Abstract
Purpose
Thyroid hormone receptors (THR) have manifold functions and are involved in the carcinogenesis of several tumor types. Within this study, we aimed to investigate the expression pattern (nuclear versus cytoplasmic) of the THR alpha and its impact on patients survival in ovarian cancer (OvCa). Methods The presence of the thyroid hormone receptors THRα, THRα1 and − 2 was investigated in 156 ovarian cancer samples using immunohistochemistry (IHC) using semi-quantitative immunoreactivity (IR) scores and correlated with clinical, pathological data, subtype of ovarian cancer, clinical data, staining of 20 already described OvCa marker proteins and overall survival (OS). Results Among all subtypes of OvCa, clear cell carcinomas showed the highest THRα expression. Furthermore, nuclear THRα was associated with a reduced survival in this subtype. However, nuclear expressed THRα1 turned out to be a positive prognosticator for all subtypes of OvCa patients. Nuclear THRα2 is a positive prognosticator for OvCa patients of the serous subtype. In contrast, cytoplasmic expression THRα2 was associated with a reduced OS in all subtypes of OvCa patients; while, cytoplasmic expression of THRα1 is associated with reduced OS in mucinous OvCa patients only. In addition, THRα expression correlates with gonadotropin receptors, steroid hormone receptors, TA-MUC1 and glycodelin. Conclusion Depending on nuclear or cytoplasmic expression, our study shows that THRα and its isoforms 1 and 2 provide different prognostic information for ovarian cancer patients. Further investigations should analyze if THRs may represent new endocrine targets for the treatment of ovarian cancer. Electronic supplementary material The online version of this article (10.1007/s00432-020-03241-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nina Ditsch
- Department of Obstetrics and Gynecology, LMU Munich, University Hospital, Marchioninistr. 15, 81377 Munich, Germany
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Sabine Heublein
- Department of Obstetrics and Gynecology, LMU Munich, University Hospital, Marchioninistr. 15, 81377 Munich, Germany
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Ruprecht-Karls-University of Heidelberg, 69120 Heidelberg, Germany
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, LMU Munich, University Hospital, Marchioninistr. 15, 81377 Munich, Germany
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Cornelia Sattler
- Department of Obstetrics and Gynecology, LMU Munich, University Hospital, Marchioninistr. 15, 81377 Munich, Germany
| | - Christina Kuhn
- Department of Obstetrics and Gynecology, LMU Munich, University Hospital, Marchioninistr. 15, 81377 Munich, Germany
| | - Anna Hester
- Department of Obstetrics and Gynecology, LMU Munich, University Hospital, Marchioninistr. 15, 81377 Munich, Germany
| | - Bastian Czogalla
- Department of Obstetrics and Gynecology, LMU Munich, University Hospital, Marchioninistr. 15, 81377 Munich, Germany
| | - Fabian Trillsch
- Department of Obstetrics and Gynecology, LMU Munich, University Hospital, Marchioninistr. 15, 81377 Munich, Germany
| | - Sven Mahner
- Department of Obstetrics and Gynecology, LMU Munich, University Hospital, Marchioninistr. 15, 81377 Munich, Germany
| | - Jutta Engel
- Munich Cancer Registry (MCR), Bavarian Cancer Registry—Regional Centre Munich (LGL), Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Doris Mayr
- Department of Pathology, LMU Munich, Thalkirchner Str. 36, 80337 Munich, Germany
| | - Elisa Schmoeckel
- Department of Pathology, LMU Munich, Thalkirchner Str. 36, 80337 Munich, Germany
| |
Collapse
|