Sex-Specific Effects of THRβ Signaling on Metabolic Responses to High Fat Diet in Mice.

IF 3.8 3区 医学 Q2 ENDOCRINOLOGY & METABOLISM
Aruljothi Muralidharan, Gustavo A Gomez, Chandrasekhar Kesavan, Sheila Pourteymoor, Destiney Larkin, William Tambunan, V Franklin Sechriest, Subburaman Mohan
{"title":"Sex-Specific Effects of THRβ Signaling on Metabolic Responses to High Fat Diet in Mice.","authors":"Aruljothi Muralidharan, Gustavo A Gomez, Chandrasekhar Kesavan, Sheila Pourteymoor, Destiney Larkin, William Tambunan, V Franklin Sechriest, Subburaman Mohan","doi":"10.1210/endocr/bqae075","DOIUrl":null,"url":null,"abstract":"<p><p>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.</p>","PeriodicalId":11819,"journal":{"name":"Endocrinology","volume":null,"pages":null},"PeriodicalIF":3.8000,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11237353/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Endocrinology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1210/endocr/bqae075","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"ENDOCRINOLOGY & METABOLISM","Score":null,"Total":0}
引用次数: 0

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.

THRβ信号在改变小鼠高脂饮食代谢效应中的性别特异性效应机制
甲状腺激素(TH)在调节骨骼和脂肪组织的功能方面起着至关重要的作用。鉴于甲状腺激素通过甲状腺激素受体-β(TRβ)在肝组织中发挥降低胆固醇的作用,我们假设使用MGL3196(MGL)的TRβ激动剂疗法将能有效治疗成年C57BL/6J小鼠因12周高脂肪饮食(HFD)引起的脂肪增加和骨质流失。转录和血清分析表明,高脂饮食诱导的瘦素可促进雄性和雌性小鼠的体重增加,但 MGL 只能抑制瘦素诱导和雄性小鼠的体重增加。体外研究表明,雌激素会抑制脂肪细胞中MGL的活性,这表明雌激素可能会干扰MGL-TRβ的功能。与全身性肥胖相比,高密度脂蛋白胆固醇(HFD)会降低雄性小鼠的骨量,但不会降低雌性小鼠的骨量。矛盾的是,MGL 治疗逆转了阑尾骨的宏观骨密度损失,但显微 CT 显示,MGL 加剧了 HFD 诱导的骨小梁损失和骨强度。在关于 HFD 对骨骼影响机制的研究中,我们发现 HFD 会诱导男性股骨中 Rankl 的表达,而 MGL 可阻断这种表达。通过体外试验,我们发现 RANKL 通过诱导炎性细胞因子 TNFα、Il-1β 和 CCL2 间接抑制骨生成细胞的成骨细胞系分配。最后,我们发现MGL通过非基因组TRβ信号在全身性脂肪和骨骼中发挥作用,因为在基因组正常但非基因组TRβ信号缺陷的TRβ147F基因敲除小鼠中,HFD介导的表型没有得到挽救。我们的研究结果表明,HFD对体脂和骨骼表型的负面影响受到MGL以性别特异性方式的影响。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Endocrinology
Endocrinology 医学-内分泌学与代谢
CiteScore
8.10
自引率
4.20%
发文量
195
审稿时长
2-3 weeks
期刊介绍: The mission of Endocrinology is to be the authoritative source of emerging hormone science and to disseminate that new knowledge to scientists, clinicians, and the public in a way that will enable "hormone science to health." Endocrinology welcomes the submission of original research investigating endocrine systems and diseases at all levels of biological organization, incorporating molecular mechanistic studies, such as hormone-receptor interactions, in all areas of endocrinology, as well as cross-disciplinary and integrative studies. The editors of Endocrinology encourage the submission of research in emerging areas not traditionally recognized as endocrinology or metabolism in addition to the following traditionally recognized fields: Adrenal; Bone Health and Osteoporosis; Cardiovascular Endocrinology; Diabetes; Endocrine-Disrupting Chemicals; Endocrine Neoplasia and Cancer; Growth; Neuroendocrinology; Nuclear Receptors and Their Ligands; Obesity; Reproductive Endocrinology; Signaling Pathways; and Thyroid.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信