1
|
Dinh J, Yi D, Lin F, Xue P, Holloway ND, Xie Y, Ibe NU, Nguyen HP, Viscarra JA, Wang Y, Sul HS. The microprotein C16orf74/MICT1 promotes thermogenesis in brown adipose tissue. EMBO J 2025:10.1038/s44318-025-00444-x. [PMID: 40355556 DOI: 10.1038/s44318-025-00444-x] [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: 08/16/2024] [Revised: 03/28/2025] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
Brown and beige adipose tissues are metabolically beneficial for increasing energy expenditure via thermogenesis, mainly through UCP1 (uncoupling protein 1). Here, we identify C16orf74, subsequently named MICT1 (microprotein for thermogenesis 1), as a microprotein that is specifically and highly expressed in brown adipose tissue (BAT) and is induced upon cold exposure. MICT1 interacts with protein phosphatase 2B (PP2B, calcineurin) through the docking motif PNIIIT, thereby interfering with dephosphorylation of the regulatory subunit of protein kinase A (PKA), RIIβ, and potentiating PKA activity in brown adipocytes. Overexpression of MICT1 in differentiated brown adipocytes promotes thermogenesis, showing increased oxygen consumption rate (OCR) with higher thermogenic gene expression during β3-adrenergic stimulation, while knockdown of MICT1 impairs thermogenic responses. Moreover, BAT-specific MICT1 ablation in mice suppresses thermogenic capacity to increase adiposity and insulin resistance. Conversely, MICT1 overexpression in BAT or treating mice with a chemical inhibitor that targets the PP2B docking motif of MICT1 enhances thermogenesis. This results in cold tolerance and increased energy expenditure, protection against diet-induced and genetic obesity and insulin resistance, thus suggesting a therapeutic potential of MICT1 targeting.
Collapse
Affiliation(s)
- Jennie Dinh
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, CA, 94720, USA
| | - Danielle Yi
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, CA, 94720, USA
- Endocrinology Program, University of California, Berkeley, CA, 94720, USA
| | - Frances Lin
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, CA, 94720, USA
| | - Pengya Xue
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, CA, 94720, USA
| | - Nicholas D Holloway
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, CA, 94720, USA
- Endocrinology Program, University of California, Berkeley, CA, 94720, USA
| | - Ying Xie
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, CA, 94720, USA
| | - Nnejiuwa U Ibe
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, CA, 94720, USA
| | - Hai P Nguyen
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, CA, 94720, USA
- Endocrinology Program, University of California, Berkeley, CA, 94720, USA
- University of Texas at Austin, Austin, TX, 78723, USA
| | - Jose A Viscarra
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, CA, 94720, USA
| | - Yuhui Wang
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, CA, 94720, USA
| | - Hei Sook Sul
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, CA, 94720, USA.
- Endocrinology Program, University of California, Berkeley, CA, 94720, USA.
| |
Collapse
|
2
|
Kato YS, Shimazaki Y, Chuma S, Shiraya K, Nakane Y, Sugi T, Okabe K, Harada Y, Sotoma S. Fluorescent Thermometers Based on Carbon Quantum Dots with Various Detection Modes for Intracellular Temperature Measurement. NANO LETTERS 2025; 25:5688-5696. [PMID: 40134068 DOI: 10.1021/acs.nanolett.4c06642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
We report that carbon quantum dots (CQDs) synthesized via a hydrothermal process using anthraquinone derivatives and l-cysteine provide versatile detection modes, making them suitable for various experimental setups. By modification of the precursor structures, these CQDs can function as different types of fluorescent nanothermometers, including those based on fluorescence intensity, ratiometrics, and fluorescence lifetime. Notably, fluorescence lifetime-based CQDs demonstrate robust performance under a wide range of conditions, including variations in pH and ionic strength. The CQDs exhibit low cytotoxicity and high cellular uptake efficiency, enabling wash-free imaging and precise fluorescence lifetime-based temperature measurements at the single-cell level. Furthermore, we successfully measured temperature changes associated with biochemical reactions, including the increase in cellular temperature induced by mitochondrial depolarization. In addition, these fluorescence lifetime-based measurements could be cross-verified using their fluorescence intensity. These findings underscore the potential of CQDs as versatile and minimally invasive tools for nanoscale thermometry in live cells.
Collapse
Affiliation(s)
- Yuki S Kato
- Department of Biological Sciences, School of Science, The University of Osaka, Machikaneyamacho Toyonaka, Osaka 560-0043, Japan
| | - Yukiho Shimazaki
- Department of Biological Sciences, Graduate School of Science, The University of Osaka, Machikaneyamacho Toyonaka, Osaka 560-0043, Japan
- Institute for Protein Research, The University of Osaka, Yamadaoka Suita, Osaka 565-0871, Japan
| | - Shunsuke Chuma
- Institute for Protein Research, The University of Osaka, Yamadaoka Suita, Osaka 565-0871, Japan
| | - Kota Shiraya
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Yurina Nakane
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Kagamiyama, Higashihiroshima, Hiroshima 739-0046, Japan
| | - Takuma Sugi
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Kagamiyama, Higashihiroshima, Hiroshima 739-0046, Japan
| | - Kohki Okabe
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo Bunkyo City, Tokyo 113-0033, Japan
| | - Yoshie Harada
- Institute for Protein Research, The University of Osaka, Yamadaoka Suita, Osaka 565-0871, Japan
- Center for Quantum Information and Quantum Biology, The University of Osaka, Machikaneyamacho Toyonaka, Osaka 560-0043, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), The University of Osaka, Yamadaoka Suita, Osaka 565-0871, Japan
| | - Shingo Sotoma
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| |
Collapse
|
3
|
Wazawa T, Ozaki-Noma R, Kai L, Fukushima SI, Matsuda T, Nagai T. Genetically-encoded temperature indicators for thermal biology. Biophys Physicobiol 2025; 22:e220008. [PMID: 40309302 PMCID: PMC12040488 DOI: 10.2142/biophysico.bppb-v22.0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 04/03/2025] [Indexed: 05/02/2025] Open
Abstract
Temperature crucially affects molecular processes in living organisms and thus it is one of the vital physical parameters for life. To investigate how temperature is biologically maintained and regulated and its biological impact on organisms, it is essential to measure the spatial distribution and/or temporal changes of temperature across different biological scales, from whole organism to subcellular structures. Fluorescent nanothermometers have been developed as probes for temperature measurement by fluorescence microscopy for applications in microscopic scales where macroscopic temperature sensors are inaccessible, such as embryos, tissues, cells, and organelles. Although fluorescent nanothermometers have been developed from various materials, fluorescent protein-based ones are especially of interest because they can be introduced into cells as the transgenes for expression with or without specific localization, making them suitable for less-invasive temperature observation in living biological samples. In this article, we review protein-based fluorescent nanothermometers also known as genetically-encoded temperature indicators (GETIs), covering most published GETIs, for developers, users, and researchers in thermal biology as well as interested readers. We provide overviews of the temperature sensing mechanisms and measurement methods of these protein-based fluorescent nanothermometers. We then outline key information for GETI development, focusing on unique protein engineering techniques and building blocks distinct to GETIs, unlike other fluorescent nanothermometers. Furthermore, we propose several standards for the characterization of GETIs. Additionally, we explore various issues and offer perspectives in the field of thermal biology.
Collapse
Affiliation(s)
- Tetsuichi Wazawa
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Ibaraki, Osaka 567-0047, Japan
| | - Ryohei Ozaki-Noma
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Ibaraki, Osaka 567-0047, Japan
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Lu Kai
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Ibaraki, Osaka 567-0047, Japan
| | - Shun-ichi Fukushima
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Ibaraki, Osaka 567-0047, Japan
| | - Tomoki Matsuda
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Ibaraki, Osaka 567-0047, Japan
- Department of Biosciences, School of Science, Kitasato University, Sagamihara, Kanagawa 252-0373, Japan
| | - Takeharu Nagai
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Ibaraki, Osaka 567-0047, Japan
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
4
|
Wang Y, Xie H, Liu Q, Wang N, Luo X, Sun F, Zhu J, Dong R, Wang Y, Gao J, Gao Z, Huang T, Liu X, Yu Q, Wang T, Li Y, Song D, Liu S, Zhang S, Yin H, Kong W, Wang CY. Kdm2a inhibition in skeletal muscle improves metabolic flexibility in obesity. Nat Metab 2025; 7:383-400. [PMID: 39870955 PMCID: PMC11860252 DOI: 10.1038/s42255-024-01210-9] [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: 07/17/2024] [Accepted: 12/17/2024] [Indexed: 01/29/2025]
Abstract
Skeletal muscle is a critical organ in maintaining homoeostasis against metabolic stress, and histone post-translational modifications are pivotal in those processes. However, the intricate nature of histone methylation in skeletal muscle and its impact on metabolic homoeostasis have yet to be elucidated. Here, we report that mitochondria-rich slow-twitch myofibers are characterized by significantly higher levels of H3K36me2 along with repressed expression of Kdm2a, an enzyme that specifically catalyses H3K36me2 demethylation. Deletion or inhibition of Kdm2a shifts fuel use from glucose under cold challenge to lipids under obese conditions by increasing the proportion of mitochondria-rich slow-twitch myofibers. This protects mice against cold insults and high-fat-diet-induced obesity and insulin resistance. Mechanistically, Kdm2a deficiency leads to a marked increase in H3K36me2 levels, which then promotes the recruitment of Mrg15 to the Esrrg locus to process its precursor messenger RNA splicing, thereby reshaping skeletal muscle metabolic profiles to induce slow-twitch myofiber transition. Collectively, our data support the role of Kdm2a as a viable target against metabolic stress.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Xie
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qianrui Liu
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Wang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Luo
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Sun
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinghan Zhu
- Department of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruihan Dong
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Wang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Gao
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhichao Gao
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Teng Huang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Liu
- Department of Interventional Radiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qilin Yu
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Wang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Li
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danni Song
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiwei Liu
- Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Third Hospital of Shanxi Medical University, the Key Laboratory of Endocrine and Metabolic Diseases of Shanxi Province, Taiyuan, China
| | - Shu Zhang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Hao Yin
- Organ Transplant Center, Shanghai Changzheng Hospital (Second Affiliated Hospital of Naval Medical University), Shanghai, China.
| | - Wen Kong
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Cong-Yi Wang
- Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Third Hospital of Shanxi Medical University, the Key Laboratory of Endocrine and Metabolic Diseases of Shanxi Province, Taiyuan, China.
- The Center for Biomedical Research, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
5
|
Kang GS, Kim YE, Oh HR, Jo HJ, Bok S, Jeon YK, Cheon GJ, Roh TY, Chang YT, Park DJ, Ahn GO. Hypoxia-inducible factor-1α-deficient adipose-tissue macrophages produce the heat to mediate lipolysis of white adipose tissue through uncoupling protein-1. Lab Anim Res 2024; 40:37. [PMID: 39473019 PMCID: PMC11523771 DOI: 10.1186/s42826-024-00224-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Uncoupling protein 1 (UCP1) is a proton uncoupler located across the mitochondrial membrane generally involved in thermogenesis of brown adipose tissues. Although UCP1 is known to be strongly expressed in brown adipocytes, recent evidence suggest that white adipocytes can also express UCP1 under certain circumstances such as cold- or β-adrenergic receptor-stimulation, allowing them to acquire brown adipocyte-like features thereby becoming 'beige' adipocytes. RESULTS In this study, we report that UCP1 can be expressed in adipose-tissue macrophages (ATM) lacking functional hypoxia-inducible factor-1 (HIF-1) and this does not require cold- nor β-adrenergic receptor activation. By using myeloid-specific Hif-1α knockout (KO) mice, we observed that these mice were protected from diet-induced obesity and exhibited an improved thermogenic tolerance upon cold challenge. ATM isolated from white adipose tissues (WAT) of these mice fed with high fat diet exhibited significantly higher M2-polarization, decreased glycolysis, increased mitochondrial functions and acetyl-CoA levels, along with increased expression of Ucp1, peroxisome proliferator activated receptor-gamma co-activator-1a, and others involved in histone acetylation. Consistent with the increased Ucp1 gene expression, these ATM produced a significant amount of heat mediating lipolysis of co-cultured adipocytes liberating free fatty acid. Treating ATM with acetate, a substrate for acetyl-CoA synthesis was able to boost the heat production in wild-type or Hif-1α-deficient but not UCP1-deficient macrophages, indicating that UCP1 was necessary for the heat production in macrophages. Lastly, we observed a significant inverse correlation between the number of UCP1-expressing ATM in WAT and the body mass index of human individuals. CONCLUSIONS UCP1-expressing ATM produce the heat to mediate lipolysis of adipocytes, indicating that this can be a novel strategy to treat and prevent diet-induced obesity.
Collapse
Affiliation(s)
- Gi-Sue Kang
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Young-Eun Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
| | - Ho Rim Oh
- Department of Nuclear Medicine, College of Medicine, Seoul National University, Seoul, 03080, Korea
| | - Hye-Ju Jo
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Seoyeon Bok
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
| | - Yoon Kyung Jeon
- Department of Pathology, College of Medicine, Seoul National University, Seoul, 03080, Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, College of Medicine, Seoul National University, Seoul, 03080, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Korea
- College of Medicine, Cancer Research Institute, Seoul National University, Seoul, 03080, Korea
| | - Tae-Young Roh
- Department of Life Sciences, Ewha Womans University, Seoul, 03760, Korea
| | | | - Do Joong Park
- Department of Surgery, College of Medicine, Seoul National University, Seoul, 03080, Korea
| | - G-One Ahn
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea.
- College of Medicine, Cancer Research Institute, Seoul National University, Seoul, 03080, Korea.
| |
Collapse
|
6
|
Guarnieri AR, Anthony SR, Acharya P, Wen BY, Lanzillotta L, Gavin R, Tranter M. HuR-dependent expression of RyR2 contributes to calcium-mediated thermogenesis in brown adipocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619637. [PMID: 39484459 PMCID: PMC11527003 DOI: 10.1101/2024.10.22.619637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Several uncoupling protein 1 (UCP1)-independent thermogenic pathways have been described in thermogenic adipose tissue, including calcium-mediated thermogenesis in beige adipocytes via sarco/endoplasmic reticulum ATPase (SERCA). We have previously shown that adipocyte-specific deletion of the RNA binding protein human antigen R (HuR) results in thermogenic dysfunction independent of UCP1 expression. RNA sequencing revealed the downregulation of several genes involved in calcium ion transport upon HuR deletion. The goal of this work was to define the HuR-dependent mechanisms of calcium driven thermogenesis in brown adipocytes. We generated (BAT)-specific HuR-deletion (BAT-HuR -/- ) mice and show that their body weight, glucose tolerance, brown and white adipose tissue weights, and total lipid droplet size were not significantly different compared to wild-type. Similar to our initial findings in Adipo-HuR -/- mice, mice with BAT-specific HuR deletion are cold intolerant following acute thermal challenge at 4°C, demonstrating specificity of acute HuR-dependent thermogenesis to BAT. We also found decreased expression of ryanodine receptor 2 (RyR2), but no changes in RyR2, SERCA1, SERCA2, or UCP1 expression, in BAT from BAT-HuR -/- mice. Next, we used Fluo-4 calcium indicator dye to show that genetic deletion or pharmacological inhibition of HuR blunts the increase in cytosolic calcium concentration in SVF-derived primary brown adipocytes. Moreover, we saw a similar blunting in β-adrenergic-mediated heat generation, as assessed by ERtherm AC fluorescence, in SVF-derived brown adipocytes following HuR inhibition or deletion. Mechanistically, we show that HuR directly binds and reduces the decay rate of RyR2 mRNA in brown adipocytes, and stabilization of RyR2 via S107 rescues β-adrenergic-mediated cytosolic calcium increase and heat generation in HuR deficient brown adipocytes. In conclusion, our results suggest that HuR-dependent control of RyR2 expression plays a significant role in the thermogenic function of brown adipose tissue through modulation of SR calcium cycling.
Collapse
|
7
|
Baroi S, Czernik PJ, Khan MP, Letson J, Crowe E, Chougule A, Griffin PR, Rosen CJ, Lecka-Czernik B. PPARG in osteocytes controls cell bioenergetics and systemic energy metabolism independently of sclerostin levels in circulation. Mol Metab 2024; 88:102000. [PMID: 39074536 PMCID: PMC11367276 DOI: 10.1016/j.molmet.2024.102000] [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: 04/08/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024] Open
Abstract
OBJECTIVE The skeleton is one of the largest organs in the body, wherein metabolism is integrated with systemic energy metabolism. However, the bioenergetic programming of osteocytes, the most abundant bone cells coordinating bone metabolism, is not well defined. Here, using a mouse model with partial penetration of an osteocyte-specific PPARG deletion, we demonstrate that PPARG controls osteocyte bioenergetics and their contribution to systemic energy metabolism independently of circulating sclerostin levels, which were previously correlated with metabolic status of extramedullary fat depots. METHODS In vivo and in vitro models of osteocyte-specific PPARG deletion, i.e. Dmp1CrePparγflfl male and female mice (γOTKO) and MLO-Y4 osteocyte-like cells with either siRNA-silenced or CRISPR/Cas9-edited Pparγ. As applicable, the models were analyzed for levels of energy metabolism, glucose metabolism, and metabolic profile of extramedullary adipose tissue, as well as the osteocyte transcriptome, mitochondrial function, bioenergetics, insulin signaling, and oxidative stress. RESULTS Circulating sclerostin levels of γOTKO male and female mice were not different from control mice. Male γOTKO mice exhibited a high energy phenotype characterized by increased respiration, heat production, locomotion and food intake. This high energy phenotype in males did not correlate with "beiging" of peripheral adipose depots. However, both sexes showed a trend for reduced fat mass and apparent insulin resistance without changes in glucose tolerance, which correlated with decreased osteocytic responsiveness to insulin measured by AKT activation. The transcriptome of osteocytes isolated from γOTKO males suggested profound changes in cellular metabolism, fuel transport, mitochondria dysfunction, insulin signaling and increased oxidative stress. In MLO-Y4 osteocytes, PPARG deficiency correlated with highly active mitochondria, increased ATP production, and accumulation of reactive oxygen species (ROS). CONCLUSIONS PPARG in male osteocytes acts as a molecular break on mitochondrial function, and protection against oxidative stress and ROS accumulation. It also regulates osteocyte insulin signaling and fuel usage to produce energy. These data provide insight into the connection between osteocyte bioenergetics and their sex-specific contribution to the balance of systemic energy metabolism. These findings support the concept that the skeleton controls systemic energy expenditure via osteocyte metabolism.
Collapse
Affiliation(s)
- Sudipta Baroi
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| | - Piotr J Czernik
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| | - Mohd Parvez Khan
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| | - Joshua Letson
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| | - Emily Crowe
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| | - Amit Chougule
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| | - Patrick R Griffin
- The Wertheim UF Scripps Institute, University of Florida, Jupiter, FL 33458, USA.
| | | | - Beata Lecka-Czernik
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| |
Collapse
|
8
|
Fricke TC, Leffler A. TRPV2: a universal regulator in cellular physiology with a yet poorly defined thermosensitivity. J Physiol Sci 2024; 74:42. [PMID: 39285320 PMCID: PMC11403965 DOI: 10.1186/s12576-024-00936-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 08/31/2024] [Indexed: 09/22/2024]
Abstract
Transient receptor potential (TRP) ion channels serve as sensors for variations in ambient temperature, modulating both thermoregulation and temperature responsive cellular processes. Among these, the vanilloid TRP subfamily (TRPV) comprises six members and at least four of these members (TRPV1-TRPV4) have been associated with thermal sensation. TRPV2 has been described as a sensor for noxious heat, but subsequent studies have unveiled a more complex role for TRPV2 beyond temperature perception. This comprehensive review aims to elucidate the intricate thermosensitivity of TRPV2 by synthesizing current knowledge on its biophysical properties, expression pattern and known physiological functions associated with thermosensation.
Collapse
Affiliation(s)
- Tabea C Fricke
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Carl-Neuberg Strasse 1, 30625, Hannover, Germany
| | - Andreas Leffler
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Carl-Neuberg Strasse 1, 30625, Hannover, Germany.
| |
Collapse
|
9
|
Bolin AP, de Fatima Silva F, Salgueiro RB, Dos Santos BA, Komino ACM, Andreotti S, de Sousa É, de Castro É, Real CC, de Paula Faria D, Souza GP, Camara H, Sorgi CA, Tseng YH, Lima FB, Rodrigues AC. Glucocorticoid modulates oxidative and thermogenic function of rat brown adipose tissue and human brown adipocytes. J Cell Physiol 2024; 239:1-12. [PMID: 39091018 DOI: 10.1002/jcp.31397] [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: 03/25/2024] [Revised: 07/09/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024]
Abstract
Chronic and excessive glucocorticoid (GC) exposure can cause Cushing's syndrome, resulting in fat accumulation in selected body areas. Particularly in the brown adipose tissue (BAT), GC acts negatively, resulting in whitening of the tissue. We hypothesized that dysregulation of microRNAs by GC could be an additional mechanism to explain its negative actions in BAT. Male Wistar rats were divided into two groups: (1) Control sham and (2) GC group that was administered dexamethasone 6.25 mg/200 μL via osmotic pump implantation over 28 days. After this period, the animals were euthanized and BAT tissue was properly stored. Human fat cells treated with dexamethasone were used to translate the experimental results found in animals to human biology. GC-treated rat BAT presented with large lipid droplets, severely impaired thermogenic activation, and reduced glucose uptake measured by 18F-FDG PET/CT. GC exposure induced a reduction in the mitochondrial OXPHOS system and oxygen consumption. MicroRNA profiling of BAT revealed five top-regulated microRNAs and among them miR-21-5p was the most significantly upregulated in GC-treated rats compared to the control group. Although upregulation of miR-21-5p in the tissue, differentiated primary brown adipocytes from GC-treated rats had decreased miR-21-5p levels compared to the control group. To translate these results to the clinic, human brown adipocytes were treated with dexamethasone and miR-21-5p inhibitor. In human brown cells, inhibition of miR-21-5p increased brown adipocyte differentiation and prevented GC-induced glucose uptake, resulting in a lower glycolysis rate. In conclusion, high-dose GC therapy significantly impacts brown adipose tissue function, with a notable association between glucose uptake and miR-21-5p.
Collapse
Affiliation(s)
- Anaysa Paola Bolin
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Flaviane de Fatima Silva
- Department of Physiology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Rafael Barrera Salgueiro
- Department of Physiology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Bruna Araújo Dos Santos
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | | | - Sandra Andreotti
- Department of Physiology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Érica de Sousa
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Érique de Castro
- Department of Physiology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Caroline Cristiano Real
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Daniele de Paula Faria
- Department of Radiology and Oncology, Laboratory of Nuclear Medicine (LIM43), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Gerson Profeta Souza
- Department of Medicine, Section on Integrative Physiology and Metabolism, Joslin Diabetes Center Harvard Medical School, Boston, Massachusetts, USA
| | - Henrique Camara
- Department of Medicine, Section on Integrative Physiology and Metabolism, Joslin Diabetes Center Harvard Medical School, Boston, Massachusetts, USA
| | - Carlos Arterio Sorgi
- Department of Biochemistry and Immunology, Faculdade de Medicina de Ribeirão Preto - FMRP/USP, Ribeirão Preto, Brazil
- Department of Chemistry, Faculdade de Filosofia, Ciencias e Letras de Ribeirão Preto - FFCLRP/USP, Ribeirão Preto, Brazil
| | - Yu-Hua Tseng
- Department of Medicine, Section on Integrative Physiology and Metabolism, Joslin Diabetes Center Harvard Medical School, Boston, Massachusetts, USA
| | - Fábio Bessa Lima
- Department of Physiology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Alice Cristina Rodrigues
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
Fukushima SI, Wazawa T, Sugiura K, Nagai T. Extremely Sensitive Genetically Encoded Temperature Indicator Enabling Measurement at the Organelle Level. ACS Sens 2024; 9:3889-3897. [PMID: 39042704 PMCID: PMC11348412 DOI: 10.1021/acssensors.3c02658] [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: 12/11/2023] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/25/2024]
Abstract
Intracellular temperature is a fundamental parameter in biochemical reactions. Genetically encoded fluorescent temperature indicators (GETIs) have been developed to visualize intracellular thermogenesis; however, the temperature sensitivity or localization capability in specific organelles should have been further improved to clearly capture when and where intracellular temperature changes at the subcellular level occur. Here, we developed a new GETI, gMELT, composed of donor and acceptor subunits, in which cyan and yellow fluorescent proteins, respectively, as a Förster resonance energy transfer (FRET) pair were fused with temperature-sensitive domains. The donor and acceptor subunits associated and dissociated in response to temperature changes, altering the FRET efficiency. Consequently, gMELT functioned as a fluorescence ratiometric indicator. Untagged gMELT was expressed in the cytoplasm, whereas versions fused with specific localization signals were targeted to the endoplasmic reticulum (ER) or mitochondria. All gMELT variations enabled more sensitive temperature measurements in cellular compartments than those in previous GETIs. The gMELTs, tagged with ER or mitochondrial targeting sequences, were used to detect thermogenesis in organelles stimulated chemically, a method previously known to induce thermogenesis. The observed temperature changes were comparable to previous reports, assuming that the fluorescence readout changes were exclusively due to temperature variations. Furthermore, we demonstrated how macromolecular crowding influences gMELT fluorescence given that this factor can subtly affect the fluorescence readout. Investigating thermogenesis with gMELT, accounting for factors such as macromolecular crowding, will enhance our understanding of intracellular thermogenesis phenomena.
Collapse
Affiliation(s)
| | | | | | - Takeharu Nagai
- SANKEN, The University of Osaka, Ibaraki, Osaka 567-0047, Japan
- OTRI, The University of Osaka, Suita, Osaka 565-0871, Japan
- Research
Institute for Electronic Science, Hokkaido
University, Sapporo, Hokkaido 001-0021, Japan
| |
Collapse
|
11
|
Wang CH, Tsuji T, Wu LH, Yang CY, Huang TL, Sato M, Shamsi F, Tseng YH. Endothelin 3/EDNRB signaling induces thermogenic differentiation of white adipose tissue. Nat Commun 2024; 15:7215. [PMID: 39174539 PMCID: PMC11341701 DOI: 10.1038/s41467-024-51579-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/12/2024] [Indexed: 08/24/2024] Open
Abstract
Thermogenic adipose tissue, consisting of brown and beige fat, regulates nutrient utilization and energy metabolism. Human brown fat is relatively scarce and decreases with obesity and aging. Hence, inducing thermogenic differentiation of white fat offers an attractive way to enhance whole-body metabolic capacity. Here, we show the role of endothelin 3 (EDN3) and endothelin receptor type B (EDNRB) in promoting the browning of white adipose tissue (WAT). EDNRB overexpression stimulates thermogenic differentiation of human white preadipocytes through cAMP-EPAC1-ERK activation. In mice, cold induces the expression of EDN3 and EDNRB in WAT. Deletion of EDNRB in adipose progenitor cells impairs cold-induced beige adipocyte formation in WAT, leading to excessive weight gain, glucose intolerance, and insulin resistance upon high-fat feeding. Injection of EDN3 into WAT promotes browning and improved whole-body glucose metabolism. The findings shed light on the mechanism of WAT browning and offer potential therapeutics for obesity and metabolic disorders.
Collapse
Affiliation(s)
- Chih-Hao Wang
- Graduate Institute of Cell Biology, China Medical University, Taichung City, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City, Taiwan.
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| | - Tadataka Tsuji
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Li-Hong Wu
- Graduate Institute of Cell Biology, China Medical University, Taichung City, Taiwan
| | - Cheng-Ying Yang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City, Taiwan
| | - Tian Lian Huang
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Mari Sato
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Farnaz Shamsi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
12
|
Dukic B, Ruppert Z, Tóth ME, Hunya Á, Czibula Á, Bíró P, Tiszlavicz Á, Péter M, Balogh G, Erdélyi M, Timinszky G, Vígh L, Gombos I, Török Z. Mild Hyperthermia-Induced Thermogenesis in the Endoplasmic Reticulum Defines Stress Response Mechanisms. Cells 2024; 13:1141. [PMID: 38994992 PMCID: PMC11240596 DOI: 10.3390/cells13131141] [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: 06/05/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/13/2024] Open
Abstract
Previous studies reported that a mild, non-protein-denaturing, fever-like temperature increase induced the unfolded protein response (UPR) in mammalian cells. Our dSTORM super-resolution microscopy experiments revealed that the master regulator of the UPR, the IRE1 (inositol-requiring enzyme 1) protein, is clustered as a result of UPR activation in a human osteosarcoma cell line (U2OS) upon mild heat stress. Using ER thermo yellow, a temperature-sensitive fluorescent probe targeted to the endoplasmic reticulum (ER), we detected significant intracellular thermogenesis in mouse embryonic fibroblast (MEF) cells. Temperatures reached at least 8 °C higher than the external environment (40 °C), resulting in exceptionally high ER temperatures similar to those previously described for mitochondria. Mild heat-induced thermogenesis in the ER of MEF cells was likely due to the uncoupling of the Ca2+/ATPase (SERCA) pump. The high ER temperatures initiated a pronounced cytosolic heat-shock response in MEF cells, which was significantly lower in U2OS cells in which both the ER thermogenesis and SERCA pump uncoupling were absent. Our results suggest that depending on intrinsic cellular properties, mild hyperthermia-induced intracellular thermogenesis defines the cellular response mechanism and determines the outcome of hyperthermic stress.
Collapse
Affiliation(s)
- Barbara Dukic
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.D.); (L.V.); (I.G.)
- Doctoral School of Environmental Sciences, Faculty of Science and Informatics, University of Szeged, 6720 Szeged, Hungary
| | - Zsófia Ruppert
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.D.); (L.V.); (I.G.)
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, 6720 Szeged, Hungary
| | - Melinda E. Tóth
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.D.); (L.V.); (I.G.)
| | - Ákos Hunya
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.D.); (L.V.); (I.G.)
| | - Ágnes Czibula
- Laboratory of DNA Damage and Nuclear Dynamics, Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
- Department of Immunology, University of Szeged, 6720 Szeged, Hungary
| | - Péter Bíró
- Department of Optics and Quantum Electronics, University of Szeged, 6720 Szeged, Hungary
| | - Ádám Tiszlavicz
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.D.); (L.V.); (I.G.)
| | - Mária Péter
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.D.); (L.V.); (I.G.)
| | - Gábor Balogh
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.D.); (L.V.); (I.G.)
| | - Miklós Erdélyi
- Department of Optics and Quantum Electronics, University of Szeged, 6720 Szeged, Hungary
| | - Gyula Timinszky
- Laboratory of DNA Damage and Nuclear Dynamics, Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - László Vígh
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.D.); (L.V.); (I.G.)
| | - Imre Gombos
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.D.); (L.V.); (I.G.)
| | - Zsolt Török
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary; (B.D.); (L.V.); (I.G.)
| |
Collapse
|
13
|
Kołodziej T, Mrózek M, Sengottuvel S, Głowacki MJ, Ficek M, Gawlik W, Rajfur Z, Wojciechowski AM. Multimodal analysis of traction forces and the temperature dynamics of living cells with a diamond-embedded substrate. BIOMEDICAL OPTICS EXPRESS 2024; 15:4024-4043. [PMID: 39022544 PMCID: PMC11249686 DOI: 10.1364/boe.524293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 07/20/2024]
Abstract
Cells and tissues are constantly exposed to chemical and physical signals that regulate physiological and pathological processes. This study explores the integration of two biophysical methods: traction force microscopy (TFM) and optically detected magnetic resonance (ODMR) to concurrently assess cellular traction forces and the local relative temperature. We present a novel elastic substrate with embedded nitrogen-vacancy microdiamonds that facilitate ODMR-TFM measurements. Optimization efforts focused on minimizing sample illumination and experiment duration to mitigate biological perturbations. Our hybrid ODMR-TFM technique yields TFM maps and achieves approximately 1 K precision in relative temperature measurements. Our setup employs a simple wide-field fluorescence microscope with standard components, demonstrating the feasibility of the proposed technique in life science laboratories. By elucidating the physical aspects of cellular behavior beyond the existing methods, this approach opens avenues for a deeper understanding of cellular processes and may inspire the development of diverse biomedical applications.
Collapse
Affiliation(s)
- Tomasz Kołodziej
- Jagiellonian University Medical School, Faculty of Pharmacy, Kraków, Poland
- Jagiellonian University , Faculty of Physics, Astronomy, and Applied Computer Science, Kraków, Poland
| | - Mariusz Mrózek
- Jagiellonian University , Faculty of Physics, Astronomy, and Applied Computer Science, Kraków, Poland
| | - Saravanan Sengottuvel
- Jagiellonian University , Faculty of Physics, Astronomy, and Applied Computer Science, Kraków, Poland
- Jagiellonian University, Doctoral School of Exact and Natural Sciences, Kraków, Poland
| | - Maciej J Głowacki
- Gdansk University of Technology, Faculty of Electronics, Telecommunications, and Informatics, Department of Metrology and Optoelectronics, Gdańsk, Poland
| | - Mateusz Ficek
- Gdansk University of Technology, Faculty of Electronics, Telecommunications, and Informatics, Department of Metrology and Optoelectronics, Gdańsk, Poland
| | - Wojciech Gawlik
- Jagiellonian University , Faculty of Physics, Astronomy, and Applied Computer Science, Kraków, Poland
| | - Zenon Rajfur
- Jagiellonian University , Faculty of Physics, Astronomy, and Applied Computer Science, Kraków, Poland
| | - Adam M Wojciechowski
- Jagiellonian University , Faculty of Physics, Astronomy, and Applied Computer Science, Kraków, Poland
| |
Collapse
|
14
|
Kang MG, Kim HR, Lee HY, Kwak C, Koh H, Kang BH, Roe JS, Rhee HW. Mitochondrial Thermogenesis Can Trigger Heat Shock Response in the Nucleus. ACS CENTRAL SCIENCE 2024; 10:1231-1241. [PMID: 38947196 PMCID: PMC11212142 DOI: 10.1021/acscentsci.3c01589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/05/2024] [Accepted: 05/15/2024] [Indexed: 07/02/2024]
Abstract
Mitochondrial thermogenesis is a process in which heat is generated by mitochondrial respiration. In living organisms, the thermogenic mechanisms that maintain body temperature have been studied extensively in fat cells with little knowledge on how mitochondrial heat may act beyond energy expenditure. Here, we highlight that the exothermic oxygen reduction reaction (ΔH f° = -286 kJ/mol) is the main source of the protonophore-induced mitochondrial thermogenesis, and this heat is conducted to other cellular organelles, including the nucleus. As a result, mitochondrial heat that reached the nucleus initiated the classical heat shock response, including the formation of nuclear stress granules and the localization of heat shock factor 1 (HSF1) to chromatin. Consequently, activated HSF1 increases the level of gene expression associated with the response to thermal stress in mammalian cells. Our results illustrate heat generated within the cells as a potential source of mitochondria-nucleus communication and expand our understanding of the biological functions of mitochondria in cell physiology.
Collapse
Affiliation(s)
- Myeong-Gyun Kang
- Department
of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Hwa-Ryeon Kim
- Department
of Biochemistry, Yonsei University, Seoul 03722, Korea
| | - Hee Yong Lee
- Department
of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Chulhwan Kwak
- Department
of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Hyewon Koh
- Department
of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Byoung Heon Kang
- Department
of Biological Sciences, Ulsan National Institute
of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Jae-Seok Roe
- Department
of Biochemistry, Yonsei University, Seoul 03722, Korea
| | - Hyun-Woo Rhee
- Department
of Chemistry, Seoul National University, Seoul 08826, Korea
- School
of Biological Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
15
|
Stafeev I, Agareva M, Michurina S, Tomilova A, Shestakova E, Zubkova E, Sineokaya M, Ratner E, Menshikov M, Parfyonova Y, Shestakova M. Semaglutide 6-months therapy of type 2 diabetes mellitus restores adipose progenitors potential to develop metabolically active adipocytes. Eur J Pharmacol 2024; 970:176476. [PMID: 38493915 DOI: 10.1016/j.ejphar.2024.176476] [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: 12/11/2023] [Revised: 02/20/2024] [Accepted: 03/01/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Nowadays type 2 diabetes mellitus (T2DM) leads to population mortality growth. Today glucagon-like peptide type 1 receptor agonists (GLP-1 RA) are one of the most promising glucose-lowered drugs with anorexigenic and cardioprotective effects. The present study aims to determine the effects of GLP-1 RA semaglutide 6-month therapy on T2DM patient metabolic parameters and adipose progenitor cell health. METHODS T2DM patients (N = 8) underwent clinical characterization and subcutaneous fat biopsy at start point and after semaglutide 6-month therapy. Adipose-derived stem cells (ADSC) were isolated by enzymatic method. Cell proliferation analysis was performed by MTT and immunocytochemistry. White and beige adipogenesis was analyzed by BODIPY493/503 staining and confocal microscopy. Adipocyte's metabolic properties were estimated by 3H- and 14C-based metabolic assays. Thermogenesis analysis was performed by ERthermAC staining and confocal microscopy. Protein markers were assessed by Western blotting. RESULTS Semaglutide 6-month therapy demonstrated significant anorexigenic and glucose-lowering effects. However, insulin sensitivity (HOMA-IR and M-index) was unchanged after therapy. Semaglutide 6-month therapy increased ADSC proliferation and white and beige adipogenesis. Moreover, lipid droplets fragmentation was observed in beige adipocytes. Both white and beige adipocytes after semaglutide therapy demonstrated 2-3 fold growth of glucose uptake without changes in insulin sensitivity. Newly formed white adipocytes demonstrated glucose utilization for active ATP synthesis, whereas beige adipocytes for canonical thermogenesis. CONCLUSIONS Our study has revealed that semaglutide 6-month therapy has not only systemic anorexigenic effects, but can markedly improve adipose tissue health. We have demonstrated critical restoration of ADSC renewal functions, which potentially can be involved in semaglutide based weight loss.
Collapse
Affiliation(s)
- I Stafeev
- National Medical Research Centre of Cardiology Named After Academician E.I.Chazov, 121552, Moscow, Russia.
| | - M Agareva
- National Medical Research Centre of Cardiology Named After Academician E.I.Chazov, 121552, Moscow, Russia; Lomonosov Moscow State University, 119991, Moscow, Russia
| | - S Michurina
- National Medical Research Centre of Cardiology Named After Academician E.I.Chazov, 121552, Moscow, Russia; Lomonosov Moscow State University, 119991, Moscow, Russia
| | - A Tomilova
- Endocrinology Research Centre, 117292, Moscow, Russia
| | - E Shestakova
- Endocrinology Research Centre, 117292, Moscow, Russia
| | - E Zubkova
- National Medical Research Centre of Cardiology Named After Academician E.I.Chazov, 121552, Moscow, Russia
| | - M Sineokaya
- Endocrinology Research Centre, 117292, Moscow, Russia
| | - E Ratner
- National Medical Research Centre of Cardiology Named After Academician E.I.Chazov, 121552, Moscow, Russia
| | - M Menshikov
- National Medical Research Centre of Cardiology Named After Academician E.I.Chazov, 121552, Moscow, Russia
| | - Ye Parfyonova
- National Medical Research Centre of Cardiology Named After Academician E.I.Chazov, 121552, Moscow, Russia; Lomonosov Moscow State University, 119991, Moscow, Russia
| | - M Shestakova
- Lomonosov Moscow State University, 119991, Moscow, Russia; Endocrinology Research Centre, 117292, Moscow, Russia
| |
Collapse
|
16
|
Baroi S, Czernik PJ, Khan MP, Letson J, Crowe E, Chougule A, Griffin PR, Rosen CJ, Lecka-Czernik B. PPARG in osteocytes controls cell bioenergetics and systemic energy metabolism independently of sclerostin levels in circulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588029. [PMID: 38645043 PMCID: PMC11030235 DOI: 10.1101/2024.04.04.588029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Objective The skeleton is one of the largest organs in the body, wherein metabolism is integrated with systemic energy metabolism. However, the bioenergetic programming of osteocytes, the most abundant bone cells coordinating bone metabolism, is not well defined. Here, using a mouse model with partial penetration of an osteocyte-specific PPARG deletion, we demonstrate that PPARG controls osteocyte bioenergetics and their contribution to systemic energy metabolism independently of circulating sclerostin levels. Methods In vivo and in vitro models of osteocyte-specific PPARG deletion, i.e. Dmp 1 Cre Pparγ flfl male and female mice (γOT KO ) and MLO-Y4 osteocyte-like cells with either siRNA-silenced or CRISPR/Cas9-edited Pparγ . As applicable, the models were analyzed for levels of energy metabolism, glucose metabolism, and metabolic profile of extramedullary adipose tissue, as well as the osteocyte transcriptome, mitochondrial function, bioenergetics, insulin signaling, and oxidative stress. Results Circulating sclerostin levels of γOT KO male and female mice were not different from control mice. Male γOT KO mice exhibited a high energy phenotype characterized by increased respiration, heat production, locomotion and food intake. This high energy phenotype in males did not correlate with "beiging" of peripheral adipose depots. However, both sexes showed a trend for reduced fat mass and apparent insulin resistance without changes in glucose tolerance, which correlated with decreased osteocytic responsiveness to insulin measured by AKT activation. The transcriptome of osteocytes isolated from γOT KO males suggested profound changes in cellular metabolism, fuel transport and usage, mitochondria dysfunction, insulin signaling and increased oxidative stress. In MLO-Y4 osteocytes, PPARG deficiency correlated with highly active mitochondria, increased ATP production, shifts in fuel utilization, and accumulation of reactive oxygen species (ROS). Conclusions PPARG in male osteocytes acts as a molecular break on mitochondrial function, and protection against oxidative stress and ROS accumulation. It also regulates osteocyte insulin signaling and fuel usage to produce energy. These data provide insight into the connection between osteocyte bioenergetics and their sex-specific contribution to the balance of systemic energy metabolism. These findings support the concept that the skeleton controls systemic energy expenditure via osteocyte metabolism. Highlights Osteocytes function as a body energostat via their bioenergeticsPPARG protein acts as a "molecular break" of osteocyte mitochondrial activityPPARG deficiency activates TCA cycle, oxidative stress and ROS accumulationPPARG controls osteocyte insulin signaling and fuel utilization.
Collapse
|
17
|
Li S, Li Y, Zhang S, Fang H, Huang Z, Zhang D, Ding A, Uvdal K, Hu Z, Huang K, Li L. Response strategies and biological applications of organic fluorescent thermometry: cell- and mitochondrion-level detection. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:1968-1984. [PMID: 38511286 DOI: 10.1039/d4ay00117f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Temperature homeostasis is critical for cells to perform their physiological functions. Among the diverse methods for temperature detection, fluorescent temperature probes stand out as a proven and effective tool, especially for monitoring temperature in cells and suborganelles, with a specific emphasis on mitochondria. The utilization of these probes provides a new opportunity to enhance our understanding of the mechanisms and interconnections underlying various physiological activities related to temperature homeostasis. However, the complexity and variability of cells and suborganelles necessitate fluorescent temperature probes with high resolution and sensitivity. To meet the demanding requirements for intracellular/subcellular temperature detection, several strategies have been developed, offering a range of options to address this challenge. This review examines four fundamental temperature-response strategies employed by small molecule and polymer probes, including intramolecular rotation, polarity sensitivity, Förster resonance energy transfer, and structural changes. The primary emphasis was placed on elucidating molecular design and biological applications specific to each type of probe. Furthermore, this review provides an insightful discussion on factors that may affect fluorescent thermometry, providing valuable perspectives for future development in the field. Finally, the review concludes by presenting cutting-edge response strategies and research insights for mitigating biases in temperature sensing.
Collapse
Affiliation(s)
- Shuai Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Yaoxuan Li
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Shiji Zhang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Haixiao Fang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
- Future Display Institute in Xiamen, Xiamen 361005, China.
| | - Ze Huang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Duoteng Zhang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Aixiang Ding
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Kajsa Uvdal
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, 58183, Sweden.
| | - Zhangjun Hu
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, 58183, Sweden.
| | - Kai Huang
- Future Display Institute in Xiamen, Xiamen 361005, China.
| | - Lin Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
- Future Display Institute in Xiamen, Xiamen 361005, China.
| |
Collapse
|
18
|
Morocho-Jaramillo PA, Kotlar-Goldaper I, Zakarauskas-Seth BI, Purfürst B, Filosa A, Sawamiphak S. The zebrafish heart harbors a thermogenic beige fat depot analog of human epicardial adipose tissue. Cell Rep 2024; 43:113955. [PMID: 38507414 DOI: 10.1016/j.celrep.2024.113955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 01/25/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Epicardial adipose tissue (eAT) is a metabolically active fat depot that has been associated with a wide array of cardiac homeostatic functions and cardiometabolic diseases. A full understanding of its diverse physiological and pathological roles is hindered by the dearth of animal models. Here, we show, in the heart of an ectothermic teleost, the zebrafish, the existence of a fat depot localized underneath the epicardium, originating from the epicardium and exhibiting the molecular signature of beige adipocytes. Moreover, a subset of adipocytes within this cardiac fat tissue exhibits primitive thermogenic potential. Transcriptomic profiling and cross-species analysis revealed elevated glycolytic and cardiac homeostatic gene expression with downregulated obesity and inflammatory hallmarks in the teleost eAT compared to that of lean aged humans. Our findings unveil epicardium-derived beige fat in the heart of an ectotherm considered to possess solely white adipocytes for energy storage and identify pathways that may underlie age-driven remodeling of human eAT.
Collapse
Affiliation(s)
- Paul-Andres Morocho-Jaramillo
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Ilan Kotlar-Goldaper
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Bhakti I Zakarauskas-Seth
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Bettina Purfürst
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Alessandro Filosa
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Suphansa Sawamiphak
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany.
| |
Collapse
|
19
|
Cheng Y, Wu J, Cui Y, Zhai J, Wu M, Xie X. Photoswitchable Temperature Nanosensors Based on the Chemical Kinetics of Photochromic Naphthopyran for Live Cell Imaging. Anal Chem 2024; 96:4605-4611. [PMID: 38457774 DOI: 10.1021/acs.analchem.3c05568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2024]
Abstract
Microscopic temperature imaging holds significant importance in various fields, particularly in the development of nanomaterials for photothermal therapy (PTT). In this study, we present an analytical method to probe cellular temperature based on chemical kinetics and additional luminescence quenching by photoswitchable naphthopyrans. Taking advantage of the rapid ring-closing reaction of naphthopyran, temperature sensing was realized with a linear relationship between the logarithmic decay time constant (ln τ) and the reciprocal temperature (T-1). To create luminescent temperature nanosensors, we harnessed the ability of ring-opened naphthopyran to quench the luminescence of a semiconducting polymer, resulting in a diverse array of probes. Structural modifications on the naphthopyran also provided a way to fine-tune the sensitivity and response window of the nanosensors. The method allowed cellular temperature imaging on a cost-effective fluorescence microscopic setup. As an application, the temperature increase induced by gold nanorods (AuNRs) in cell lysosomes was successfully monitored, laying the foundation for a new class of photoswitchable nanosensors with promising biological applications.
Collapse
Affiliation(s)
- Yu Cheng
- Department of Chemistry, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jianhong Wu
- Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yunxin Cui
- Department of Chemistry, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jingying Zhai
- Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Minghui Wu
- Department of Chemistry, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Xiaojiang Xie
- Department of Chemistry, Southern University of Science and Technology, Shenzhen, 518055, China
| |
Collapse
|
20
|
Ma J, Sun R, Xia K, Xia Q, Liu Y, Zhang X. Design and Application of Fluorescent Probes to Detect Cellular Physical Microenvironments. Chem Rev 2024; 124:1738-1861. [PMID: 38354333 DOI: 10.1021/acs.chemrev.3c00573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
The microenvironment is indispensable for functionality of various biomacromolecules, subcellular compartments, living cells, and organisms. In particular, physical properties within the biological microenvironment could exert profound effects on both the cellular physiology and pathology, with parameters including the polarity, viscosity, pH, and other relevant factors. There is a significant demand to directly visualize and quantitatively measure the fluctuation in the cellular microenvironment with spatiotemporal resolution. To satisfy this need, analytical methods based on fluorescence probes offer great opportunities due to the facile, sensitive, and dynamic detection that these molecules could enable in varying biological settings from in vitro samples to live animal models. Herein, we focus on various types of small molecule fluorescent probes for the detection and measurement of physical parameters of the microenvironment, including pH, polarity, viscosity, mechanical force, temperature, and electron potential. For each parameter, we primarily describe the chemical mechanisms underlying how physical properties are correlated with changes of various fluorescent signals. This review provides both an overview and a perspective for the development of small molecule fluorescent probes to visualize the dynamic changes in the cellular environment, to expand the knowledge for biological process, and to enrich diagnostic tools for human diseases.
Collapse
Affiliation(s)
- Junbao Ma
- Department of Chemistry and Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou 310030, Zhejiang Province, China
- Westlake Laboratory of Life Sciences and Biomedicine, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
- Institute of Natural Sciences, Westlake Institute for Advanced Study, Hangzhou 310030, Zhejiang Province, China
| | - Rui Sun
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- University of the Chinese Academy of Sciences, 19 A Yuquan Road, Shijingshan District, Beijing 100049, China
| | - Kaifu Xia
- Department of Chemistry and Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou 310030, Zhejiang Province, China
- Westlake Laboratory of Life Sciences and Biomedicine, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
- Institute of Natural Sciences, Westlake Institute for Advanced Study, Hangzhou 310030, Zhejiang Province, China
| | - Qiuxuan Xia
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- University of the Chinese Academy of Sciences, 19 A Yuquan Road, Shijingshan District, Beijing 100049, China
| | - Yu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, Chinese Academy of Sciences Dalian Liaoning 116023, China
| | - Xin Zhang
- Department of Chemistry and Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou 310030, Zhejiang Province, China
- Westlake Laboratory of Life Sciences and Biomedicine, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| |
Collapse
|
21
|
Michurina S, Agareva M, Zubkova E, Menshikov M, Stafeev I, Parfyonova Y. IL-4 activates the futile triacylglyceride cycle for glucose utilization in white adipocytes. Biochem J 2024; 481:329-344. [PMID: 38323641 DOI: 10.1042/bcj20230486] [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: 12/01/2023] [Revised: 01/22/2024] [Accepted: 02/06/2024] [Indexed: 02/08/2024]
Abstract
The development of cardiometabolic complications during obesity is strongly associated with chronic latent inflammation in hypertrophied adipose tissue (AT). IL-4 is an anti-inflammatory cytokine, playing a protective role against insulin resistance, glucose intolerance and weight gain. The positive effects of IL-4 are associated not only with the activation of anti-inflammatory immune cells in AT, but also with the modulation of adipocyte metabolism. IL-4 is known to activate lipolysis and glucose uptake in adipocytes, but the precise regulatory mechanisms and physiological significance of these processes remain unclear. In this study, we detail IL-4 effects on glucose and triacylglycerides (TAGs) metabolism and propose mechanisms of IL-4 metabolic action in adipocytes. We have shown that IL-4 activates glucose oxidation, lipid droplet (LD) fragmentation, lipolysis and thermogenesis in mature 3T3-L1 adipocytes. We found that lipolysis was not accompanied by fatty acids (FAs) release from adipocytes, suggesting FA re-esterification. Moreover, glucose oxidation and thermogenesis stimulation depended on adipocyte triglyceride lipase (ATGL) activity, but not the uncoupling protein (UCP1) expression. Based on these data, IL-4 may activate the futile TAG-FA cycle in adipocytes, which enhances the oxidative activity of cells and heat production. Thus, the positive effect of IL-4 on systemic metabolism can be the result of the activation of non-canonical thermogenic mechanism in AT, increasing TAG turnover and utilization of excessive glucose.
Collapse
Affiliation(s)
- Svetlana Michurina
- Department of Angiogenesis, National Medical Research Centre for Cardiology named after academician E.I.Chazov, 121552, Moscow, Russia
| | - Margarita Agareva
- Department of Angiogenesis, National Medical Research Centre for Cardiology named after academician E.I.Chazov, 121552, Moscow, Russia
- Faculty of Basic Medicine, Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Ekaterina Zubkova
- Department of Angiogenesis, National Medical Research Centre for Cardiology named after academician E.I.Chazov, 121552, Moscow, Russia
| | - Mikhail Menshikov
- Department of Angiogenesis, National Medical Research Centre for Cardiology named after academician E.I.Chazov, 121552, Moscow, Russia
| | - Iurii Stafeev
- Department of Angiogenesis, National Medical Research Centre for Cardiology named after academician E.I.Chazov, 121552, Moscow, Russia
| | - Yelena Parfyonova
- Department of Angiogenesis, National Medical Research Centre for Cardiology named after academician E.I.Chazov, 121552, Moscow, Russia
- Faculty of Basic Medicine, Lomonosov Moscow State University, 119991, Moscow, Russia
| |
Collapse
|
22
|
Terzioglu M, Veeroja K, Montonen T, Ihalainen TO, Salminen TS, Bénit P, Rustin P, Chang YT, Nagai T, Jacobs HT. Mitochondrial temperature homeostasis resists external metabolic stresses. eLife 2023; 12:RP89232. [PMID: 38079477 PMCID: PMC10712956 DOI: 10.7554/elife.89232] [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] [Indexed: 12/18/2023] Open
Abstract
Based on studies with a fluorescent reporter dye, Mito Thermo Yellow (MTY), and the genetically encoded gTEMP ratiometric fluorescent temperature indicator targeted to mitochondria, the temperature of active mitochondria in four mammalian and one insect cell line was estimated to be up to 15°C above that of the external environment to which the cells were exposed. High mitochondrial temperature was maintained in the face of a variety of metabolic stresses, including substrate starvation or modification, decreased ATP demand due to inhibition of cytosolic protein synthesis, inhibition of the mitochondrial adenine nucleotide transporter and, if an auxiliary pathway for electron transfer was available via the alternative oxidase, even respiratory poisons acting downstream of oxidative phosphorylation (OXPHOS) complex I. We propose that the high temperature of active mitochondria is an inescapable consequence of the biochemistry of OXPHOS and is homeostatically maintained as a primary feature of mitochondrial metabolism.
Collapse
Affiliation(s)
- Mügen Terzioglu
- Faculty of Medicine and Health Technology, Tampere UniversityTampereFinland
| | - Kristo Veeroja
- Faculty of Medicine and Health Technology, Tampere UniversityTampereFinland
| | - Toni Montonen
- Faculty of Medicine and Health Technology, Tampere UniversityTampereFinland
| | - Teemu O Ihalainen
- Faculty of Medicine and Health Technology, Tampere UniversityTampereFinland
| | - Tiina S Salminen
- Faculty of Medicine and Health Technology, Tampere UniversityTampereFinland
| | - Paule Bénit
- Université Paris Cité, Inserm, Maladies Neurodéveloppementales et NeurovasculairesParisFrance
| | - Pierre Rustin
- Université Paris Cité, Inserm, Maladies Neurodéveloppementales et NeurovasculairesParisFrance
| | - Young-Tae Chang
- SANKEN (The Institute of Scientific and Industrial Research), Osaka UniversityIbarakiJapan
| | | | - Howard T Jacobs
- Faculty of Medicine and Health Technology, Tampere UniversityTampereFinland
- Department of Environment and Genetics, La Trobe UniversityMelbourneAustralia
| |
Collapse
|
23
|
Kruglov AG, Romshin AM, Nikiforova AB, Plotnikova A, Vlasov II. Warm Cells, Hot Mitochondria: Achievements and Problems of Ultralocal Thermometry. Int J Mol Sci 2023; 24:16955. [PMID: 38069275 PMCID: PMC10707128 DOI: 10.3390/ijms242316955] [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: 11/10/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Temperature is a crucial regulator of the rate and direction of biochemical reactions and cell processes. The recent data indicating the presence of local thermal gradients associated with the sites of high-rate thermogenesis, on the one hand, demonstrate the possibility for the existence of "thermal signaling" in a cell and, on the other, are criticized on the basis of thermodynamic calculations and models. Here, we review the main thermometric techniques and sensors developed for the determination of temperature inside living cells and diverse intracellular compartments. A comparative analysis is conducted of the results obtained using these methods for the cytosol, nucleus, endo-/sarcoplasmic reticulum, and mitochondria, as well as their biological consistency. Special attention is given to the limitations, possible sources of errors and ambiguities of the sensor's responses. The issue of biological temperature limits in cells and organelles is considered. It is concluded that the elaboration of experimental protocols for ultralocal temperature measurements that take into account both the characteristics of biological systems, as well as the properties and limitations of each type of sensor is of critical importance for the generation of reliable results and further progress in this field.
Collapse
Affiliation(s)
- Alexey G. Kruglov
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Alexey M. Romshin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Anna B. Nikiforova
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Arina Plotnikova
- Institute for Physics and Engineering in Biomedicine, National Research Nuclear University MEPhI (Moscow Engineering Physics Institute MEPhI), 115409 Moscow, Russia;
| | - Igor I. Vlasov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 119991 Moscow, Russia;
| |
Collapse
|
24
|
Rao J, Djeffal Y, Chal J, Marchianò F, Wang CH, Al Tanoury Z, Gapon S, Mayeuf-Louchart A, Glass I, Sefton EM, Habermann B, Kardon G, Watt FM, Tseng YH, Pourquié O. Reconstructing human brown fat developmental trajectory in vitro. Dev Cell 2023; 58:2359-2375.e8. [PMID: 37647896 PMCID: PMC10873093 DOI: 10.1016/j.devcel.2023.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 08/23/2022] [Accepted: 08/01/2023] [Indexed: 09/01/2023]
Abstract
Brown adipocytes (BAs) represent a specialized cell type that is able to uncouple nutrient catabolism from ATP generation to dissipate energy as heat. In humans, the brown fat tissue is composed of discrete depots found throughout the neck and trunk region. BAs originate from a precursor common to skeletal muscle, but their developmental trajectory remains poorly understood. Here, we used single-cell RNA sequencing to characterize the development of interscapular brown fat in mice. Our analysis identified a transient stage of BA differentiation characterized by the expression of the transcription factor GATA6. We show that recapitulating the sequence of signaling cues identified in mice can lead to efficient differentiation of BAs in vitro from human pluripotent stem cells. These precursors can in turn be efficiently converted into functional BAs that can respond to signals mimicking adrenergic stimuli by increasing their metabolism, resulting in heat production.
Collapse
Affiliation(s)
- Jyoti Rao
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Yannis Djeffal
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Jerome Chal
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Fabio Marchianò
- Aix-Marseille University, CNRS, IBDM, The Turing Center for Living Systems, 13009 Marseille, France
| | - Chih-Hao Wang
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ziad Al Tanoury
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Svetlana Gapon
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | | | - Ian Glass
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Elizabeth M Sefton
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Bianca Habermann
- Aix-Marseille University, CNRS, IBDM, The Turing Center for Living Systems, 13009 Marseille, France
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Fiona M Watt
- King's College London Centre for Stem Cells and Regenerative Medicine, Great Maze Pond, London SE1 9RT, UK
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
25
|
Hu D, Tan M, Lu D, Kleiboeker B, Liu X, Park H, Kravitz AV, Shoghi KI, Tseng YH, Razani B, Ikeda A, Lodhi IJ. TMEM135 links peroxisomes to the regulation of brown fat mitochondrial fission and energy homeostasis. Nat Commun 2023; 14:6099. [PMID: 37773161 PMCID: PMC10541902 DOI: 10.1038/s41467-023-41849-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 09/20/2023] [Indexed: 10/01/2023] Open
Abstract
Mitochondrial morphology, which is controlled by mitochondrial fission and fusion, is an important regulator of the thermogenic capacity of brown adipocytes. Adipose-specific peroxisome deficiency impairs thermogenesis by inhibiting cold-induced mitochondrial fission due to decreased mitochondrial membrane content of the peroxisome-derived lipids called plasmalogens. Here, we identify TMEM135 as a critical mediator of the peroxisomal regulation of mitochondrial fission and thermogenesis. Adipose-specific TMEM135 knockout in mice blocks mitochondrial fission, impairs thermogenesis, and increases diet-induced obesity and insulin resistance. Conversely, TMEM135 overexpression promotes mitochondrial division, counteracts obesity and insulin resistance, and rescues thermogenesis in peroxisome-deficient mice. Mechanistically, thermogenic stimuli promote association between peroxisomes and mitochondria and plasmalogen-dependent localization of TMEM135 in mitochondria, where it mediates PKA-dependent phosphorylation and mitochondrial retention of the fission factor Drp1. Together, these results reveal a previously unrecognized inter-organelle communication regulating mitochondrial fission and energy homeostasis and identify TMEM135 as a potential target for therapeutic activation of BAT.
Collapse
Affiliation(s)
- Donghua Hu
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Min Tan
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Dongliang Lu
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian Kleiboeker
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Xuejing Liu
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Hongsuk Park
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexxai V Kravitz
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Kooresh I Shoghi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Babak Razani
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
26
|
Choi J, Kim DI, Kim JY, Pané S, Nelson BJ, Chang YT, Choi H. Magnetically Enhanced Intracellular Uptake of Superparamagnetic Iron Oxide Nanoparticles for Antitumor Therapy. ACS NANO 2023; 17:15857-15870. [PMID: 37477428 DOI: 10.1021/acsnano.3c03780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have been widely employed in biomedical fields, including targeted delivery of antitumor therapy. Conventional magnetic tumor targeting has used simple static magnetic fields (SMFs), which cause SPIONs to linearly aggregate into a long chain-like shape. Such agglomeration greatly hinders the intracellular targeting of SPIONs into tumors, thus reducing the therapeutic efficacy. In this study, we investigated the enhancement of the intracellular uptake of SPIONs through the application of rotating magnetic fields (RMFs). Based on the physical principles of SPION chain disassembly, we investigated physical parameters to predict the chain length favorable for intracellular uptake. Our prediction was validated by clear visualization of the intracellular distributions of SPIONs in tumor cells at both cellular and three-dimensional microtissue levels. To identify the potential therapeutic effects of enhanced intracellular uptake, magnetic hyperthermia as antitumor therapy was investigated under varying conditions of magnetic hyperthermia and RMFs. The results showed that enhanced intracellular uptake reduced magnetic hyperthermia time and strength as well as particle concentration. The proposed method will be useful in the development of techniques to determine the optimized physical conditions for the enhanced intracellular uptake of SPIONs in antitumor therapy.
Collapse
Affiliation(s)
- Junhee Choi
- Department of Robotics and Mechatronics Engineering, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- DGIST-ETH Microrobotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Dong-In Kim
- Department of Robotics and Mechatronics Engineering, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- DGIST-ETH Microrobotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Jin-Young Kim
- Department of Robotics and Mechatronics Engineering, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- DGIST-ETH Microrobotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- Robotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- Division of Biotechnology, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- IMsystem Co., Ltd., Daegu 42988, Republic of Korea
| | - Salvador Pané
- DGIST-ETH Microrobotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- Multi-Scale Robotics Lab, Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich CH-8092, Switzerland
| | - Bradley J Nelson
- DGIST-ETH Microrobotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- Multi-Scale Robotics Lab, Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich CH-8092, Switzerland
| | - Young-Tae Chang
- Center for Self-assembly and Complexity, Institute for Basic Science (IBS), Pohang, Gyeongbuk 37673, Republic of Korea
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea
| | - Hongsoo Choi
- Department of Robotics and Mechatronics Engineering, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- DGIST-ETH Microrobotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- Robotics Research Center, Daegu Gyeong-buk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| |
Collapse
|
27
|
Gupta A, Efthymiou V, Kodani SD, Shamsi F, Patti ME, Tseng YH, Streets A. Mapping the transcriptional landscape of human white and brown adipogenesis using single-nuclei RNA-seq. Mol Metab 2023; 74:101746. [PMID: 37286033 PMCID: PMC10338377 DOI: 10.1016/j.molmet.2023.101746] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/09/2023] Open
Abstract
Adipogenesis is key to maintaining organism-wide energy balance and healthy metabolic phenotype, making it critical to thoroughly comprehend its molecular regulation in humans. By single-nuclei RNA-sequencing (snRNA-seq) of over 20,000 differentiating white and brown preadipocytes, we constructed a high-resolution temporal transcriptional landscape of human white and brown adipogenesis. White and brown preadipocytes were isolated from a single individual's neck region, thereby eliminating inter-subject variability across two distinct lineages. These preadipocytes were also immortalized to allow for controlled, in vitro differentiation, allowing sampling of distinct cellular states across the spectrum of adipogenic progression. Pseudotemporal cellular ordering revealed the dynamics of ECM remodeling during early adipogenesis, and lipogenic/thermogenic response during late white/brown adipogenesis. Comparison with adipogenic regulation in murine models Identified several novel transcription factors as potential targets for adipogenic/thermogenic drivers in humans. Among these novel candidates, we explored the role of TRPS1 in adipocyte differentiation and showed that its knockdown impairs white adipogenesis in vitro. Key adipogenic and lipogenic markers revealed in our analysis were applied to analyze publicly available scRNA-seq datasets; these confirmed unique cell maturation features in recently discovered murine preadipocytes, and revealed inhibition of adipogenic expansion in humans with obesity. Overall, our study presents a comprehensive molecular description of both white and brown adipogenesis in humans and provides an important resource for future studies of adipose tissue development and function in both health and metabolic disease state.
Collapse
Affiliation(s)
- Anushka Gupta
- University of California at Berkeley, University of California at San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA
| | - Vissarion Efthymiou
- Department of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Sean D Kodani
- Department of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Farnaz Shamsi
- Department of Molecular Pathobiology, New York University, New York, NY 10010, USA
| | - Mary Elizabeth Patti
- Department of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Yu-Hua Tseng
- Department of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Aaron Streets
- University of California at Berkeley, University of California at San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA; Biophysics Graduate Group, University of California at Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
28
|
Suzuki M, Liu C, Oyama K, Yamazawa T. Trans-scale thermal signaling in biological systems. J Biochem 2023; 174:217-225. [PMID: 37461189 PMCID: PMC10464929 DOI: 10.1093/jb/mvad053] [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: 04/30/2023] [Accepted: 06/21/2023] [Indexed: 08/31/2023] Open
Abstract
Biochemical reactions in cells serve as the endogenous source of heat, maintaining a constant body temperature. This process requires proper control; otherwise, serious consequences can arise due to the unwanted but unavoidable responses of biological systems to heat. This review aims to present a range of responses to heat in biological systems across various spatial scales. We begin by examining the impaired thermogenesis of malignant hyperthermia in model mice and skeletal muscle cells, demonstrating that the progression of this disease is caused by a positive feedback loop between thermally driven Ca2+ signaling and thermogenesis at the subcellular scale. After we explore thermally driven force generation in both muscle and non-muscle cells, we illustrate how in vitro assays using purified proteins can reveal the heat-responsive properties of proteins and protein assemblies. Building on these experimental findings, we propose the concept of 'trans-scale thermal signaling'.
Collapse
Key Words
- ATPase
- fluorescence microscopy
- heat-induced calcium release
- microheating
- type 1 ryanodine receptor.
Abbreviations: [Ca2+]i, intracellular Ca2+ concentration; CICR, Ca2+-induced Ca2+ release; ER, endoplasmic reticulum; FDB, flexor digitorum brevis; HEK293 cell, human embryonic kidney 293 cell; HICR, heat-induced Ca2+ release; IP3R, inositol 1,4,5-trisphosphate receptor; MH, malignant hyperthermia; RCC, rapid cooling contracture; RyR1, type 1 ryanodine receptor; SERCA, sarco/endoplasmic reticulum Ca2+-ATPase; SR, sarcoplasmic reticulum; TRP, transient receptor potential; WT, wild type
Collapse
Affiliation(s)
- Madoka Suzuki
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Chujie Liu
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1, Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
| | - Kotaro Oyama
- Foundational Quantum Technology Research Directorate, National Institutes for Quantum Science and Technology, 1233 Watanukimachi, Takasaki-shi, Gunma 370-1292, Japan
| | - Toshiko Yamazawa
- Core Research Facilities, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| |
Collapse
|
29
|
Onikanni SA, Yang CY, Noriega L, Wang CH. U0126 Compound Triggers Thermogenic Differentiation in Preadipocytes via ERK-AMPK Signaling Axis. Int J Mol Sci 2023; 24:ijms24097987. [PMID: 37175694 PMCID: PMC10178890 DOI: 10.3390/ijms24097987] [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: 03/07/2023] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
In recent years, thermogenic differentiation and activation in brown and white adipose tissues have been regarded as one of the major innovative and promising strategies for the treatment and amelioration of obesity. However, the pharmacological approach towards this process has had limited and insufficient commitments, which presents a greater challenge for obesity treatment. This research evaluates the effects of U0126 compound on the activation of thermogenic differentiation during adipogenesis. The results show that U0126 pretreatment primes both white and brown preadipocytes to upregulate thermogenic and mitochondrial genes as well as enhance functions during the differentiation process. We establish that U0126-mediated thermogenic differentiation induction occurs partially via AMPK activation signaling. The findings of this research suggest U0126 as a promising alternative ligand in pursuit of a pharmacological option to increase thermogenic adipocyte formation and improve energy expenditure. Thus it could pave the way for the discovery of therapeutic drugs for the treatment of obesity and its related complications.
Collapse
Affiliation(s)
- Sunday Amos Onikanni
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
- Department of Chemical Sciences, Biochemistry Unit, Afe Babalola University, Ado-Ekiti 360101, Ekiti State, Nigeria
| | - Cheng-Ying Yang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
| | - Lloyd Noriega
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
| | - Chih-Hao Wang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
- Graduate Institute of Cell Biology, College of Life Sciences, China Medical University, Taichung 406040, Taiwan
| |
Collapse
|
30
|
How hot can mitochondria be? Incubation at temperatures above 43 °C induces the degradation of respiratory complexes and supercomplexes in intact cells and isolated mitochondria. Mitochondrion 2023; 69:83-94. [PMID: 36764502 DOI: 10.1016/j.mito.2023.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/25/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023]
Abstract
Mitochondrial function generates an important fraction of the heat that contributes to cellular and organismal temperature maintenance, but the actual values of this parameter reached in the organelles is a matter of debate. The studies addressing this issue have reported divergent results: from detecting in the organelles the same temperature as the cell average or the incubation temperature, to increasing differences of up to 10 degrees above the incubation value. Theoretical calculations based on physical laws exclude the possibility of relevant temperature gradients between mitochondria and their surroundings. These facts have given rise to a conundrum or paradox about hot mitochondria. We have examined by Blue-Native electrophoresis, both in intact cells and in isolated organelles, the stability of respiratory complexes and supercomplexes at different temperatures to obtain information about their tolerance to heat stress. We observe that, upon incubation at values above 43 °C and after relatively short periods, respiratory complexes, and especially complex I and its supercomplexes, are unstable even when the respiratory activity is inhibited. These results support the conclusion that high temperatures (>43 °C) cause damage to mitochondrial structure and function and question the proposal that these organelles can physiologically work at close to 50 °C.
Collapse
|
31
|
Inada N. A Guide to Plant Intracellular Temperature Imaging using Fluorescent Thermometers. PLANT & CELL PHYSIOLOGY 2023; 64:7-18. [PMID: 36039974 DOI: 10.1093/pcp/pcac123] [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: 02/20/2022] [Revised: 07/06/2022] [Accepted: 08/30/2022] [Indexed: 06/15/2023]
Abstract
All aspects of plant physiology are influenced by temperature. Changes in environmental temperature alter the temperatures of plant tissues and cells, which then affect various cellular activities, such as gene expression, protein stability and enzyme activities. In turn, changes in cellular activities, which are associated with either exothermic or endothermic reactions, can change the local temperature in cells and tissues. In the past 10 years, a number of fluorescent probes that detect temperature and enable intracellular temperature imaging have been reported. Intracellular temperature imaging has revealed that there is a temperature difference >1°C inside cells and that the treatment of cells with mitochondrial uncoupler or ionomycin can cause more than a 1°C intracellular temperature increase in mammalian cultured cells. Thermogenesis mechanisms in brown adipocytes have been revealed with the aid of intracellular temperature imaging. While there have been no reports on plant intracellular temperature imaging thus far, intracellular temperature imaging is expected to provide a new way to analyze the mechanisms underlying the various activities of plant cells. In this review, I will first summarize the recent progress in the development of fluorescent thermometers and their biological applications. I will then discuss the selection of fluorescent thermometers and experimental setup for the adaptation of intracellular temperature imaging to plant cells. Finally, possible applications of intracellular temperature imaging to investigate plant cell functions will be discussed.
Collapse
Affiliation(s)
- Noriko Inada
- Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai-shi, Osaka, 599-8531 Japan
- School of Agriculture, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai-shi, Osaka, 599-8531 Japan
| |
Collapse
|
32
|
Michurina S, Stafeev I, Boldyreva M, Truong VA, Ratner E, Menshikov M, Hu YC, Parfyonova Y. Transplantation of Adipose-Tissue-Engineered Constructs with CRISPR-Mediated UCP1 Activation. Int J Mol Sci 2023; 24:ijms24043844. [PMID: 36835254 PMCID: PMC9959691 DOI: 10.3390/ijms24043844] [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: 11/02/2022] [Revised: 02/05/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
Thermogenic adipocytes have potential utility for the development of approaches to treat type 2 diabetes and obesity-associated diseases. Although several reports have proved the positive effect of beige and brown adipocyte transplantation in obese mice, translation to human cell therapy needs improvement. Here, we describe the application of CRISPR activation (CRISPRa) technology for generating safe and efficient adipose-tissue-engineered constructs with enhanced mitochondrial uncoupling protein 1 (UCP1) expression. We designed the CRISPRa system for the activation of UCP1 gene expression. CRISPRa-UCP1 was delivered into mature adipocytes by a baculovirus vector. Modified adipocytes were transplanted in C57BL/6 mice, followed by analysis of grafts, inflammation and systemic glucose metabolism. Staining of grafts on day 8 after transplantation shows them to contain UCP1-positive adipocytes. Following transplantation, adipocytes remain in grafts and exhibit expression of PGC1α transcription factor and hormone sensitive lipase (HSL). Transplantation of CRISPRa-UCP1-modified adipocytes does not influence glucose metabolism or inflammation in recipient mice. We show the utility and safety of baculovirus vectors for CRISPRa-based thermogenic gene activation. Our findings suggest a means of improving existing cell therapy approaches using baculovirus vectors and CRISPRa for modification and transplantation of non-immunogenic adipocytes.
Collapse
Affiliation(s)
- Svetlana Michurina
- National Medical Research Centre of Cardiology Named after Academician E. I. Chazov, 121552 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- Correspondence: (S.M.); (I.S.)
| | - Iurii Stafeev
- National Medical Research Centre of Cardiology Named after Academician E. I. Chazov, 121552 Moscow, Russia
- Correspondence: (S.M.); (I.S.)
| | - Maria Boldyreva
- National Medical Research Centre of Cardiology Named after Academician E. I. Chazov, 121552 Moscow, Russia
- Cell and Molecular Biology Unit, Faculty of Biology and Biotechnology, National Research University Higher School of Economics, 101000 Moscow, Russia
| | - Vu Anh Truong
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Elizaveta Ratner
- National Medical Research Centre of Cardiology Named after Academician E. I. Chazov, 121552 Moscow, Russia
| | - Mikhail Menshikov
- National Medical Research Centre of Cardiology Named after Academician E. I. Chazov, 121552 Moscow, Russia
| | - Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300044, Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Yelena Parfyonova
- National Medical Research Centre of Cardiology Named after Academician E. I. Chazov, 121552 Moscow, Russia
- Faculty of Basic Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| |
Collapse
|
33
|
Veysset D, Zhuo Y, Hattori J, Buckhory M, Palanker D. Interferometric thermometry of ocular tissues for retinal laser therapy. BIOMEDICAL OPTICS EXPRESS 2023; 14:37-53. [PMID: 36698667 PMCID: PMC9842005 DOI: 10.1364/boe.475705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/13/2022] [Accepted: 11/14/2022] [Indexed: 06/17/2023]
Abstract
Controlling the tissue temperature rise during retinal laser therapy is highly desirable for predictable and reproducible outcomes of the procedure, especially with non-damaging settings. In this work, we demonstrate a method for determining the optical absorption, the thermal conductivity, and the thermal expansion coefficients of RPE and choroid using phase-resolved optical coherence tomography (pOCT). These parameters are extracted from the measured changes in the optical path length (ΔOPL) using an axisymmetric thermo-mechanical model. This allows the calculation of the temperature rise during hyperthermia, which was further validated by imaging the temperature-sensitive fluorescence at the same location. We demonstrate that, with a temperature uncertainty of ±0.9°C and a peak heating of about 17°C following a laser pulse of 20 ms, this methodology is expected to be safe and sufficiently precise for calibration of the non-damaging retinal laser therapy. The method is directly translatable to in-vivo studies, where we expect a similar precision.
Collapse
Affiliation(s)
- David Veysset
- Hansen Experimental Physics Laboratory, Stanford University, Stanford, CA 94305, USA
- Department of Ophthalmology, Stanford University, Stanford, CA 94305, USA
| | - Yueming Zhuo
- Hansen Experimental Physics Laboratory, Stanford University, Stanford, CA 94305, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Junya Hattori
- Hansen Experimental Physics Laboratory, Stanford University, Stanford, CA 94305, USA
- Department of Mechanical Engineering, The University of Tokyo, Tokyo, Japan
| | - Mohajeet Buckhory
- Hansen Experimental Physics Laboratory, Stanford University, Stanford, CA 94305, USA
- Department of Ophthalmology, Stanford University, Stanford, CA 94305, USA
| | - Daniel Palanker
- Hansen Experimental Physics Laboratory, Stanford University, Stanford, CA 94305, USA
- Department of Ophthalmology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
34
|
Tsuji T, Kajimoto K, Inada N. Measurement of Intracellular Temperature in Brown Adipocytes Using a Cationic Fluorescent Polymeric Thermometer. Methods Mol Biol 2023; 2662:87-102. [PMID: 37076673 DOI: 10.1007/978-1-0716-3167-6_8] [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: 04/21/2023]
Abstract
Brown adipose tissue specializes in expending energy through non-shivering thermogenesis, and many studies have associated its activity with protection and treatment of obesity and metabolic diseases. To reveal the mechanisms involved in heat production, primary cultured brown adipose cells (BACs) have been used because of their ease of genetic engineering and similarity to living tissue. However, thermogenic activity has often been evaluated as an indirect method, such as the measurement of oxygen consumption. Recently, fluorescent nanothermometers for the direct measurement of intracellular temperature have been developed and applied to elucidate the mechanisms of heat production in BACs. In this chapter, we introduce a protocol that uses a cationic fluorescent polymeric thermometer to directly measure the temperature within primary cultured BACs. We anticipate that this protocol will be beneficial in elucidating the mechanism of thermogenesis in BACs.
Collapse
Affiliation(s)
- Toshikazu Tsuji
- Kirin Central Research Institute, Kirin Holdings Company Limited, Fujisawa, Kanagawa, Japan.
| | - Kazuaki Kajimoto
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa, Japan
| | - Noriko Inada
- School of Agriculture, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
35
|
Liu X, Yamazaki T, Kwon HY, Arai S, Chang YT. A palette of site-specific organelle fluorescent thermometers. Mater Today Bio 2022; 16:100405. [PMID: 36060107 PMCID: PMC9434161 DOI: 10.1016/j.mtbio.2022.100405] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 11/04/2022] Open
Abstract
Intracellular micro-temperature is closely related to cellular processes. Such local temperature inside cells can be measured by fluorescent thermometers, which are a series of fluorescent materials that convert the temperature information to detectable fluorescence signals. To investigate the intracellular temperature fluctuation in various organelles, it is essential to develop site-specific organelle thermometers. In this study, we develop a new series of fluorescent thermometers, Thermo Greens (TGs), to visualize the temperature change in almost all typical organelles. Through fluorescence lifetime-based cell imaging, it was proven that TGs allow the organelle-specific monitoring of temperature gradients created by external heating. The fluorescence lifetime-based thermometry shows that each organelle experiences a distinct temperature increment which depends on the distance away from the heat source. TGs are further demonstrated in the quantitative imaging of heat production at different organelles such as mitochondria and endoplasmic reticulum in brown adipocytes. To date, TGs are the first palette batch of small molecular fluorescent thermometers that can cover almost all typical organelles. These findings can inspire the development of new fluorescent thermometers and enhance the understanding of thermal biology in the future.
Collapse
Affiliation(s)
- Xiao Liu
- Center for Self-assembly and Complexity, Institute for Basic Science (IBS), Pohang, Gyeongbuk, 37673, South Korea
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, South Korea
| | - Takeru Yamazaki
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Haw-Young Kwon
- Center for Self-assembly and Complexity, Institute for Basic Science (IBS), Pohang, Gyeongbuk, 37673, South Korea
| | - Satoshi Arai
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Young-Tae Chang
- Center for Self-assembly and Complexity, Institute for Basic Science (IBS), Pohang, Gyeongbuk, 37673, South Korea
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, South Korea
| |
Collapse
|
36
|
Pittman M, Ali AM, Chen Y. How sticky? How tight? How hot? Imaging probes for fluid viscosity, membrane tension and temperature measurements at the cellular level. Int J Biochem Cell Biol 2022; 153:106329. [PMID: 36336304 PMCID: PMC10148659 DOI: 10.1016/j.biocel.2022.106329] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/22/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
We review the progress made in imaging probes for three important physical parameters: viscosity, membrane tension, and temperature, all of which play important roles in many cellular processes. Recent evidences showed that cell migration speed can be modulated by extracellular fluid viscosity; membrane tension contributes to the regulation of cell motility, exo-/endo-cytosis, and cell spread area; and temperature affects neural activity and adipocyte differentiation. We discuss the techniques implementing imaging-based probes to measure viscosity, membrane tension, and temperature at subcellular resolution dynamically. The merits and shortcomings of each technique are examined, and the future applications of the recently developed techniques are also explored.
Collapse
Affiliation(s)
- Matthew Pittman
- Department of Mechanical Engineering, Johns Hopkins University, MD, USA; Center for Cell Dynamics, Johns Hopkins University, MD, USA; Institute for NanoBio Technology, Johns Hopkins University, MD, USA
| | - Abdulla M Ali
- Center for Cell Dynamics, Johns Hopkins University, MD, USA; Institute for NanoBio Technology, Johns Hopkins University, MD, USA; T.C. Jenkins Department of Biophysics, Johns Hopkins University, MD, USA
| | - Yun Chen
- Department of Mechanical Engineering, Johns Hopkins University, MD, USA; Center for Cell Dynamics, Johns Hopkins University, MD, USA; Institute for NanoBio Technology, Johns Hopkins University, MD, USA.
| |
Collapse
|
37
|
Tsuboi Y, Oyama K, Kobirumaki-Shimozawa F, Murayama T, Kurebayashi N, Tachibana T, Manome Y, Kikuchi E, Noguchi S, Inoue T, Inoue YU, Nishino I, Mori S, Ishida R, Kagechika H, Suzuki M, Fukuda N, Yamazawa T. Mice with R2509C-RYR1 mutation exhibit dysfunctional Ca2+ dynamics in primary skeletal myocytes. J Gen Physiol 2022; 154:213526. [PMID: 36200983 PMCID: PMC9546722 DOI: 10.1085/jgp.202213136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/22/2022] [Accepted: 09/14/2022] [Indexed: 11/20/2022] Open
Abstract
Type 1 ryanodine receptor (RYR1) is a Ca2+ release channel in the sarcoplasmic reticulum (SR) of the skeletal muscle and plays a critical role in excitation-contraction coupling. Mutations in RYR1 cause severe muscle diseases, such as malignant hyperthermia, a disorder of Ca2+-induced Ca2+ release (CICR) through RYR1 from the SR. We recently reported that volatile anesthetics induce malignant hyperthermia (MH)-like episodes through enhanced CICR in heterozygous R2509C-RYR1 mice. However, the characterization of Ca2+ dynamics has yet to be investigated in skeletal muscle cells from homozygous mice because these animals die in utero. In the present study, we generated primary cultured skeletal myocytes from R2509C-RYR1 mice. No differences in cellular morphology were detected between wild type (WT) and mutant myocytes. Spontaneous Ca2+ transients and cellular contractions occurred in WT and heterozygous myocytes, but not in homozygous myocytes. Electron microscopic observation revealed that the sarcomere length was shortened to ∼1.7 µm in homozygous myocytes, as compared to ∼2.2 and ∼2.3 µm in WT and heterozygous myocytes, respectively. Consistently, the resting intracellular Ca2+ concentration was higher in homozygous myocytes than in WT or heterozygous myocytes, which may be coupled with a reduced Ca2+ concentration in the SR. Finally, using infrared laser-based microheating, we found that heterozygous myocytes showed larger heat-induced Ca2+ transients than WT myocytes. Our findings suggest that the R2509C mutation in RYR1 causes dysfunctional Ca2+ dynamics in a mutant-gene dose-dependent manner in the skeletal muscles, in turn provoking MH-like episodes and embryonic lethality in heterozygous and homozygous mice, respectively.
Collapse
Affiliation(s)
- Yoshitaka Tsuboi
- Core Research Facilities, The Jikei University School of Medicine, Tokyo, Japan.,Department of Molecular Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kotaro Oyama
- Quantum Beam Science Research Directorate, National Institutes for Quantum Science and Technology, Gunma, Japan.,Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | | | - Takashi Murayama
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nagomi Kurebayashi
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toshiaki Tachibana
- Core Research Facilities, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshinobu Manome
- Core Research Facilities, The Jikei University School of Medicine, Tokyo, Japan
| | - Emi Kikuchi
- Core Research Facilities, The Jikei University School of Medicine, Tokyo, Japan
| | - Satoru Noguchi
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takayoshi Inoue
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yukiko U Inoue
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Shuichi Mori
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryosuke Ishida
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Kagechika
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Madoka Suzuki
- Institute for Protein Research, Osaka University, Osaka, Japan
| | - Norio Fukuda
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Toshiko Yamazawa
- Core Research Facilities, The Jikei University School of Medicine, Tokyo, Japan.,Department of Molecular Physiology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
38
|
Qi Y, Hui XH. The Single-Cell Revelation of Thermogenic Adipose Tissue. Mol Cells 2022; 45:673-684. [PMID: 36254709 PMCID: PMC9589375 DOI: 10.14348/molcells.2022.0092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 11/07/2022] Open
Abstract
The past two decades have witnessed an upsurge in the appreciation of adipose tissue (AT) as an immuno-metabolic hub harbouring heterogeneous cell populations that collectively fine-tune systemic metabolic homeostasis. Technological advancements, especially single-cell transcriptomics, have offered an unprecedented opportunity for dissecting the sophisticated cellular networks and compositional dynamics underpinning AT remodelling. The "re-discovery" of functional brown adipose tissue dissipating heat energy in human adults has aroused tremendous interest in exploiting the mechanisms underpinning the engagement of AT thermogenesis for combating human obesity. In this review, we aim to summarise and evaluate the use of single-cell transcriptomics that contribute to a better appreciation of the cellular plasticity and intercellular crosstalk in thermogenic AT.
Collapse
Affiliation(s)
- Yue Qi
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoyan Hannah Hui
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
39
|
Liao FH, Yao CN, Chen SP, Wu TH, Lin SY. Transdermal Delivery of Succinate Accelerates Energy Dissipation of Brown Adipocytes to Reduce Remote Fat Accumulation. Mol Pharm 2022; 19:4299-4310. [DOI: 10.1021/acs.molpharmaceut.2c00628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Fang-Hsuean Liao
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35 Keyan Road,
Zhunan Town, Miaoli County 35053, Taiwan
| | - Chun-Nien Yao
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35 Keyan Road,
Zhunan Town, Miaoli County 35053, Taiwan
| | - Shu-Ping Chen
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35 Keyan Road,
Zhunan Town, Miaoli County 35053, Taiwan
| | - Te-Haw Wu
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35 Keyan Road,
Zhunan Town, Miaoli County 35053, Taiwan
| | - Shu-Yi Lin
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35 Keyan Road,
Zhunan Town, Miaoli County 35053, Taiwan
| |
Collapse
|
40
|
Heat-hypersensitive mutants of ryanodine receptor type 1 revealed by microscopic heating. Proc Natl Acad Sci U S A 2022; 119:e2201286119. [PMID: 35925888 PMCID: PMC9371657 DOI: 10.1073/pnas.2201286119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Malignant hyperthermia (MH) is a life-threatening disorder caused largely by mutations in ryanodine receptor type 1 (RyR1) Ca2+-release channels. Enhanced Ca2+ release through the mutant channels induces excessive heat development upon exposure to volatile anesthetics. However, the mechanism by which Ca2+ release is accelerated at an elevated temperature is yet to be identified. Fluorescence Ca2+ imaging with rapid heating by an infrared laser beam provides direct evidence that heat induces Ca2+ release through the RyR1 channel. And the mutant channels are more heat sensitive than the wild-type channels, thereby causing an increase in the cytosolic Ca2+ concentration in mutant cells. It is likely that the heat-induced Ca2+ release participates as an enhancer in the cellular mechanism of MH. Thermoregulation is an important aspect of human homeostasis, and high temperatures pose serious stresses for the body. Malignant hyperthermia (MH) is a life-threatening disorder in which body temperature can rise to a lethal level. Here we employ an optically controlled local heat-pulse method to manipulate the temperature in cells with a precision of less than 1 °C and find that the mutants of ryanodine receptor type 1 (RyR1), a key Ca2+ release channel underlying MH, are heat hypersensitive compared with the wild type (WT). We show that the local heat pulses induce an intracellular Ca2+ burst in human embryonic kidney 293 cells overexpressing WT RyR1 and some RyR1 mutants related to MH. Fluorescence Ca2+ imaging using the endoplasmic reticulum–targeted fluorescent probes demonstrates that the Ca2+ burst originates from heat-induced Ca2+ release (HICR) through RyR1-mutant channels because of the channels’ heat hypersensitivity. Furthermore, the variation in the heat hypersensitivity of four RyR1 mutants highlights the complexity of MH. HICR likewise occurs in skeletal muscles of MH model mice. We propose that HICR contributes an additional positive feedback to accelerate thermogenesis in patients with MH.
Collapse
|
41
|
Substantial impact of 3-iodothyronamine (T1AM) on the regulations of fluorescent thermoprobe-measured cellular temperature and natriuretic peptide expression in cardiomyocytes. Sci Rep 2022; 12:12740. [PMID: 35882940 PMCID: PMC9325765 DOI: 10.1038/s41598-022-17086-2] [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: 03/12/2022] [Accepted: 07/20/2022] [Indexed: 11/11/2022] Open
Abstract
There is growing interest in 3-iodothyronamine (T1AM), an active thyroid hormone metabolite, that induces negative inotropic and chronotropic actions in the heart and exerts systemic hypothermic action. We explored the direct impact of T1AM on cardiomyocytes with a focus on the regulation of the intracellular temperature and natriuretic peptide (NP) expression. A thermoprobe was successfully introduced into neonatal rat cardiomyocytes, and the temperature-dependent changes in the fluorescence intensity ratio were measured using a fluorescence microscope. After one-hour incubation with T1AM, the degree of change in the fluorescence intensity ratio was significantly lower in T1AM-treated cardiomyocytes than in equivalent solvent-treated controls (P < 0.01), indicating the direct hypothermic action of T1AM on cardiomyocytes. Furthermore, T1AM treatment upregulated B-type NP (BNP) gene expression comparable to treatment with endothelin-1 or phenylephrine. Of note, ERK phosphorylation was markedly increased after T1AM treatment, and inhibition of ERK phosphorylation by an MEK inhibitor completely cancelled both T1AM-induced decrease in thermoprobe-measured temperature and the increase in BNP expression. In summary, T1AM decreases fluorescent thermoprobe-measured temperatures (estimated intracellular temperatures) and increases BNP expression in cardiomyocytes by activating the MEK/ERK pathway. The present findings provide new insight into the direct myocardial cellular actions of T1AM in patients with severe heart failure.
Collapse
|
42
|
Li BY, Guo YY, Xiao G, Guo L, Tang QQ. SERPINA3C ameliorates adipose tissue inflammation through the Cathepsin G/Integrin/AKT pathway. Mol Metab 2022; 61:101500. [PMID: 35436587 PMCID: PMC9062745 DOI: 10.1016/j.molmet.2022.101500] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/10/2022] [Accepted: 04/13/2022] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE Due to the increasing prevalence of obesity and insulin resistance, there is an urgent need for better treatment of obesity and its related metabolic disorders. This study aimed to elucidate the role of SERPINA3C, an adipocyte secreted protein, in obesity and related metabolic disorders. METHODS Male wild type (WT) and knockout (KO) mice were fed with high-fat diet (HFD) for 16 weeks, adiposity, insulin resistance, and inflammation were assessed. AAV-mediated overexpression of SERPINA3C was injected locally in inguinal white adipose tissue (iWAT) to examine the effect of SERPINA3C. In vitro analyses were conducted in 3T3-L1 adipocytes to explore the molecular pathways underlying the function of SERPINA3C. RESULTS Functional exploration of the SERPINA3C knockout mice revealed that SERPINA3C deficiency led to an impaired metabolic phenotype (more severe obesity, lower metabolic rates, worse glucose intolerance and insulin insensitivity), which was associated with anabatic inflammation and apoptosis of white adipose tissues. Consistent with these results, overexpression of SERPINA3C in inguinal adipose tissue protected mice against diet-induced obesity and metabolic disorders with less inflammation and apoptosis in adipose tissue. Mechanistically, SERPINA3C inhibited Cathepsin G activity, acting as a serine protease inhibitor, which blocked Cathepsin G-mediated turnover of α5/β1 Integrin protein. Then, the preserved integrity (increase) of α5/β1 Integrin signaling activated AKT to decrease JNK phosphorylation, thereby inhibiting inflammation and promoting insulin sensitivity in adipocytes. CONCLUSIONS/INTERPRETATION These findings demonstrate a previously unknown SERPINA3C/Cathepsin G/Integrin/AKT pathway in regulating adipose tissue inflammation, and suggest the therapeutic potential of targeting SERPINA3C/Cathepsin G axis in adipose tissue for the treatment of obesity and metabolic diseases.
Collapse
Affiliation(s)
- Bai-Yu Li
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ying-Ying Guo
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Gang Xiao
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Liang Guo
- School of Kinesiology, and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, 200438, China.
| | - Qi-Qun Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
43
|
Sugimoto S, Mena HA, Sansbury BE, Kobayashi S, Tsuji T, Wang CH, Yin X, Huang TL, Kusuyama J, Kodani SD, Darcy J, Profeta G, Pereira N, Tanzi RE, Zhang C, Serwold T, Kokkotou E, Goodyear LJ, Cypess AM, Leiria LO, Spite M, Tseng YH. Brown adipose tissue-derived MaR2 contributes to cold-induced resolution of inflammation. Nat Metab 2022; 4:775-790. [PMID: 35760872 PMCID: PMC9792164 DOI: 10.1038/s42255-022-00590-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 05/18/2022] [Indexed: 12/30/2022]
Abstract
Obesity induces chronic inflammation resulting in insulin resistance and metabolic disorders. Cold exposure can improve insulin sensitivity in humans and rodents, but the mechanisms have not been fully elucidated. Here, we find that cold resolves obesity-induced inflammation and insulin resistance and improves glucose tolerance in diet-induced obese mice. The beneficial effects of cold exposure on improving obesity-induced inflammation and insulin resistance depend on brown adipose tissue (BAT) and liver. Using targeted liquid chromatography with tandem mass spectrometry, we discovered that cold and β3-adrenergic stimulation promote BAT to produce maresin 2 (MaR2), a member of the specialized pro-resolving mediators of bioactive lipids that play a role in the resolution of inflammation. Notably, MaR2 reduces inflammation in obesity in part by targeting macrophages in the liver. Thus, BAT-derived MaR2 could contribute to the beneficial effects of BAT activation in resolving obesity-induced inflammation and may inform therapeutic approaches to combat obesity and its complications.
Collapse
Affiliation(s)
- Satoru Sugimoto
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Hebe Agustina Mena
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Brian E Sansbury
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Shio Kobayashi
- Section of Immunobiology, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Tadataka Tsuji
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Chih-Hao Wang
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Xuanzhi Yin
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tian Lian Huang
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Joji Kusuyama
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Sean D Kodani
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Justin Darcy
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Gerson Profeta
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Nayara Pereira
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Can Zhang
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Thomas Serwold
- Section of Immunobiology, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Efi Kokkotou
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Luiz Osório Leiria
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Matthew Spite
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
44
|
Kassotis CD, Vom Saal FS, Babin PJ, Lagadic-Gossmann D, Le Mentec H, Blumberg B, Mohajer N, Legrand A, Munic Kos V, Martin-Chouly C, Podechard N, Langouët S, Touma C, Barouki R, Kim MJ, Audouze K, Choudhury M, Shree N, Bansal A, Howard S, Heindel JJ. Obesity III: Obesogen assays: Limitations, strengths, and new directions. Biochem Pharmacol 2022; 199:115014. [PMID: 35393121 PMCID: PMC9050906 DOI: 10.1016/j.bcp.2022.115014] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/11/2022]
Abstract
There is increasing evidence of a role for environmental contaminants in disrupting metabolic health in both humans and animals. Despite a growing need for well-understood models for evaluating adipogenic and potential obesogenic contaminants, there has been a reliance on decades-old in vitro models that have not been appropriately managed by cell line providers. There has been a quick rise in available in vitro models in the last ten years, including commercial availability of human mesenchymal stem cell and preadipocyte models; these models require more comprehensive validation but demonstrate real promise in improved translation to human metabolic health. There is also progress in developing three-dimensional and co-culture techniques that allow for the interrogation of a more physiologically relevant state. While diverse rodent models exist for evaluating putative obesogenic and/or adipogenic chemicals in a physiologically relevant context, increasing capabilities have been identified for alternative model organisms such as Drosophila, C. elegans, zebrafish, and medaka in metabolic health testing. These models have several appreciable advantages, including most notably their size, rapid development, large brood sizes, and ease of high-resolution lipid accumulation imaging throughout the organisms. They are anticipated to expand the capabilities of metabolic health research, particularly when coupled with emerging obesogen evaluation techniques as described herein.
Collapse
Affiliation(s)
- Christopher D Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, United States.
| | - Frederick S Vom Saal
- Division of Biological Sciences, The University of Missouri, Columbia, MO 65211, United States
| | - Patrick J Babin
- Department of Life and Health Sciences, University of Bordeaux, INSERM, Pessac, France
| | - Dominique Lagadic-Gossmann
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Helene Le Mentec
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, The University of California, Irvine, Irvine CA 92697, United States
| | - Nicole Mohajer
- Department of Developmental and Cell Biology, The University of California, Irvine, Irvine CA 92697, United States
| | - Antoine Legrand
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Vesna Munic Kos
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Corinne Martin-Chouly
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Normand Podechard
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Sophie Langouët
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Charbel Touma
- Univ Rennes, Inserm, EHESP, Irset (Research Institute for Environmental and Occupational Health) - UMR_S 1085, 35 000 Rennes, France
| | - Robert Barouki
- Department of Biochemistry, University of Paris, INSERM, Paris, France
| | - Min Ji Kim
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | | | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Texas A & M University, College Station, TX 77843, United States
| | - Nitya Shree
- Department of Pharmaceutical Sciences, Texas A & M University, College Station, TX 77843, United States
| | - Amita Bansal
- College of Health & Medicine, Australian National University, Canberra, ACT, 2611, Australia
| | - Sarah Howard
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States
| | - Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States
| |
Collapse
|
45
|
Rakoczy RJ, Schiebrel CM, Wyatt CN. Acute Oxygen-Sensing via Mitochondria-Generated Temperature Transients in Rat Carotid Body Type I Cells. Front Physiol 2022; 13:874039. [PMID: 35510145 PMCID: PMC9060449 DOI: 10.3389/fphys.2022.874039] [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: 02/11/2022] [Accepted: 03/28/2022] [Indexed: 12/28/2022] Open
Abstract
The Carotid Bodies (CB) are peripheral chemoreceptors that detect changes in arterial oxygenation and, via afferent inputs to the brainstem, correct the pattern of breathing to restore blood gas homeostasis. Herein, preliminary evidence is presented supporting a novel oxygen-sensing hypothesis which suggests CB Type I cell “hypoxic signaling” may in part be mediated by mitochondria-generated thermal transients in TASK-channel-containing microdomains. Distances were measured between antibody-labeled mitochondria and TASK-potassium channels in primary rat CB Type I cells. Sub-micron distance measurements (TASK-1: 0.33 ± 0.04 µm, n = 47 vs TASK-3: 0.32 ± 0.03 µm, n = 54) provided evidence for CB Type I cell oxygen-sensing microdomains. A temperature-sensitive dye (ERthermAC) indicated that inhibition of mitochondrial activity in isolated cells caused a rapid and reversible inhibition of mitochondrial thermogenesis and thus temperature in these microdomains. Whole-cell perforated-patch current-clamp electrophysiological recordings demonstrated sensitivity of resting membrane potential (Vm) to temperature: lowering bath temperature from 37°C to 24°C induced consistent and reversible depolarizations (Vm at 37°C: 48.4 ± 4.11 mV vs 24°C: 31.0 ± 5.69 mV; n = 5; p < 0.01). These data suggest that hypoxic inhibition of mitochondrial thermogenesis may play an important role in oxygen chemotransduction in the CB. A reduction in temperature within cellular microdomains will inhibit plasma membrane ion channels, influence the balance of cellular phosphorylation–dephosphorylation, and may extend the half-life of reactive oxygen species. The characterization of a thermosensory chemotransduction mechanism, that may also be used by other oxygen-sensitive cell types and may impact multiple other chemotransduction mechanisms is critical if we are to fully understand how the CBs, and potentially other oxygen-sensitive cells, respond to hypoxia.
Collapse
|
46
|
Li Y, Wang D, Ping X, Zhang Y, Zhang T, Wang L, Jin L, Zhao W, Guo M, Shen F, Meng M, Chen X, Zheng Y, Wang J, Li D, Zhang Q, Hu C, Xu L, Ma X. Local hyperthermia therapy induces browning of white fat and treats obesity. Cell 2022; 185:949-966.e19. [PMID: 35247329 DOI: 10.1016/j.cell.2022.02.004] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/28/2021] [Accepted: 02/02/2022] [Indexed: 02/08/2023]
Abstract
Beige fat plays key roles in the regulation of systemic energy homeostasis; however, detailed mechanisms and safe strategy for its activation remain elusive. In this study, we discovered that local hyperthermia therapy (LHT) targeting beige fat promoted its activation in humans and mice. LHT achieved using a hydrogel-based photothermal therapy activated beige fat, preventing and treating obesity in mice without adverse effects. HSF1 is required for the effects since HSF1 deficiency blunted the metabolic benefits of LHT. HSF1 regulates Hnrnpa2b1 (A2b1) transcription, leading to increased mRNA stability of key metabolic genes. Importantly, analysis of human association studies followed by functional analysis revealed that the HSF1 gain-of-function variant p.P365T is associated with improved metabolic performance in humans and increased A2b1 transcription in mice and cells. Overall, we demonstrate that LHT offers a promising strategy against obesity by inducing beige fat activation via HSF1-A2B1 transcriptional axis.
Collapse
Affiliation(s)
- Yu Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Dongmei Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiaodan Ping
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yankang Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Ting Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Li Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Li Jin
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Wenjun Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Mingwei Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Fei Shen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Meiyao Meng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xin Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Ying Zheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jiqiu Wang
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China; Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Qiang Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China; Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 201499, China.
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China; Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China; Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 201499, China; Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
47
|
Murakami A, Nagao K, Sakaguchi R, Kida K, Hara Y, Mori Y, Okabe K, Harada Y, Umeda M. Cell-autonomous control of intracellular temperature by unsaturation of phospholipid acyl chains. Cell Rep 2022; 38:110487. [PMID: 35294880 DOI: 10.1016/j.celrep.2022.110487] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 12/03/2021] [Accepted: 02/14/2022] [Indexed: 11/25/2022] Open
Abstract
Intracellular temperature affects a wide range of cellular functions in living organisms. However, it remains unclear whether temperature in individual animal cells is controlled autonomously as a response to fluctuations in environmental temperature. Using two distinct intracellular thermometers, we find that the intracellular temperature of steady-state Drosophila S2 cells is maintained in a manner dependent on Δ9-fatty acid desaturase DESAT1, which introduces a double bond at the Δ9 position of the acyl moiety of acyl-CoA. The DESAT1-mediated increase of intracellular temperature is caused by the enhancement of F1Fo-ATPase-dependent mitochondrial respiration, which is coupled with thermogenesis. We also reveal that F1Fo-ATPase-dependent mitochondrial respiration is potentiated by cold exposure through the remodeling of mitochondrial cristae structures via DESAT1-dependent unsaturation of mitochondrial phospholipid acyl chains. Based on these findings, we propose a cell-autonomous mechanism for intracellular temperature control during environmental temperature changes.
Collapse
Affiliation(s)
- Akira Murakami
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan; Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Kohjiro Nagao
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan.
| | - Reiko Sakaguchi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Keisuke Kida
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Yuji Hara
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Kohki Okabe
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yoshie Harada
- Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Masato Umeda
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan.
| |
Collapse
|
48
|
Li Y, Fromme T. Uncoupling Protein 1 Does Not Produce Heat without Activation. Int J Mol Sci 2022; 23:2406. [PMID: 35269549 PMCID: PMC8910648 DOI: 10.3390/ijms23052406] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial uncoupling protein 1 (UCP1) is the crucial mechanistic component of heat production in classical brown fat and the newly identified beige or brite fat. Thermogenesis inevitably comes at a high energetic cost and brown fat, ultimately, is an energy-wasting organ. A constrained strategy that minimizes brown fat activity unless obligate will have been favored during natural selection to safeguard metabolic thriftiness. Accordingly, UCP1 is constitutively inhibited and is inherently not leaky without activation. It follows that increasing brown adipocyte number or UCP1 abundance genetically or pharmacologically does not lead to an automatic increase in thermogenesis or subsequent metabolic consequences in the absence of a plausible route of concomitant activation. Despite its apparent obviousness, this tenet is frequently ignored. Consequently, incorrect conclusions are often drawn from increased BAT or brite/beige depot mass, e.g., predicting or causally linking beneficial metabolic effects. Here, we highlight the inherently inactive nature of UCP1, with a particular emphasis on the molecular brakes and releases of UCP1 activation under physiological conditions. These controls of UCP1 activity represent potential targets of therapeutic interventions to unlock constraints and efficiently harness the energy-expending potential of brown fat to prevent and treat obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Yongguo Li
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Tobias Fromme
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| |
Collapse
|
49
|
Gupta A, Shamsi F, Altemose N, Dorlhiac GF, Cypess AM, White AP, Yosef N, Patti ME, Tseng YH, Streets A. Characterization of transcript enrichment and detection bias in single-nucleus RNA-seq for mapping of distinct human adipocyte lineages. Genome Res 2022; 32:242-257. [PMID: 35042723 PMCID: PMC8805720 DOI: 10.1101/gr.275509.121] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 12/10/2021] [Indexed: 02/02/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) enables molecular characterization of complex biological tissues at high resolution. The requirement of single-cell extraction, however, makes it challenging for profiling tissues such as adipose tissue, for which collection of intact single adipocytes is complicated by their fragile nature. For such tissues, single-nucleus extraction is often much more efficient and therefore single-nucleus RNA sequencing (snRNA-seq) presents an alternative to scRNA-seq. However, nuclear transcripts represent only a fraction of the transcriptome in a single cell, with snRNA-seq marked with inherent transcript enrichment and detection biases. Therefore, snRNA-seq may be inadequate for mapping important transcriptional signatures in adipose tissue. In this study, we compare the transcriptomic landscape of single nuclei isolated from preadipocytes and mature adipocytes across human white and brown adipocyte lineages, with whole-cell transcriptome. We show that snRNA-seq is capable of identifying the broad cell types present in scRNA-seq at all states of adipogenesis. However, we also explore how and why the nuclear transcriptome is biased and limited, as well as how it can be advantageous. We robustly characterize the enrichment of nuclear-localized transcripts and adipogenic regulatory lncRNAs in snRNA-seq, while also providing a detailed understanding for the preferential detection of long genes upon using this technique. To remove such technical detection biases, we propose a normalization strategy for a more accurate comparison of nuclear and cellular data. Finally, we show successful integration of scRNA-seq and snRNA-seq data sets with existing bioinformatic tools. Overall, our results illustrate the applicability of snRNA-seq for the characterization of cellular diversity in the adipose tissue.
Collapse
Affiliation(s)
- Anushka Gupta
- University of California at Berkeley-University of California at San Francisco Graduate Program in Bioengineering, Berkeley, California 94720, USA
| | - Farnaz Shamsi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Nicolas Altemose
- University of California at Berkeley-University of California at San Francisco Graduate Program in Bioengineering, Berkeley, California 94720, USA
| | - Gabriel F Dorlhiac
- Biophysics Graduate Group, University of California at Berkeley, Berkeley, California 94720, USA
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Andrew P White
- Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Nir Yosef
- Center for Computational Biology, University of California, Berkeley, Berkeley, California 94720, USA
- Department of Electrical Engineering and Computer Sciences, University of California at Berkeley, Berkeley, California 94720, USA
- Chan Zuckerberg Biohub, San Francisco, California 94158, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts 02139, USA
| | | | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Aaron Streets
- University of California at Berkeley-University of California at San Francisco Graduate Program in Bioengineering, Berkeley, California 94720, USA
- Biophysics Graduate Group, University of California at Berkeley, Berkeley, California 94720, USA
- Chan Zuckerberg Biohub, San Francisco, California 94158, USA
| |
Collapse
|
50
|
Okabe K, Uchiyama S. Intracellular thermometry uncovers spontaneous thermogenesis and associated thermal signaling. Commun Biol 2021; 4:1377. [PMID: 34887517 PMCID: PMC8660847 DOI: 10.1038/s42003-021-02908-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/17/2021] [Indexed: 02/06/2023] Open
Abstract
Conventional thermal biology has elucidated the physiological function of temperature homeostasis through spontaneous thermogenesis and responses to variations in environmental temperature in organisms. In addition to research on individual physiological phenomena, the molecular mechanisms of fever and physiological events such as temperature-dependent sex determination have been intensively addressed. Thermosensitive biomacromolecules such as heat shock proteins (HSPs) and transient receptor potential (TRP) channels were systematically identified, and their sophisticated functions were clarified. Complementarily, recent progress in intracellular thermometry has opened new research fields in thermal biology. High-resolution intracellular temperature mapping has uncovered thermogenic organelles, and the thermogenic functions of brown adipocytes were ascertained by the combination of intracellular thermometry and classic molecular biology. In addition, intracellular thermometry has introduced a new concept, "thermal signaling", in which temperature variation within biological cells acts as a signal in a cascade of intriguing biological events.
Collapse
Affiliation(s)
- Kohki Okabe
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.
- JST, PRESTO, Saitama, Japan.
| | - Seiichi Uchiyama
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|