1
|
Duda M, Joshi P, Borodziuk A, Sobczak K, Sikora-Dobrowolska B, Maćkowski S, Dennis AM, Kłopotowski Ł. Multimodal Temperature Readout Boosts the Performance of CuInS 2/ZnS Quantum Dot Nanothermometers. ACS APPLIED MATERIALS & INTERFACES 2024; 16:60008-60017. [PMID: 39437320 DOI: 10.1021/acsami.4c14541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Fluorescent nanothermometers are positioned to revolutionize research into cell functions and provide strategies for early diagnostics. Fluorescent nanostructures hold particular promise to fulfill this potential if nontoxic, stable varieties allowing for precise temperature measurement with high thermal sensitivities can be fabricated. In this work, we investigate the performance of micelle-encapsulated CuInS2/ZnS core/shell colloidal quantum dots (QDs) as fluorescent nanothermometers. We demonstrate four temperature readout modes, which are based on variations in the photoluminescence intensity, energy, and lifetime and on a specific ratio of excitation efficiencies. We further leverage this multimodal readout to construct a fifth, multiparametric thermometer calibration based on the multiple linear regression (MLR) model. We show that the MLR approach boosts the thermometer sensitivity by up to 7-fold while reducing the readout error by about a factor of 3. As a result, our QDs offer the highest sensitivities among semiconducting QDs emitting in the first biological window. The obtained results indicate that CuInS2/ZnS QDs are excellent candidates for intracellular in vivo thermometry and provide guidelines for further optimization of their performance.
Collapse
Affiliation(s)
- Magdalena Duda
- Institute of Physics, Polish Academy of Sciences, 02-668 Warsaw, Poland
| | - Pushkar Joshi
- Institute of Physics, Polish Academy of Sciences, 02-668 Warsaw, Poland
| | - Anna Borodziuk
- Institute of Physics, Polish Academy of Sciences, 02-668 Warsaw, Poland
| | - Kamil Sobczak
- University of Warsaw Biological and Chemical Research Centre, 02-089 Warsaw, Poland
| | | | - Sebastian Maćkowski
- Institute of Physics, Faculty of Physics, Nicolaus Copernicus University, Astronomy and Informatics, 87-100 Toruń, Poland
| | - Allison M Dennis
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | | |
Collapse
|
2
|
Ding F, Zheng P, Yan XY, Chen HJ, Fang HT, Luo YY, Peng YX, Zhang L, Yan YE. Adipocyte-secreted PRELP promotes adipocyte differentiation and adipose tissue fibrosis by binding with p75 NTR to activate FAK/MAPK signaling. Int J Biol Macromol 2024; 279:135376. [PMID: 39244119 DOI: 10.1016/j.ijbiomac.2024.135376] [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: 05/03/2024] [Revised: 08/20/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Adipocyte-secreted factors intricately regulate adipose tissue function, and the underlying molecular mechanisms are only partially understood. However, the function of PRELP, which is a key component of the extracellular matrix (ECM) in adipocytes, remains largely unknown. In this study, we demonstrate that PRELP was upregulated in both obese humans and mice, which exhibited a positive correlation with metabolic disorders. PRELP knockout could resist HFD-induced obesity and inhibit adipocyte differentiation. PRELP knockout improved glucose tolerance, insulin sensitivity and alleviated adipose tissue fibrosis. Mechanistically, PRELP was secreted into the ECM and bound to the extracellular domain of its receptor p75NTR in adipocytes, which further activated the FAK/MAPK (JNK, p38 MAPK, ERK1/2) signaling pathway, promoting adipocyte differentiation and exacerbating adipocyte fibrosis. Adipocyte PRELP plays a pivotal role in regulating obesity and adipose tissue fibrosis through an autocrine manner, and PRELP may be a therapeutic target for obesity and its related metabolic disorders.
Collapse
Affiliation(s)
- Fei Ding
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Peng Zheng
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Xi-Yue Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Hui-Jian Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Hong-Ting Fang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Yuan-Yuan Luo
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Yu-Xuan Peng
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Li Zhang
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - You-E Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China.
| |
Collapse
|
3
|
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
|
4
|
Davidsen LI, Hagberg CE, Goitea V, Lundby SM, Larsen S, Ebbesen MF, Stanic N, Topel H, Kornfeld JW. Mouse vascularized adipose spheroids: an organotypic model for thermogenic adipocytes. Front Endocrinol (Lausanne) 2024; 15:1396965. [PMID: 38982992 PMCID: PMC11231189 DOI: 10.3389/fendo.2024.1396965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/30/2024] [Indexed: 07/11/2024] Open
Abstract
Adipose tissues, particularly beige and brown adipose tissue, play crucial roles in energy metabolism. Brown adipose tissues' thermogenic capacity and the appearance of beige cells within white adipose tissue have spurred interest in their metabolic impact and therapeutic potential. Brown and beige fat cells, activated by environmental factors like cold exposure or by pharmacology, share metabolic mechanisms that drive non-shivering thermogenesis. Understanding these two cell types requires advanced, yet broadly applicable in vitro models that reflect the complex microenvironment and vasculature of adipose tissues. Here we present mouse vascularized adipose spheroids of the stromal vascular microenvironment from inguinal white adipose tissue, a tissue with 'beiging' capacity in mice and humans. We show that adding a scaffold improves vascular sprouting, enhances spheroid growth, and upregulates adipogenic markers, thus reflecting increased adipocyte maturity. Transcriptional profiling via RNA sequencing revealed distinct metabolic pathways upregulated in our vascularized adipose spheroids, with increased expression of genes involved in glucose metabolism, lipid metabolism, and thermogenesis. Functional assessment demonstrated increased oxygen consumption in vascularized adipose spheroids compared to classical 2D cultures, which was enhanced by β-adrenergic receptor stimulation correlating with elevated β-adrenergic receptor expression. Moreover, stimulation with the naturally occurring adipokine, FGF21, induced Ucp1 mRNA expression in the vascularized adipose spheroids. In conclusion, vascularized inguinal white adipose tissue spheroids provide a physiologically relevant platform to study how the stromal vascular microenvironment shapes adipocyte responses and influence activated thermogenesis in beige adipocytes.
Collapse
Affiliation(s)
- Laura Ingeborg Davidsen
- Functional Genomics and Metabolism Research Unit, Institute of Biochemistry and Molecular Biology, Faculty of Science, University of Southern Denmark, Odense, Denmark
| | - Carolina E. Hagberg
- Division of Cardiovascular Medicine, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Victor Goitea
- Functional Genomics and Metabolism Research Unit, Institute of Biochemistry and Molecular Biology, Faculty of Science, University of Southern Denmark, Odense, Denmark
- Novo Nordisk Foundation Center for Adipocyte Signaling (ADIPOSIGN), University of Southern Denmark, Odense, Denmark
| | - Stine Meinild Lundby
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Steen Larsen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Frendø Ebbesen
- Danish Molecular Biomedical Imaging Center (DaMBIC), Institute of Biochemistry and Molecular Biology, Faculty of Science, University of Southern Denmark, Odense, Denmark
| | - Natasha Stanic
- Functional Genomics and Metabolism Research Unit, Institute of Biochemistry and Molecular Biology, Faculty of Science, University of Southern Denmark, Odense, Denmark
| | - Hande Topel
- Functional Genomics and Metabolism Research Unit, Institute of Biochemistry and Molecular Biology, Faculty of Science, University of Southern Denmark, Odense, Denmark
- Novo Nordisk Foundation Center for Adipocyte Signaling (ADIPOSIGN), University of Southern Denmark, Odense, Denmark
| | - Jan-Wilhelm Kornfeld
- Functional Genomics and Metabolism Research Unit, Institute of Biochemistry and Molecular Biology, Faculty of Science, University of Southern Denmark, Odense, Denmark
- Novo Nordisk Foundation Center for Adipocyte Signaling (ADIPOSIGN), University of Southern Denmark, Odense, Denmark
| |
Collapse
|
5
|
Okabe K, Sotoma S, Harada Y. Cellular Thermal Biology Using Fluorescent Nanothermometers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1461:97-108. [PMID: 39289276 DOI: 10.1007/978-981-97-4584-5_7] [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: 09/19/2024]
Abstract
It has been known that cells have mechanisms to sense and respond to environmental noxiousness and mild temperature changes, such as heat shock response and thermosensitive TRP channels. Meanwhile, new methods of measuring temperature at the cellular level has recently been developed using fluorescent nanothermometers. Among these thermometers, fluorescent polymeric thermometers and fluorescent nanodiamonds excel in the properties required for intracellular thermometry. By using these novel methods to measure the temperature of single cells in cultures and tissues, it was revealed that spontaneous spatiotemporal temperature fluctuations occur within cells. Furthermore, the temperature fluctuations were related to organelles such as mitochondria and cellular and physiological functions, revealing a close relationship between intracellular temperature and cellular functions.
Collapse
Affiliation(s)
- Kohki Okabe
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.
- JST, PRESTO, Saitama, Japan.
| | - Shingo Sotoma
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Kyoto, Japan.
| | - Yoshie Harada
- Institute for Protein Research, Osaka University, Osaka, Japan
- Center for Quantum Information and Quantum Biology, Osaka, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Osaka, Japan
| |
Collapse
|
6
|
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
|
7
|
Shi F, Collins S. Regulation of mTOR Signaling: Emerging Role of Cyclic Nucleotide-Dependent Protein Kinases and Implications for Cardiometabolic Disease. Int J Mol Sci 2023; 24:11497. [PMID: 37511253 PMCID: PMC10380887 DOI: 10.3390/ijms241411497] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) kinase is a central regulator of cell growth and metabolism. It is the catalytic subunit of two distinct large protein complexes, mTOR complex 1 (mTORC1) and mTORC2. mTOR activity is subjected to tight regulation in response to external nutrition and growth factor stimulation. As an important mechanism of signaling transduction, the 'second messenger' cyclic nucleotides including cAMP and cGMP and their associated cyclic nucleotide-dependent kinases, including protein kinase A (PKA) and protein kinase G (PKG), play essential roles in mediating the intracellular action of a variety of hormones and neurotransmitters. They have also emerged as important regulators of mTOR signaling in various physiological and disease conditions. However, the mechanism by which cAMP and cGMP regulate mTOR activity is not completely understood. In this review, we will summarize the earlier work establishing the ability of cAMP to dampen mTORC1 activation in response to insulin and growth factors and then discuss our recent findings demonstrating the regulation of mTOR signaling by the PKA- and PKG-dependent signaling pathways. This signaling framework represents a new non-canonical regulation of mTOR activity that is independent of AKT and could be a novel mechanism underpinning the action of a variety of G protein-coupled receptors that are linked to the mTOR signaling network. We will further review the implications of these signaling events in the context of cardiometabolic disease, such as obesity, non-alcoholic fatty liver disease, and cardiac remodeling. The metabolic and cardiac phenotypes of mouse models with targeted deletion of Raptor and Rictor, the two essential components for mTORC1 and mTORC2, will be summarized and discussed.
Collapse
Affiliation(s)
- Fubiao Shi
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Chen K, Cheong LY, Gao Y, Zhang Y, Feng T, Wang Q, Jin L, Honoré E, Lam KSL, Wang W, Hui X, Xu A. Adipose-targeted triiodothyronine therapy counteracts obesity-related metabolic complications and atherosclerosis with negligible side effects. Nat Commun 2022; 13:7838. [PMID: 36539421 PMCID: PMC9767940 DOI: 10.1038/s41467-022-35470-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Thyroid hormone (TH) is a thermogenic activator with anti-obesity potential. However, systemic TH administration has no obvious clinical benefits on weight reduction. Herein we selectively delivered triiodothyronine (T3) to adipose tissues by encapsulating T3 in liposomes modified with an adipose homing peptide (PLT3). Systemic T3 administration failed to promote thermogenesis in brown and white adipose tissues (WAT) due to a feedback suppression of sympathetic innervation. PLT3 therapy effectively obviated this feedback suppression on adrenergic inputs, and potently induced browning and thermogenesis of WAT, leading to alleviation of obesity, glucose intolerance, insulin resistance, and fatty liver in obese mice. Furthermore, PLT3 was much more effective than systemic T3 therapy in reducing hypercholesterolemia and atherosclerosis in apoE-deficient mice. These findings uncover WAT as a viable target mediating the therapeutic benefits of TH and provide a safe and efficient therapeutic strategy for obesity and its complications by delivering TH to adipose tissue.
Collapse
Affiliation(s)
- Kang Chen
- grid.194645.b0000000121742757State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong China ,grid.194645.b0000000121742757Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China ,grid.194645.b0000000121742757Dr Li Dak-Sum Research Centre, The University of Hong Kong-Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Lai Yee Cheong
- grid.194645.b0000000121742757State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong China ,grid.194645.b0000000121742757Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yuan Gao
- grid.194645.b0000000121742757State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong China ,grid.194645.b0000000121742757Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yaming Zhang
- grid.194645.b0000000121742757State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong China ,grid.194645.b0000000121742757Dr Li Dak-Sum Research Centre, The University of Hong Kong-Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Pokfulam, Hong Kong China ,grid.194645.b0000000121742757Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Tianshi Feng
- grid.194645.b0000000121742757State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong China ,grid.194645.b0000000121742757Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Qin Wang
- grid.194645.b0000000121742757State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong China ,grid.194645.b0000000121742757Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Leigang Jin
- grid.194645.b0000000121742757State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong China ,grid.194645.b0000000121742757Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Eric Honoré
- Université Côte d’Azur, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut de Pharmacologie Moléculaire et Cellulaire, Labex ICST, Valbonne, France
| | - Karen S. L. Lam
- grid.194645.b0000000121742757State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong China ,grid.194645.b0000000121742757Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Weiping Wang
- grid.194645.b0000000121742757State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong China ,grid.194645.b0000000121742757Dr Li Dak-Sum Research Centre, The University of Hong Kong-Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Pokfulam, Hong Kong China ,grid.194645.b0000000121742757Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Xiaoyan Hui
- grid.194645.b0000000121742757State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong China ,grid.194645.b0000000121742757Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- grid.194645.b0000000121742757State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong China ,grid.194645.b0000000121742757Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
11
|
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: 4] [Impact Index Per Article: 2.0] [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
|
12
|
Lednovich KR, Gough S, Brenner M, Qadri T, Layden BT. G
Protein‐Coupled Receptors in Metabolic Disease. GPCRS AS THERAPEUTIC TARGETS 2022:521-552. [DOI: 10.1002/9781119564782.ch15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
13
|
Becerril S, Tuero C, Cienfuegos JA, Rodríguez A, Catalán V, Ramírez B, Valentí V, Moncada R, Unamuno X, Gómez-Ambrosi J, Frühbeck G. Improved Adipose Tissue Function after Single Anastomosis Duodeno-Ileal Bypass with Sleeve-Gastrectomy (SADI-S) in Diet-Induced Obesity. Int J Mol Sci 2022; 23:ijms231911641. [PMID: 36232953 PMCID: PMC9570280 DOI: 10.3390/ijms231911641] [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: 07/27/2022] [Revised: 09/27/2022] [Accepted: 09/27/2022] [Indexed: 11/09/2022] Open
Abstract
Bariatric surgery has been recognized as the safest and most effective procedure for controlling type 2 diabetes (T2D) and obesity in carefully selected patients. The aim of the present study was to compare the effects of Sleeve Gastrectomy (SG) and Single Anastomosis Duodenoileal Bypass with SG (SADI-S) on the metabolic profile of diet-induced obese rats. A total of 35 four-week-old male Wistar rats were submitted to surgical interventions (sham operation, SG and SADI-S) after 4 months of being fed a high-fat diet. Body weight, metabolic profile and the expression of molecules involved in the control of subcutaneous white (SCWAT), brown (BAT) and beige (BeAT) adipose tissue function were analyzed. SADI-S surgery was associated with significantly decreased amounts of total fat pads (p < 0.001) as well as better control of lipid and glucose metabolism compared to the SG counterparts. An improved expression of molecules involved in fat browning in SCWAT and in the control of BAT and BeAT differentiation and function was observed following SADI-S. Together, our findings provide evidence that the enhanced metabolic improvement and their continued durability after SADI-S compared to SG rely, at least in part, on the improvement of the BeAT phenotype and function.
Collapse
Affiliation(s)
- Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
- Correspondence:
| | - Carlota Tuero
- Department of Surgery, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Javier A. Cienfuegos
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Surgery, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Víctor Valentí
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Surgery, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Rafael Moncada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Anesthesia, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Xabier Unamuno
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Medical Engineering Laboratory, University of Navarra, 31008 Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| |
Collapse
|
14
|
Chen Z, Cai Z, Liu W, Yan Z. Optical trapping and manipulation for single-particle spectroscopy and microscopy. J Chem Phys 2022; 157:050901. [DOI: 10.1063/5.0086328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Optical tweezers can control the position and orientation of individual colloidal particles in solution. Such control is often desirable but challenging for single-particle spectroscopy and microscopy, especially at the nanoscale. Functional nanoparticles that are optically trapped and manipulated in a three-dimensional (3D) space can serve as freestanding nanoprobes, which provide unique prospects of sensing and mapping the surrounding environment of the nanoparticles and studying their interactions with biological systems. In this perspective, we will first describe the optical forces underlying the optical trapping and manipulation of microscopic particles, then review the combinations and applications of different spectroscopy and microscopy techniques with optical tweezers. Finally, we will discuss the challenges of performing spectroscopy and microscopy on single nanoparticles with optical tweezers, the possible routes to address these challenges, and the new opportunities that will arise.
Collapse
Affiliation(s)
- Zhenzhen Chen
- The University of North Carolina at Chapel Hill, United States of America
| | - Zhewei Cai
- Clarkson University, United States of America
| | - Wenbo Liu
- The University of North Carolina at Chapel Hill, United States of America
| | - Zijie Yan
- University of North Carolina at Chapel Hill, United States of America
| |
Collapse
|
15
|
Zhang C, Yu J, Yang C, Shang S, Lv X, Cui B, Hua F. Crosstalk between ferroptosis and stress-Implications in cancer therapeutic responses. CANCER INNOVATION 2022; 1:92-113. [PMID: 38089453 PMCID: PMC10686180 DOI: 10.1002/cai2.7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/15/2022] [Indexed: 07/05/2024]
Abstract
Ferroptosis is a newly discovered form of cell death that is characterized by the accumulation of iron-dependent lipid peroxidation. Research on ferroptosis has seen exponential growth over the past few years. Tumor cells are strongly dependent on iron for their growth, which makes them develop mechanisms to increase iron uptake and inhibit iron output, thereby completing iron accumulation. Ferroptosis can be induced or inhibited by various stresses through multiple mechanisms, making it stands at the crossroads of stresses related cancer cell fate determination. In this review, we give a brief summary of ferroptosis hallmarks and provide a systematic analysis of the current molecular mechanisms and regulatory networks of diverse stress conditions on ferroptosis. We also discuss the relationships between ferroptosis and cancer therapy responses to further understand potential targets and therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Cheng Zhang
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Jiao‐jiao Yu
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Chen Yang
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Shuang Shang
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Xiao‐xi Lv
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Bing Cui
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Fang Hua
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| |
Collapse
|
16
|
Alizadeh B, Salehzadeh A, Ranji N, Arasteh A. Effects of N-Acetyl Cysteine on Genes Expression of c-myc, and Ask-1, Histopathological, Oxidative Stress, Inflammation, and Apoptosis in the Liver of Male Rats Exposed to Cadmium. Biol Trace Elem Res 2022; 200:661-668. [PMID: 33740178 DOI: 10.1007/s12011-021-02670-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/08/2021] [Indexed: 12/18/2022]
Abstract
This study aimed to consider the oxidative damage induced by cadmium (Cd) and apoptosis and the role of N-acetylcysteine (NAC) in preserving hepatic cells against Cd toxicity. Male rats were randomly divided into seven groups including G1 (control), G2 (single dose of Cd), G3 (continuous dose of Cd), G4 (single dose of Cd + continuous dose of NAC), and G5 (continuous dose of Cd + continuous dose of NAC). Hepatic cells apoptosis was measured using TUNEL assay method. Levels of malondialdehyde (MDA), TNF-α, IL-10, and total antioxidant capacity (TAC) were measured by specific kits. Expression of c-myc and Ask-1 genes was considered using RT-PCR. NAC treatments significantly improved TAC and IL-10, but decreased MDA and TNF-α values in rats that were exposed to a single and continuous dose of Cd (p < 0.05). Exposure to a single and continuous dose of Cd caused a significant increase in c-myc expression by 3.76-fold (p < 0.001) and 8.17-fold (p < 0.0001), respectively. Single and continuous dose treatment of Cd led to a significant increase in Ask1 expression by 4.38-fold (p < 0.001) and 13.52-fold (p < 0.001), respectively. NAC treatments significantly decreased the expression of c-myc, and Ask-1 in rats exposed to single or continuous Cd. Cd exposure is strongly associated with oxidative stress, inflammation, antioxidant depletion, and liver cells apoptosis. NAC can protect liver tissue against Cd by elevating antioxidants capacity, mitigating oxidative stress and inflammation, as well as down-regulating of apoptotic genes.
Collapse
Affiliation(s)
- Bentolhoda Alizadeh
- Department of Biology, Faculty of Sciences, Rasht Branch, Islamic Azad University, Rasht, Iran
| | - Ali Salehzadeh
- Department of Biology, Faculty of Sciences, Rasht Branch, Islamic Azad University, Rasht, Iran.
| | - Najmeh Ranji
- Department of Biology, Faculty of Sciences, Rasht Branch, Islamic Azad University, Rasht, Iran
| | - Amir Arasteh
- Department of Biology, Faculty of Sciences, Rasht Branch, Islamic Azad University, Rasht, Iran
| |
Collapse
|
17
|
ASK1 signaling regulates phase-specific glial interactions during neuroinflammation. Proc Natl Acad Sci U S A 2022; 119:2103812119. [PMID: 35101972 PMCID: PMC8832969 DOI: 10.1073/pnas.2103812119] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2021] [Indexed: 01/08/2023] Open
Abstract
Neuroinflammation is associated with many neurodegenerative diseases such as Alzheimer’s disease and multiple sclerosis (MS). Thus, decreasing neuroinflammation may be a promising treatment for these diseases. Apoptosis signal-related kinase 1 (ASK1) has been shown to cause neuroinflammation in neurodegenerative disease models, but its mechanism of action has been unclear. Here, we generated conditional knockout mice that lack ASK1 in T cells, dendritic cells, microglia/macrophages, microglia, or astrocytes, to assess the roles of ASK1 during experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. We propose that ASK1 is required in microglia and astrocytes to cause and maintain neuroinflammation by a feedback loop between these two cell types. Neuroinflammation is well known to be associated with neurodegenerative diseases. Apoptosis signal-regulating kinase 1 (ASK1) is a mitogen-activated protein kinase kinase kinase that has been implicated in neuroinflammation, but its precise cellular and molecular mechanisms remain unknown. In this study, we generated conditional knockout (CKO) mice that lack ASK1 in T cells, dendritic cells, microglia/macrophages, microglia, or astrocytes, to assess the roles of ASK1 during experimental autoimmune encephalomyelitis (EAE). We found that neuroinflammation was reduced in both the early and later stages of EAE in microglia/macrophage-specific ASK1 knockout mice, whereas only the later-stage neuroinflammation was ameliorated in astrocyte-specific ASK1 knockout mice. ASK1 deficiency in T cells and dendritic cells had no significant effects on EAE severity. Further, we found that ASK1 in microglia/macrophages induces a proinflammatory environment, which subsequently activates astrocytes to exacerbate neuroinflammation. Microglia-specific ASK1 deletion was achieved using a CX3CR1CreER system, and we found that ASK1 signaling in microglia played a major role in generating and maintaining disease. Activated astrocytes produce key inflammatory mediators, including CCL2, that further activated and recruited microglia/macrophages, in an astrocytic ASK1-dependent manner. Astrocyte-specific analysis revealed CCL2 expression was higher in the later stage compared with the early stage, suggesting a greater proinflammatory role of astrocytes in the later stage. Our findings demonstrate cell-type–specific roles of ASK1 and suggest phase-specific ASK1-dependent glial cell interactions in EAE pathophysiology. We propose glial ASK1 as a promising therapeutic target for reducing neuroinflammation.
Collapse
|
18
|
Feng G, Zhang H, Zhu X, Zhang J, Fang J. Fluorescence Thermometer: Intermediation of the Fontal Temperature and Light. Biomater Sci 2022; 10:1855-1882. [DOI: 10.1039/d1bm01912k] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The rapid advance of thermal materials and fluorescence spectroscopy has extensively promoted micro-scale fluorescence thermometry development in recent years. Based on the advantages of fast response, high sensitivity, simple operation,...
Collapse
|
19
|
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: 15] [Impact Index Per Article: 5.0] [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
|
20
|
Takayanagi S, Watanabe K, Maruyama T, Ogawa M, Morishita K, Soga M, Hatta T, Natsume T, Hirano T, Kagechika H, Hattori K, Naguro I, Ichijo H. ASKA technology-based pull-down method reveals a suppressive effect of ASK1 on the inflammatory NOD-RIPK2 pathway in brown adipocytes. Sci Rep 2021; 11:22009. [PMID: 34759307 PMCID: PMC8581049 DOI: 10.1038/s41598-021-01123-7] [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: 09/18/2020] [Accepted: 10/22/2021] [Indexed: 11/13/2022] Open
Abstract
Recent studies have shown that adipose tissue is an immunological organ. While inflammation in energy-storing white adipose tissues has been the focus of intense research, the regulatory mechanisms of inflammation in heat-producing brown adipose tissues remain largely unknown. We previously identified apoptosis signal-regulating kinase 1 (ASK1) as a critical regulator of brown adipocyte maturation; the PKA-ASK1-p38 axis facilitates uncoupling protein 1 (UCP1) induction cell-autonomously. Here, we show that ASK1 suppresses an innate immune pathway and contributes to maintenance of brown adipocytes. We report a novel chemical pull-down method for endogenous kinases using analog sensitive kinase allele (ASKA) technology and identify an ASK1 interactor in brown adipocytes, receptor-interacting serine/threonine-protein kinase 2 (RIPK2). ASK1 disrupts the RIPK2 signaling complex and inhibits the NOD-RIPK2 pathway to downregulate the production of inflammatory cytokines. As a potential biological significance, an in vitro model for intercellular regulation suggests that ASK1 facilitates the expression of UCP1 through the suppression of inflammatory cytokine production. In parallel to our previous report on the PKA-ASK1-p38 axis, our work raises the possibility of an auxiliary role of ASK1 in brown adipocyte maintenance through neutralizing the thermogenesis-suppressive effect of the NOD-RIPK2 pathway.
Collapse
Affiliation(s)
- Saki Takayanagi
- grid.26999.3d0000 0001 2151 536XLaboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Kengo Watanabe
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Takeshi Maruyama
- grid.26999.3d0000 0001 2151 536XLaboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Motoyuki Ogawa
- grid.26999.3d0000 0001 2151 536XLaboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Kazuhiro Morishita
- grid.26999.3d0000 0001 2151 536XLaboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Mayumi Soga
- grid.26999.3d0000 0001 2151 536XLaboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Tomohisa Hatta
- grid.208504.b0000 0001 2230 7538Molecular Profiling Research Center for Drug Discovery, The National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo, 135-0064 Japan
| | - Tohru Natsume
- grid.208504.b0000 0001 2230 7538Cellular and Molecular Biotechnology Research Institute, The National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo, 135-0064 Japan
| | - Tomoya Hirano
- grid.265073.50000 0001 1014 9130Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kandasurugadai, Chiyoda-ku, Tokyo, 101-0062 Japan ,Present Address: Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094 Japan
| | - Hiroyuki Kagechika
- grid.265073.50000 0001 1014 9130Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kandasurugadai, Chiyoda-ku, Tokyo, 101-0062 Japan
| | - Kazuki Hattori
- grid.26999.3d0000 0001 2151 536XLaboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Isao Naguro
- grid.26999.3d0000 0001 2151 536XLaboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
21
|
Glycogen metabolism links glucose homeostasis to thermogenesis in adipocytes. Nature 2021; 599:296-301. [PMID: 34707293 PMCID: PMC9186421 DOI: 10.1038/s41586-021-04019-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 09/13/2021] [Indexed: 12/13/2022]
Abstract
Adipocytes increase energy expenditure in response to prolonged sympathetic activation via persistent expression of uncoupling protein 1 (UCP1)1,2. Here we report that the regulation of glycogen metabolism by catecholamines is critical for UCP1 expression. Chronic β-adrenergic activation leads to increased glycogen accumulation in adipocytes expressing UCP1. Adipocyte-specific deletion of a scaffolding protein, protein targeting to glycogen (PTG), reduces glycogen levels in beige adipocytes, attenuating UCP1 expression and responsiveness to cold or β-adrenergic receptor-stimulated weight loss in obese mice. Unexpectedly, we observed that glycogen synthesis and degradation are increased in response to catecholamines, and that glycogen turnover is required to produce reactive oxygen species leading to the activation of p38 MAPK, which drives UCP1 expression. Thus, glycogen has a key regulatory role in adipocytes, linking glucose metabolism to thermogenesis.
Collapse
|
22
|
Treatment with atrial natriuretic peptide induces adipose tissue browning and exerts thermogenic actions in vivo. Sci Rep 2021; 11:17466. [PMID: 34465848 PMCID: PMC8408225 DOI: 10.1038/s41598-021-96970-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/18/2021] [Indexed: 01/14/2023] Open
Abstract
Increasing evidence suggests natriuretic peptides (NPs) coordinate inter-organ metabolic crosstalk with adipose tissues and play a critical role in energy metabolism. We recently reported A-type NP (ANP) raises intracellular temperature in cultured adipocytes in a low-temperature-sensitive manner. We herein investigated whether exogenous ANP-treatment exerts a significant impact on adipose tissues in vivo. Mice fed a high-fat-diet (HFD) or normal-fat-diet (NFD) for 13 weeks were treated with or without ANP infusion subcutaneously for another 3 weeks. ANP-treatment significantly ameliorated HFD-induced insulin resistance. HFD increased brown adipose tissue (BAT) cell size with the accumulation of lipid droplets (whitening), which was suppressed by ANP-treatment (re-browning). Furthermore, HFD induced enlarged lipid droplets in inguinal white adipose tissue (iWAT), crown-like structures in epididymal WAT, and hepatic steatosis, all of which were substantially attenuated by ANP-treatment. Likewise, ANP-treatment markedly increased UCP1 expression, a specific marker of BAT, in iWAT (browning). ANP also further increased UCP1 expression in BAT with NFD. Accordingly, cold tolerance test demonstrated ANP-treated mice were tolerant to cold exposure. In summary, exogenous ANP administration ameliorates HFD-induced insulin resistance by attenuating hepatic steatosis and by inducing adipose tissue browning (activation of the adipose tissue thermogenic program), leading to in vivo thermogenesis during cold exposure.
Collapse
|
23
|
Wang Z, Zhu M, Wang M, Gao Y, Zhang C, Liu S, Qu S, Liu Z, Zhang C. Integrated Multiomic Analysis Reveals the High-Fat Diet Induced Activation of the MAPK Signaling and Inflammation Associated Metabolic Cascades via Histone Modification in Adipose Tissues. Front Genet 2021; 12:650863. [PMID: 34262592 PMCID: PMC8273343 DOI: 10.3389/fgene.2021.650863] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/10/2021] [Indexed: 11/13/2022] Open
Abstract
Background The number of diet induced obese population is increasing every year, and the incidence of type 2 diabetes is also on the rise. Histone methylation and acetylation have been shown to be associated with lipogenesis and obesity by manipulating gene expression via the formation of repression or activation domains on chromosomes. Objective In this study, we aimed to explore gene activation or repression and related biological processes by histone modification across the whole genome on a high-fat diet (HFD) condition. We also aimed to elucidate the correlation of these genes that modulated by histone modification with energy metabolism and inflammation under both short-term and long-term HFD conditions. Method We performed ChIP-seq analysis of H3K9me2 and H3K9me3 in brown and white adipose tissues (WATs; subcutaneous adipose tissue) from mice fed with a standard chow diet (SCD) or HFD and a composite analysis of the histone modification of H3K9me2, H3K9me3, H3K4me1 and H3K27ac throughout the whole genome. We also employed and integrated two bulk RNA-seq and a single-nuclei RNA sequencing dataset and performed western blotting (WB) to confirm the gene expression levels in adipose tissue of the SCD and HFD groups. Results The ChIP-seq and transcriptome analysis of mouse adipose tissues demonstrated that a series of genes were activated by the histone modification of H3K9me2, H3K9me3, H3K4me1, and H3K27ac in response to HFD condition. These genes were enriched in Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways involved in lipogenesis, energy metabolism and inflammation. Several genes in the activated mitogen-activated protein kinase (MAPK) pathway might be related to both inflammation and energy metabolism in mice, rats and humans fed with HFD for a short or long term, as showed by bulk RNA-seq and single nuclei RNA-seq datasets. Western blot analyses further confirmed the increased expression of MET, VEGFA and the enhanced phosphorylation ratio of p44/42 MAPK upon HFD treatment. Conclusion This study expanded our understanding of the influence of eating behavior on obesity and could assist the identification of putative therapeutic targets for the prevention and treatment of metabolic disorders in the future.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Zhu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Meng Wang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yihui Gao
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Cong Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shangyun Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shen Qu
- Department of Endocrinology and Metabolism, National Metabolic Management Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhongmin Liu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Department of Cardiac Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chao Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
24
|
Zhang Z, Wang L, Liu Y, Luan Y, Zhu K, Tian Y, Liu Y, Zheng X. Activation of type 4 metabotropic glutamate receptor attenuates oxygen and glucose deprivation-induced apoptosis in human neural stem cells via inhibition of ASK1-p38 signaling pathway. Brain Res 2021; 1767:147561. [PMID: 34133989 DOI: 10.1016/j.brainres.2021.147561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/19/2021] [Accepted: 06/11/2021] [Indexed: 10/21/2022]
Abstract
Hypoxic ischemic brain injury (HIBI) has been one of the most severe central nervous system (CNS) diseases with high fatality and disability rate. Neural stem cells (NSCs) persist in the mammalian brain throughout life and NSCs-associated therapies might be a promising strategy for the HIBI treatment. In this study, we identified that type 4 metabotropic glutamate receptor (mGluR4) was expressed in cultured human NSCs (hNSCs) isolated from the human fetus cortex and further established the oxygen and glucose deprivation (OGD) model in hNSCs to study the role of mGluR4 in hypoxic and ischemic injury. The results indicated that mGluR4 activation by using VU0155041 (mGluR4-specific agonist) markedly attenuated the OGD-induced alterations in TUNEL staining, apoptosis rate, cleavages of pro-caspase-8, -9, -3, and Bcl-2/Bax expression balance. Furthermore, mGluR4 activation inhibited the ASK1/p38 signaling pathway. Asiatic acid, as a p38 MAPK activator, is capable of abolishing the neuroprotective effect of mGluR4, while both NQDI-1 (ASK-1 inhibitor) and SB203580 (p38 MAPK inhibitor) exerted similar effects to VU0155041 in the OGD-induced hNSC damage. In conclusion, this study indicates that mGluR4 activation protects hNSCs against the OGD-induced cell death via inhibiting the ASK1-p38 pathway. Activation of mGluR4 might be a promising strategy for enhancing NSCs survival in hypoxic and ischemic injury.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Stomatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Li Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, China
| | - Yingfei Liu
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Yan Luan
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Kun Zhu
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710061, China
| | - Yumei Tian
- Department of Rehabilitation, Xi'an Mental Health Center, Xi'an, Shaanxi 710061, China
| | - Yong Liu
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China.
| | - Xiaoyan Zheng
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
25
|
Fujimoto M, Kamiyama M, Fuse K, Ryuno H, Odawara T, Furukawa N, Yoshimatsu Y, Watabe T, Prchal-Murphy M, Sexl V, Tahara H, Hayakawa Y, Sato T, Takeda K, Naguro I, Ichijo H. ASK1 suppresses NK cell-mediated intravascular tumor cell clearance in lung metastasis. Cancer Sci 2021; 112:1633-1643. [PMID: 33565179 PMCID: PMC8019214 DOI: 10.1111/cas.14842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2021] [Indexed: 12/16/2022] Open
Abstract
Tumor metastasis is the leading cause of death worldwide and involves an extremely complex process composed of multiple steps. Our previous study demonstrated that apoptosis signal‐regulating kinase 1 (ASK1) deficiency in mice attenuates tumor metastasis in an experimental lung metastasis model. However, the steps of tumor metastasis regulated by ASK1 remain unclear. Here, we showed that ASK1 deficiency in mice promotes natural killer (NK) cell‐mediated intravascular tumor cell clearance in the initial hours of metastasis. In response to tumor inoculation, ASK1 deficiency upregulated immune response‐related genes, including interferon‐gamma (IFNγ). We also revealed that NK cells are required for these anti‐metastatic phenotypes. ASK1 deficiency augmented cytokine production chemoattractive to NK cells possibly through induction of the ligand for NKG2D, a key activating receptor of NK cells, leading to further recruitment of NK cells into the lung. These results indicate that ASK1 negatively regulates NK cell‐dependent anti‐tumor immunity and that ASK1‐targeted therapy can provide a new tool for cancer immunotherapy to overcome tumor metastasis.
Collapse
Affiliation(s)
- Makoto Fujimoto
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Miki Kamiyama
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kosuke Fuse
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroki Ryuno
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Takeru Odawara
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Natsuki Furukawa
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yasuhiro Yoshimatsu
- Division of Pharmacology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Tetsuro Watabe
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Michaela Prchal-Murphy
- Department of Biomedical Sciences, Institute of Pharmacology and Toxicology, University of Veterinary Medicine of Vienna, Wien, Austria
| | - Veronika Sexl
- Department of Biomedical Sciences, Institute of Pharmacology and Toxicology, University of Veterinary Medicine of Vienna, Wien, Austria
| | - Hideaki Tahara
- Department of Cancer Drug Discovery and Development, Research Center, Osaka International Cancer Institute, Osaka, Japan.,Project Division of Cancer Biomolecular Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoshihiro Hayakawa
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Takehiro Sato
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kohsuke Takeda
- Division of Cell Regulation, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Isao Naguro
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
26
|
Wang X, Zhao Y, Zhou D, Tian Y, Feng G, Lu Z. Gab2 deficiency suppresses high-fat diet-induced obesity by reducing adipose tissue inflammation and increasing brown adipose function in mice. Cell Death Dis 2021; 12:212. [PMID: 33637697 PMCID: PMC7910586 DOI: 10.1038/s41419-021-03519-9] [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: 11/30/2020] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022]
Abstract
Obesity is caused by a long-term imbalance between energy intake and consumption and is regulated by multiple signals. This study investigated the effect of signaling scaffolding protein Gab2 on obesity and its relevant regulation mechanism. Gab2 knockout (KO) and wild-type (WT) mice were fed with a standard diet (SD) or high-fat diet (HFD) for 12 weeks. The results showed that the a high-fat diet-induced Gab2 expression in adipose tissues, but deletion of Gab2 attenuated weight gain and improved glucose tolerance in mice fed with a high-fat diet. White adipose tissue and systemic inflammations were reduced in HFD-fed Gab2 deficiency mice. Gab2 deficiency increased the expression of Ucp1 and other thermogenic genes in brown adipose tissue. Furthermore, the regulation of Gab2 on the mature differentiation and function of adipocytes was investigated in vitro using primary or immortalized brown preadipocytes. The expression of brown fat-selective genes was found to be elevated in differentiated adipocytes without Gab2. The mechanism of Gab2 regulating Ucp1 expression in brown adipocytes involved with its downstream PI3K (p85)-Akt-FoxO1 signaling pathway. Our research suggests that deletion of Gab2 suppresses diet-induced obesity by multiple pathways and Gab2 may be a novel therapeutic target for the treatment of obesity and associated complications.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/deficiency
- Adaptor Proteins, Signal Transducing/genetics
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/physiopathology
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/physiopathology
- Adiposity
- Animals
- Blood Glucose/metabolism
- Cell Line
- Class Ia Phosphatidylinositol 3-Kinase/metabolism
- Diet, High-Fat
- Disease Models, Animal
- Energy Metabolism
- Forkhead Box Protein O1/metabolism
- Insulin Resistance
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Obesity/genetics
- Obesity/metabolism
- Obesity/physiopathology
- Obesity/prevention & control
- Panniculitis/genetics
- Panniculitis/metabolism
- Panniculitis/physiopathology
- Panniculitis/prevention & control
- Proto-Oncogene Proteins c-akt/metabolism
- Signal Transduction
- Uncoupling Protein 1/metabolism
- Weight Gain
- Mice
Collapse
Affiliation(s)
- Xinhui Wang
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, 361005, Xiamen, Fujian, China
| | - Yinan Zhao
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, 361005, Xiamen, Fujian, China
| | - Dekun Zhou
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, 361005, Xiamen, Fujian, China
| | - Yingpu Tian
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, 361005, Xiamen, Fujian, China
| | - Gensheng Feng
- Department of Pathology, Division of Biological Sciences, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Zhongxian Lu
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, 361005, Xiamen, Fujian, China.
| |
Collapse
|
27
|
Canovas B, Nebreda AR. Diversity and versatility of p38 kinase signalling in health and disease. Nat Rev Mol Cell Biol 2021; 22:346-366. [PMID: 33504982 PMCID: PMC7838852 DOI: 10.1038/s41580-020-00322-w] [Citation(s) in RCA: 277] [Impact Index Per Article: 92.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2020] [Indexed: 02/06/2023]
Abstract
The ability of cells to deal with different types of stressful situations in a precise and coordinated manner is key for survival and involves various signalling networks. Over the past 25 years, p38 kinases — in particular, p38α — have been implicated in the cellular response to stress at many levels. These span from environmental and intracellular stresses, such as hyperosmolarity, oxidative stress or DNA damage, to physiological situations that involve important cellular changes such as differentiation. Given that p38α controls a plethora of functions, dysregulation of this pathway has been linked to diseases such as inflammation, immune disorders or cancer, suggesting the possibility that targeting p38α could be of therapeutic interest. In this Review, we discuss the organization of this signalling pathway focusing on the diversity of p38α substrates, their mechanisms and their links to particular cellular functions. We then address how the different cellular responses can be generated depending on the signal received and the cell type, and highlight the roles of this kinase in human physiology and in pathological contexts. p38α — the best-characterized member of the p38 kinase family — is a key mediator of cellular stress responses. p38α is activated by a plethora of signals and functions through a multitude of substrates to regulate different cellular behaviours. Understanding context-dependent p38α signalling provides important insights into p38α roles in physiology and pathology.
Collapse
Affiliation(s)
- Begoña Canovas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Angel R Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain. .,ICREA, Barcelona, Spain.
| |
Collapse
|
28
|
Hattori K, Wakatsuki H, Sakauchi C, Furutani S, Sugawara S, Hatta T, Natsume T, Ichijo H. β-adrenergic receptor signaling evokes the PKA-ASK axis in mature brown adipocytes. PLoS One 2020; 15:e0232645. [PMID: 33108364 PMCID: PMC7591029 DOI: 10.1371/journal.pone.0232645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/03/2020] [Indexed: 12/31/2022] Open
Abstract
Boosting energy expenditure by harnessing the activity of brown adipocytes is a promising strategy for combatting the global epidemic of obesity. Many studies have revealed that the β3-adrenergic receptor agonist is a potent activator of brown adipocytes, even in humans, and PKA and p38 MAPK have been demonstrated for regulating the transcription of a wide range of critical genes such as Ucp1. We previously revealed that the PKA-ASK1-p38 axis is activated in immature brown adipocytes and contributes to functional maturation. However, the downstream mechanisms of PKA that initiate the p38 MAPK cascade are still mostly unknown in mature brown adipocytes. Here, we identified the ASK family as a crucial signaling molecule bridging PKA and MAPK in mature brown adipocytes. Mechanistically, the phosphorylation of ASK1 at threonine 99 and serine 993 is critical in PKA-dependent ASK1 activation. Additionally, PKA also activates ASK2, which contributes to MAPK regulation. These lines of evidence provide new details for tailoring a βAR-dependent brown adipocyte activation strategy.
Collapse
Affiliation(s)
- Kazuki Hattori
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- * E-mail: (KH); (HI)
| | - Hiroaki Wakatsuki
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Chihiro Sakauchi
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shotaro Furutani
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Sho Sugawara
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Tomohisa Hatta
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Tohru Natsume
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Hidenori Ichijo
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- * E-mail: (KH); (HI)
| |
Collapse
|
29
|
Zhou J, Del Rosal B, Jaque D, Uchiyama S, Jin D. Advances and challenges for fluorescence nanothermometry. Nat Methods 2020; 17:967-980. [PMID: 32989319 DOI: 10.1038/s41592-020-0957-y] [Citation(s) in RCA: 185] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
Fluorescent nanothermometers can probe changes in local temperature in living cells and in vivo and reveal fundamental insights into biological properties. This field has attracted global efforts in developing both temperature-responsive materials and detection procedures to achieve sub-degree temperature resolution in biosystems. Recent generations of nanothermometers show superior performance to earlier ones and also offer multifunctionality, enabling state-of-the-art functional imaging with improved spatial, temporal and temperature resolutions for monitoring the metabolism of intracellular organelles and internal organs. Although progress in this field has been rapid, it has not been without controversy, as recent studies have shown possible biased sensing during fluorescence-based detection. Here, we introduce the design principles and advances in fluorescence nanothermometry, highlight application achievements, discuss scenarios that may lead to biased sensing, analyze the challenges ahead in terms of both fundamental issues and practical implementations, and point to new directions for improving this interdisciplinary field.
Collapse
Affiliation(s)
- Jiajia Zhou
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, Australia.
| | - Blanca Del Rosal
- ARC Centre of Excellence for Nanoscale Biophotonics, RMIT University, Melbourne, Australia
| | - Daniel Jaque
- Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Madrid, Spain. .,Fluorescence Imaging Group, Departamento de Física de Materiales-Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Seiichi Uchiyama
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Dayong Jin
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, Australia.,Department of Biomedical Engineering, Southern University of Science and Technology, Guangdong, China
| |
Collapse
|
30
|
Kang R, Nagoshi T, Kimura H, Tanaka TD, Yoshii A, Inoue Y, Morimoto S, Ogawa K, Minai K, Ogawa T, Kawai M, Yoshimura M. Possible Association Between Body Temperature and B-Type Natriuretic Peptide in Patients With Cardiovascular Diseases. J Card Fail 2020; 27:75-82. [PMID: 32871239 DOI: 10.1016/j.cardfail.2020.08.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 07/31/2020] [Accepted: 08/24/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND In addition to various biological effects of natriuretic peptides (NP) on cardiovascular systems, we recently reported that NP raises intracellular temperature in cultured adipocytes. We herein examined the possible thermogenic action of NP in consideration of hemodynamic parameters and inflammatory reaction by proposing structural equation models. METHODS AND RESULTS The study population consisted of 1985 consecutive patients who underwent cardiac catheterization. Covariance structure analyses were performed to clarify the direct contribution of plasma B-type NP (BNP) to body temperature (BT) by excluding other confounding factors. A hierarchical path model showed increase in BNP, increase in C-reactive protein and decrease in left ventricular ejection fraction were mutually associated. As expected, C-reactive protein was positively correlated with BT. Importantly, despite a negative correlation between BNP and left ventricular ejection fraction, a decrease in the left ventricular ejection fraction was associated with BT decrease, whereas elevation in BNP level was associated with BT increase independently of C-reactive protein level (P = .007). CONCLUSIONS Patients with LV dysfunction tend to manifest a decrease in BT, whereas BNP elevation is associated with an increase in BT independently of inflammatory response. These findings suggest the adaptive heat-retaining property of NP (and/or NP-associated factors) when BT falls owing to unfavorable hemodynamic conditions in a state of impaired cardiac function.
Collapse
Affiliation(s)
- Ryeonshi Kang
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Tomohisa Nagoshi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine.
| | - Haruka Kimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Toshikazu D Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Akira Yoshii
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Yasunori Inoue
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Satoshi Morimoto
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Kazuo Ogawa
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Kosuke Minai
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Takayuki Ogawa
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Makoto Kawai
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| |
Collapse
|
31
|
You L, Wang Y, Gao Y, Wang X, Cui X, Zhang Y, Pang L, Ji C, Guo X, Chi X. The role of microRNA-23b-5p in regulating brown adipogenesis and thermogenic program. Endocr Connect 2020; 9:457-470. [PMID: 32348962 PMCID: PMC7274556 DOI: 10.1530/ec-20-0124] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022]
Abstract
Enhanced brown adipose tissue (BAT) mass and activity have been demonstrated to promote the expenditure of excess stored energy and reduce prevalence of obesity. Cold is known as a potent stimulator of BAT and activates BAT primarily through the β3-adrenergic-cAMP signaling. Here, we performed RNA-sequencing to identify differential miRNAs in mouse BAT upon cold exposure and a total of 20 miRNAs were validated. With the treatment of CL-316,243 (CL) and forskolin (Fsk) in mouse and human differentiated brown adipocyte cells in vitro, miR-23b-5p, miR-133a-3p, miR-135-5p, miR-491-5p, and miR-150-3p expression decreased and miR-455-5p expression increased. Among these deferentially expressed miRNAs, miR-23b-5p expression was differentially regulated in activated and aging mouse BAT and negatively correlated with Ucp1 expression. Overexpression of miR-23b-5p in the precursor cells from BAT revealed no significant effects on lipid accumulation, but diminished mitochondrial function and decreased expression of BAT specific markers. Though luciferase reporter assays did not confirm the positive association of miR-23b-5p with the 3'UTRs of the predicted target Ern1, miR-23b-5p overexpression may affect brown adipocyte thermogenic capacity mainly through regulating genes expression involving in lipolysis and fatty acid β-oxidation pathways. Our results suggest that miRNAs are involved in cold-mediated BAT thermogenic activation and further acknowledged miR-23b-5p as a negative regulator in controlling thermogenic programs, further providing potential molecular therapeutic targets to increase surplus energy and treat obesity.
Collapse
Affiliation(s)
- Lianghui You
- Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
- Institute of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yan Wang
- Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
- Institute of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yao Gao
- Department of Endocrinology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Xingyun Wang
- Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xianwei Cui
- Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Yanyan Zhang
- Beijing Chaoyang Distirct Maternal and Child Health Care Hospital, Beijing, China
| | - Lingxia Pang
- Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Chenbo Ji
- Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
- Institute of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Xirong Guo
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Correspondence should be addressed to X Chi or X Guo: or
| | - Xia Chi
- Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
- Institute of Pediatrics, Nanjing Medical University, Nanjing, China
- Correspondence should be addressed to X Chi or X Guo: or
| |
Collapse
|
32
|
Sugawara S, Kanamaru Y, Sekine S, Maekawa L, Takahashi A, Yamamoto T, Watanabe K, Fujisawa T, Hattori K, Ichijo H. The mitochondrial protein PGAM5 suppresses energy consumption in brown adipocytes by repressing expression of uncoupling protein 1. J Biol Chem 2020; 295:5588-5601. [PMID: 32144202 PMCID: PMC7186182 DOI: 10.1074/jbc.ra119.011508] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 03/04/2020] [Indexed: 01/06/2023] Open
Abstract
Accumulating evidence suggests that brown adipose tissue (BAT) is a potential therapeutic target for managing obesity and related diseases. PGAM family member 5, mitochondrial serine/threonine protein phosphatase (PGAM5), is a protein phosphatase that resides in the mitochondria and regulates many biological processes, including cell death, mitophagy, and immune responses. Because BAT is a mitochondria-rich tissue, we have hypothesized that PGAM5 has a physiological function in BAT. We previously reported that PGAM5-knockout (KO) mice are resistant to severe metabolic stress. Importantly, lipid accumulation is suppressed in PGAM5-KO BAT, even under unstressed conditions, raising the possibility that PGAM5 deficiency stimulates lipid consumption. However, the mechanism underlying this observation is undetermined. Here, using an array of biochemical approaches, including quantitative RT-PCR, immunoblotting, and oxygen consumption assays, we show that PGAM5 negatively regulates energy expenditure in brown adipocytes. We found that PGAM5-KO brown adipocytes have an enhanced oxygen consumption rate and increased expression of uncoupling protein 1 (UCP1), a protein that increases energy consumption in the mitochondria. Mechanistically, we found that PGAM5 phosphatase activity and intramembrane cleavage are required for suppression of UCP1 activity. Furthermore, utilizing a genome-wide siRNA screen in HeLa cells to search for regulators of PGAM5 cleavage, we identified a set of candidate genes, including phosphatidylserine decarboxylase (PISD), which catalyzes the formation of phosphatidylethanolamine at the mitochondrial membrane. Taken together, these results indicate that PGAM5 suppresses mitochondrial energy expenditure by down-regulating UCP1 expression in brown adipocytes and that its phosphatase activity and intramembrane cleavage are required for UCP1 suppression.
Collapse
Affiliation(s)
- Sho Sugawara
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yusuke Kanamaru
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shiori Sekine
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Lila Maekawa
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Akinori Takahashi
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - Tadashi Yamamoto
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan; Laboratory for Immunogenetics, Center for Integrative Medical Sciences, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa 230-0045, Japan
| | - Kengo Watanabe
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takao Fujisawa
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kazuki Hattori
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
33
|
ASK1 inhibits browning of white adipose tissue in obesity. Nat Commun 2020; 11:1642. [PMID: 32242025 PMCID: PMC7118089 DOI: 10.1038/s41467-020-15483-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 03/10/2020] [Indexed: 12/13/2022] Open
Abstract
Increasing energy expenditure via induction of adipose tissue browning has become an appealing strategy to treat obesity and associated metabolic complications. Herein, we identify adipocyte-expressed apoptosis signal-regulating kinase 1 (ASK1) as regulator of adipose tissue browning. High fat diet-fed adipocyte-specific ASK1 knockout mice reveal increased UCP1 protein levels in inguinal adipose tissue concomitant with elevated energy expenditure, reduced obesity and ameliorated glucose tolerance compared to control littermates. In addition, ASK1-depletion blunts LPS-mediated downregulation of isoproterenol-induced UCP1 in subcutaneous fat both in vitro and in vivo. Conversely, adipocyte-specific ASK1 overexpression in chow-fed mice attenuates cold-induced UCP1 protein levels in inguinal fat. Mechanistically, ASK1 phosphorylates interferon regulatory factor 3 (IRF3) resulting in reduced Ucp1 expression. Taken together, our studies unravel a role of ASK1 in mediating the inhibitory effect of caloric surplus or LPS-treatment on adipose tissue browning. Adipocyte ASK1 might be a pharmacological target to combat obesity and associated morbidities.
Collapse
|
34
|
ASK1 inhibition: a therapeutic strategy with multi-system benefits. J Mol Med (Berl) 2020; 98:335-348. [PMID: 32060587 PMCID: PMC7080683 DOI: 10.1007/s00109-020-01878-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/18/2019] [Accepted: 01/15/2020] [Indexed: 12/11/2022]
Abstract
p38 mitogen-activated protein kinases (P38α and β) and c-Jun N-terminal kinases (JNK1, 2, and 3) are key mediators of the cellular stress response. However, prolonged P38 and JNK signalling is associated with damaging inflammatory responses, reactive oxygen species-induced cell death, and fibrosis in multiple tissues, such as the kidney, liver, central nervous system, and cardiopulmonary systems. These responses are associated with many human diseases, including arthritis, dementia, and multiple organ dysfunctions. Attempts to prevent P38- and JNK-mediated disease using small molecule inhibitors of P38 or JNK have generally been unsuccessful. However, apoptosis signal-regulating kinase 1 (ASK1), an upstream regulator of P38 and JNK, has emerged as an alternative drug target for limiting P38- and JNK-mediated disease. Within this review, we compile the evidence that ASK1 mediates damaging cellular responses via prolonged P38 or JNK activation. We discuss the potential benefits of ASK1 inhibition as a therapeutic and summarise the studies that have tested the effects of ASK1 inhibition in cell and animal disease models, in addition to human clinical trials for a variety of disorders.
Collapse
|
35
|
Uchiyama S. Fluorescent Sensors Based on a Novel Functional Design: Combination of an Environment-sensitive Fluorophore with Polymeric and Self-assembled Architectures. J SYN ORG CHEM JPN 2019. [DOI: 10.5059/yukigoseikyokaishi.77.1116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Seiichi Uchiyama
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| |
Collapse
|
36
|
Zhang D, Liu Y, Cui Y, Cui S. Mitogen-activated protein kinase kinase kinase 8 (MAP3K8) mediates the LH-induced stimulation of progesterone synthesis in the porcine corpus luteum. Reprod Fertil Dev 2019; 31:1444-1456. [PMID: 31039922 DOI: 10.1071/rd18478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 02/25/2019] [Indexed: 11/23/2022] Open
Abstract
Progesterone (P4) synthesized by the corpus luteum (CL) plays a key role in the establishment and maintenance of pregnancy. The LH signal is important for luteinisation and P4 synthesis in pigs. In a previous study, we demonstrated that mitogen-activated protein kinase kinase kinase 8 (MAP3K8) regulates P4 synthesis in mouse CL, but whether the function and mechanism of MAP3K8 in the pig is similar to that in the mouse is not known. Thus, in the present study we investigated the effects of MAP3K8 on porcine CL. Abundant expression of MAP3K8 was detected in porcine CL, and, in pigs, MAP3K8 expression was higher in mature CLs (or those of the mid-luteal phase) than in regressing CLs (late luteal phase). Further functional studies in cultured porcine luteal cells showed that P4 synthesis and the expression of genes encoding the key enzymes in P4 synthesis are significantly reduced when MAP3K8 is inhibited with the MAP3K8 inhibitor Tpl2 kinase inhibitor (MAP3K8i, 10μM). After 12-24h treatment of luteal cells with 100ngmL-1 LH, MAP3K8 expression and P4 secretion were significantly upregulated. In addition, the 10μM MAP3K8 inhibitor blocked the stimulatory effect of LH on P4 synthesis and extracellular signal-regulated kinase (ERK) 1/2 phosphorylation in porcine luteal cells. The LH-induced increases in MAP3K8 phosphorylation and expression, ERK1/2 phosphorylation and P4 synthesis were all blocked when protein kinase A was inhibited by its inhibitor H89 (20 μM) in porcine luteal cells. In conclusion, MAP3K8 mediates the LH-induced stimulation of P4 synthesis through the PKA/mitogen-activated protein kinase signalling pathway in porcine CL.
Collapse
Affiliation(s)
- Di Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100094, PR China
| | - Ying Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100094, PR China
| | - Yan Cui
- The 306th Hospital of People's Liberation Army, Beijing, 100101, PR China; and Corresponding authors. Emails: ;
| | - Sheng Cui
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100094, PR China; and Corresponding authors. Emails: ;
| |
Collapse
|
37
|
Temperature imaging using a cationic linear fluorescent polymeric thermometer and fluorescence lifetime imaging microscopy. Nat Protoc 2019; 14:1293-1321. [DOI: 10.1038/s41596-019-0145-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 01/25/2019] [Indexed: 12/31/2022]
|
38
|
Gao Y, Cao Y, Cui X, Wang X, Zhou Y, Huang F, Wang X, Wen J, Xie K, Xu P, Guo X, You L, Ji C. miR-199a-3p regulates brown adipocyte differentiation through mTOR signaling pathway. Mol Cell Endocrinol 2018; 476:155-164. [PMID: 29753771 DOI: 10.1016/j.mce.2018.05.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/23/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
Recent discoveries of functional brown adipocytes in mammals illuminates their therapeutic potential for combating obesity and its associated diseases. However, on account of the limited amount and activity in adult humans of brown adipocyte depots, identification of miRNAs and characterization their regulatory roles in human brown adipogenesis are urgently needed. This study focused on the role of microRNA (miR)-199a-3p in human brown adipocyte differentiation and thermogenic capacity. A decreased expression pattern of miR-199a-3p was consistently observed during the differentiation course of brown adipocytes in mice and humans. Conversely, its level was induced during the differentiation course of human white pre-adipocytes derived from visceral fat. miR-199a-3p expression was relatively abundant in interscapular BAT (iBAT) and differentially regulated in the activated and aging BAT in mice. Additionally, miR-199a-3p expression level in human brown adipocytes was observed decreased upon thermogenic activation and increased by aging-related stimuli. Using primary pre-adipocytes, miR-199a-3p over-expression was capable of attenuating lipid accumulation and adipogenic gene expression as well as impairing brown adipocytes' metabolic characteristics as revealed by decreased mitochondrial DNA content and respiration. Suppression of miR-199a-3p by a locked nucleic acid (LNA) modified-anti-miR led to increased differentiation and thermogenesis in human brown adipocytes. By combining target prediction and examination, we identified mechanistic target of rapamycin kinase (mTOR) as a direct target of miR-199a-3p that affected brown adipogenesis and thermogenesis. Our results point to a novel role for miR-199a-3p and its downstream effector mTOR in human brown adipocyte differentiation and maintenance of thermogenic characteristics, which can be manipulated as therapeutic targets against obesity and its related metabolic disorders.
Collapse
Affiliation(s)
- Yao Gao
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Yan Cao
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Xianwei Cui
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Xingyun Wang
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Yahui Zhou
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Fangyan Huang
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Xing Wang
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Juan Wen
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Kaipeng Xie
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Pengfei Xu
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Xirong Guo
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Lianghui You
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China.
| | - Chenbo Ji
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China.
| |
Collapse
|
39
|
β-TrCP-dependent degradation of ASK1 suppresses the induction of the apoptotic response by oxidative stress. Biochim Biophys Acta Gen Subj 2018; 1862:2271-2280. [DOI: 10.1016/j.bbagen.2018.07.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 12/26/2022]
|
40
|
Zhang S, Cao H, Li Y, Jing Y, Liu S, Ye C, Wang H, Yu S, Peng C, Hui L, Wang YC, Zhang H, Guo F, Zhai Q, Wang H, Huang R, Zhang L, Jiang J, Liu W, Ying H. Metabolic benefits of inhibition of p38α in white adipose tissue in obesity. PLoS Biol 2018; 16:e2004225. [PMID: 29750781 PMCID: PMC5965899 DOI: 10.1371/journal.pbio.2004225] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 05/23/2018] [Accepted: 04/09/2018] [Indexed: 01/22/2023] Open
Abstract
p38 has long been known as a central mediator of protein kinase A (PKA) signaling in brown adipocytes, which positively regulate the transcription of uncoupling protein 1 (UCP-1). However, the physiological role of p38 in adipose tissues, especially the white adipose tissue (WAT), is largely unknown. Here, we show that mice lacking p38α in adipose tissues display a lean phenotype, improved metabolism, and resistance to diet-induced obesity. Surprisingly, ablation of p38α causes minimal effects on brown adipose tissue (BAT) in adult mice, as evident from undetectable changes in UCP-1 expression, mitochondrial function, body temperature (BT), and energy expenditure. In contrast, genetic ablation of p38α in adipose tissues not only markedly facilitates the browning in WAT upon cold stress but also prevents diet-induced obesity. Consistently, pharmaceutical inhibition of p38α remarkably enhances the browning of WAT and has metabolic benefits. Furthermore, our data suggest that p38α deficiency promotes white-to-beige adipocyte reprogramming in a cell-autonomous manner. Mechanistically, inhibition of p38α stimulates the UCP-1 transcription through PKA and its downstream cAMP-response element binding protein (CREB), which form a positive feedback loop that functions to reinforce the white-to-beige phenotypic switch during cold exposure. Together, our study reveals that inhibition of p38α is able to promote WAT browning and confer metabolic benefits. Our study also indicates that p38α in WAT represents an exciting pharmacological target to combat obesity and metabolic diseases. The functional brown adipose tissue (BAT) identified in human adults consists of not only classic brown adipocytes but also brown-like adipocytes (beige adipocytes), both of which are important for energy homeostasis. Due to the same ability to convert fat into heat as brown adipocytes, beige adipocytes have been considered as a novel pharmacological target to combat obesity. Growing evidence suggests that promoting the development and formation of beige adipocytes in white adipose tissue (WAT), also called the browning of WAT, is able to prevent diet-induced obesity and improve metabolism in rodents. Thus, understanding the molecular basis for the regulation of browning in WAT may help us to develop new strategies to counteract obesity and metabolic diseases. In this study, adipocyte-specific p38α knockout (Fp38αKO) mice are generated that display a lean phenotype, improved metabolism, and resistance to diet-induced obesity. Interestingly, we found that adipocyte p38α deficiency facilitates the browning in WAT. Then, we show that pharmaceutical inhibition of p38α enhances the browning in WAT and has metabolic benefits. We propose that inhibiting p38α in WAT, possibly combined with cold exposure, could constitute an exciting pharmacological target to combat obesity and metabolic diseases.
Collapse
Affiliation(s)
- Shengjie Zhang
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hongchao Cao
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Li
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yanyan Jing
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shengnan Liu
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Cheng Ye
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hui Wang
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shuxian Yu
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chengyuan Peng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Lijian Hui
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu-cheng Wang
- Shanghai Xuhui Central Hospital, Shanghai Clinical Center, Chinese Academy of Sciences, Shanghai, China
| | - Haibing Zhang
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Feifan Guo
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiwei Zhai
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hui Wang
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
| | - Ruimin Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ling Zhang
- Department of Head and Neck Surgery, Fudan University Cancer Center and Department of Oncology, Fudan University, Shanghai Medical College, Shanghai, China
| | - Jingjing Jiang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Liu
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hao Ying
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
- * E-mail:
| |
Collapse
|
41
|
Okabe K, Sakaguchi R, Shi B, Kiyonaka S. Intracellular thermometry with fluorescent sensors for thermal biology. Pflugers Arch 2018; 470:717-731. [PMID: 29397424 PMCID: PMC5942359 DOI: 10.1007/s00424-018-2113-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/16/2018] [Accepted: 01/17/2018] [Indexed: 12/27/2022]
Abstract
Temperature influences the activities of living organisms at various levels. Cells not only detect environmental temperature changes through their unique temperature-sensitive molecular machineries but also muster an appropriate response to the temperature change to maintain their inherent functions. Despite the fundamental involvement of temperature in physiological phenomena, the mechanism by which cells produce and use heat is largely unknown. Recently, fluorescent thermosensors that function as thermometers in live cells have attracted much attention in biology. These new tools, made of various temperature-sensitive molecules, have allowed for intracellular thermometry at the single-cell level. Intriguing spatiotemporal temperature variations, including organelle-specific thermogenesis, have been revealed with these fluorescent thermosensors, which suggest an intrinsic connection between temperature and cell functions. Moreover, fluorescent thermosensors have shown that intracellular temperature changes at the microscopic level are largely different from those assumed for a water environment at the macroscopic level. Thus, the employment of fluorescent thermosensors will uncover novel mechanisms of intracellular temperature-assisted physiological functions.
Collapse
Affiliation(s)
- Kohki Okabe
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan.
- JST, PRESTO, 4-8-1 Honcho, Kawaguchi, Saitama, 332-0012, Japan.
| | - Reiko Sakaguchi
- World Premier International Research Initiative-Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Beini Shi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Shigeki Kiyonaka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan.
| |
Collapse
|
42
|
Smad proteins differentially regulate obesity-induced glucose and lipid abnormalities and inflammation via class-specific control of AMPK-related kinase MPK38/MELK activity. Cell Death Dis 2018; 9:471. [PMID: 29700281 PMCID: PMC5920110 DOI: 10.1038/s41419-018-0489-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 03/15/2018] [Accepted: 03/19/2018] [Indexed: 12/16/2022]
Abstract
Smad proteins have been implicated in metabolic processes, but little is known about how they regulate metabolism. Because Smad 2, 3, 4, and 7 have previously been shown to interact with murine protein serine-threonine kinase 38 (MPK38), an AMP-activated protein kinase (AMPK)-related kinase that has been implicated in obesity-associated metabolic defects, we investigated whether Smad proteins regulate metabolic processes via MPK38. Smads2/3/4 increased, but Smad7 decreased, MPK38-mediated apoptosis signal-regulating kinase-1 (ASK1)/transforming growth factor-β (TGF-β)/p53 signaling. However, MPK38-mediated phosphorylation-defective Smad mutants (Smad2 S245A, Smad3 S204A, Smad4 S343A, and Smad7 T96A) had no such effect. In addition, Smads2/3/4 increased, but Smad7 decreased, the stability of MPK38. Consistent with this, Smads2/3/4 attenuated complex formation between MPK38 and its negative regulator thioredoxin (Trx), whereas Smad7 increased this complex formation. However, an opposite effect was observed on complex formation between MPK38 and its positive regulator zinc-finger-like protein 9 (ZPR9). When Smads were overexpressed in high-fat diet (HFD)-fed obese mice using an adenoviral delivery system, Smads2/3/4 improved, but Smad7 worsened, obesity-associated metabolic parameters and inflammation in a MPK38 phosphorylation-dependent manner. These findings suggest that Smad proteins have class-specific impacts on obesity-associated metabolism by differentially regulating MPK38 activity in diet-induced obese mice.
Collapse
|
43
|
Uchiyama S, Tsuji T, Kawamoto K, Okano K, Fukatsu E, Noro T, Ikado K, Yamada S, Shibata Y, Hayashi T, Inada N, Kato M, Koizumi H, Tokuyama H. A Cell‐Targeted Non‐Cytotoxic Fluorescent Nanogel Thermometer Created with an Imidazolium‐Containing Cationic Radical Initiator. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201801495] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Seiichi Uchiyama
- Graduate School of Pharmaceutical Sciences The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
| | - Toshikazu Tsuji
- Central Laboratories for Key Technologies KIRIN Company Limited 1-13-5 Fukuura Kanazawa-ku Yokohama Kanagawa 236-0004 Japan
| | - Kyoko Kawamoto
- Graduate School of Pharmaceutical Sciences The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
| | - Kentaro Okano
- Graduate School of Pharmaceutical Sciences Tohoku University Sendai Miyagi 980-8578 Japan
- Department of Chemical Science and Engineering Kobe University Kobe Hyogo 657-8501 Japan
| | - Eiko Fukatsu
- Graduate School of Pharmaceutical Sciences The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
| | - Takahiro Noro
- Graduate School of Pharmaceutical Sciences Tohoku University Sendai Miyagi 980-8578 Japan
| | - Kumiko Ikado
- Central Laboratories for Key Technologies KIRIN Company Limited 1-13-5 Fukuura Kanazawa-ku Yokohama Kanagawa 236-0004 Japan
| | - Sayuri Yamada
- Central Laboratories for Key Technologies KIRIN Company Limited 1-13-5 Fukuura Kanazawa-ku Yokohama Kanagawa 236-0004 Japan
| | - Yuka Shibata
- Graduate School of Pharmaceutical Sciences The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
| | - Teruyuki Hayashi
- The Graduate School of Biological Sciences Nara Institute of Science and Technology Ikoma Nara 630-0192 Japan
- Department of Nutrition Koshien University 10-1 Momijigaoka Takarazuka Hyogo 665-0006 Japan
| | - Noriko Inada
- The Graduate School of Biological Sciences Nara Institute of Science and Technology Ikoma Nara 630-0192 Japan
| | - Masaru Kato
- Graduate School of Pharmaceutical Sciences The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
| | - Hideki Koizumi
- Central Laboratories for Key Technologies KIRIN Company Limited 1-13-5 Fukuura Kanazawa-ku Yokohama Kanagawa 236-0004 Japan
| | - Hidetoshi Tokuyama
- Graduate School of Pharmaceutical Sciences Tohoku University Sendai Miyagi 980-8578 Japan
| |
Collapse
|
44
|
Uchiyama S, Tsuji T, Kawamoto K, Okano K, Fukatsu E, Noro T, Ikado K, Yamada S, Shibata Y, Hayashi T, Inada N, Kato M, Koizumi H, Tokuyama H. A Cell‐Targeted Non‐Cytotoxic Fluorescent Nanogel Thermometer Created with an Imidazolium‐Containing Cationic Radical Initiator. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/anie.201801495] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Seiichi Uchiyama
- Graduate School of Pharmaceutical Sciences The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
| | - Toshikazu Tsuji
- Central Laboratories for Key Technologies KIRIN Company Limited 1-13-5 Fukuura Kanazawa-ku Yokohama Kanagawa 236-0004 Japan
| | - Kyoko Kawamoto
- Graduate School of Pharmaceutical Sciences The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
| | - Kentaro Okano
- Graduate School of Pharmaceutical Sciences Tohoku University Sendai Miyagi 980-8578 Japan
- Department of Chemical Science and Engineering Kobe University Kobe Hyogo 657-8501 Japan
| | - Eiko Fukatsu
- Graduate School of Pharmaceutical Sciences The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
| | - Takahiro Noro
- Graduate School of Pharmaceutical Sciences Tohoku University Sendai Miyagi 980-8578 Japan
| | - Kumiko Ikado
- Central Laboratories for Key Technologies KIRIN Company Limited 1-13-5 Fukuura Kanazawa-ku Yokohama Kanagawa 236-0004 Japan
| | - Sayuri Yamada
- Central Laboratories for Key Technologies KIRIN Company Limited 1-13-5 Fukuura Kanazawa-ku Yokohama Kanagawa 236-0004 Japan
| | - Yuka Shibata
- Graduate School of Pharmaceutical Sciences The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
| | - Teruyuki Hayashi
- The Graduate School of Biological Sciences Nara Institute of Science and Technology Ikoma Nara 630-0192 Japan
- Department of Nutrition Koshien University 10-1 Momijigaoka Takarazuka Hyogo 665-0006 Japan
| | - Noriko Inada
- The Graduate School of Biological Sciences Nara Institute of Science and Technology Ikoma Nara 630-0192 Japan
| | - Masaru Kato
- Graduate School of Pharmaceutical Sciences The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
| | - Hideki Koizumi
- Central Laboratories for Key Technologies KIRIN Company Limited 1-13-5 Fukuura Kanazawa-ku Yokohama Kanagawa 236-0004 Japan
| | - Hidetoshi Tokuyama
- Graduate School of Pharmaceutical Sciences Tohoku University Sendai Miyagi 980-8578 Japan
| |
Collapse
|
45
|
Markussen LK, Winther S, Wicksteed B, Hansen JB. GSK3 is a negative regulator of the thermogenic program in brown adipocytes. Sci Rep 2018; 8:3469. [PMID: 29472592 PMCID: PMC5823915 DOI: 10.1038/s41598-018-21795-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 02/09/2018] [Indexed: 01/23/2023] Open
Abstract
Brown adipose tissue is a promising therapeutic target in metabolic disorders due to its ability to dissipate energy and improve systemic insulin sensitivity and glucose homeostasis. β-Adrenergic stimulation of brown adipocytes leads to an increase in oxygen consumption and induction of a thermogenic gene program that includes uncoupling protein 1 (Ucp1) and fibroblast growth factor 21 (Fgf21). In kinase inhibitor screens, we have identified glycogen synthase kinase 3 (GSK3) as a negative regulator of basal and β-adrenergically stimulated Fgf21 expression in cultured brown adipocytes. In addition, inhibition of GSK3 also caused increased Ucp1 expression and oxygen consumption. β-Adrenergic stimulation triggered an inhibitory phosphorylation of GSK3 in a protein kinase A (PKA)-dependent manner. Mechanistically, inhibition of GSK3 activated the mitogen activated protein kinase (MAPK) kinase 3/6-p38 MAPK-activating transcription factor 2 signaling module. In summary, our data describe GSK3 as a novel negative regulator of β-adrenergic signaling in brown adipocytes.
Collapse
Affiliation(s)
- Lasse K Markussen
- Department of Biology, University of Copenhagen, DK-2100, Copenhagen, Denmark
| | - Sally Winther
- Department of Biology, University of Copenhagen, DK-2100, Copenhagen, Denmark
| | - Barton Wicksteed
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jacob B Hansen
- Department of Biology, University of Copenhagen, DK-2100, Copenhagen, Denmark.
| |
Collapse
|
46
|
Hayashi M, Futawaka K, Koyama R, Fan Y, Matsushita M, Hirao A, Fukuda Y, Nushida A, Nezu S, Tagami T, Moriyama K. Effects of growth hormone on uncoupling protein 1 in white adipose tissues in obese mice. Growth Horm IGF Res 2017; 37:31-39. [PMID: 29111497 DOI: 10.1016/j.ghir.2017.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 10/17/2017] [Accepted: 10/22/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The transition of white adipocytes to beige cells (a phenomenon referred to as browning or beigeing) during obesity has been previously reported. Our study aimed to examine the mechanisms through which obesity induced by a high fat diet (HFD) affects uncoupling protein 1 (UCP1) expression via signal transduction and activator of transcription 5 (STAT5s). DESIGN Seven-week-old male C57BL/6J mice were fed a normal or HFD for 11weeks. Body weight, white adipose tissue weight, and blood lipid and glucose levels were measured. To unveil the molecular mechanisms of UCP1 expression in adipose tissue, we performed further studying 3T3-L1 cells using qRT-PCR. We also measured UCP1 promoter activity in the TSA201 cell line using a dual luciferase assay. In addition, we analyzed the predicted consensus sequences for STAT5 binding in the UCP1 promoter region. RESULTS Mice fed an HFD had higher body weight and intra-abdominal adipose tissues weight and a higher expression of UCP1, GH receptor (GHR), STATs, suppressors of cytokine signaling (SOCSs), and cytokine-inducible SH2-containing protein (CISH) compared to control mice. In 3T3-L1 cell studies, GH induced phosphorylation of the STAT5, SOCSs, CISH and UCP1 expressions. UCP1 promoter activity was associated with constitutively active STAT5 in a dose-dependent manner. We confirmed functional STAT5 binding sites at -425, -279, and -178bp of the UCP1 promoter. CONCLUSION We suggest that endogenous GH induces UCP1 expression in adipose tissue via STAT5.
Collapse
Affiliation(s)
- Misa Hayashi
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Kumi Futawaka
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Rie Koyama
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Yue Fan
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Midori Matsushita
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Asuka Hirao
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Yuki Fukuda
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Ayako Nushida
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Syoko Nezu
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Tetsuya Tagami
- Clinical Research Institute for Endocrine and Metabolic Diseases, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan
| | - Kenji Moriyama
- Department of Medicine & Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan; Clinical Research Institute for Endocrine and Metabolic Diseases, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan.
| |
Collapse
|
47
|
The thermogenic actions of natriuretic peptide in brown adipocytes: The direct measurement of the intracellular temperature using a fluorescent thermoprobe. Sci Rep 2017; 7:12978. [PMID: 29021616 PMCID: PMC5636787 DOI: 10.1038/s41598-017-13563-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/26/2017] [Indexed: 12/11/2022] Open
Abstract
In addition to the various effects of natriuretic peptides (NPs) on cardiovascular systems, increasing attention is being paid to the possibility that NPs induce adipose tissue browning and activate thermogenic program. We herein established a direct intracellular temperature measurement system using a fluorescent thermoprobe and investigated the thermogenic effects of A-type NP (ANP) on brown adipocytes. The thermoprobe was successfully introduced into rat brown adipocytes, and the temperature dependent change in fluorescence intensity ratio was measured using a fluorescence microscope. After one-hour incubation with ANP, the degree of the change in fluorescence intensity ratio was significantly higher in ANP-treated (P < 0.01) adipocytes compared to untreated controls. The ANP treatment increased uncoupling protein-1 (UCP1) mRNA levels, which is one of the markers of thermogenesis in adipocytes, while the intracellular ATP content was not changed, indicating mitochondrial uncoupled respiration. Intriguingly, these thermogenic actions of ANP were more prominent when brown adipocytes were incubated at 35 °C than at 37 °C. Moreover, the increase in the intracellular temperature and the expression of UCP1 induced by ANP were cancelled by p38MAPK inhibition. Taken together, this study directly demonstrated the thermogenic actions of ANP in brown adipocytes through the use of a novel method of intracellular temperature measurement.
Collapse
|
48
|
Difference in intracellular temperature rise between matured and precursor brown adipocytes in response to uncoupler and β-adrenergic agonist stimuli. Sci Rep 2017; 7:12889. [PMID: 29018208 PMCID: PMC5635136 DOI: 10.1038/s41598-017-12634-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/13/2017] [Indexed: 01/27/2023] Open
Abstract
Brown adipocytes function to maintain body temperature by heat production. However, direct measurement of heat production at a single cell level remains difficult. Here we developed a method to measure the temperature within primary cultured brown adipocytes using a cationic fluorescent polymeric thermometer. Placement of the thermometer within a matured brown adipocyte and a precursor cell enabled the detection of heat production following uncoupler treatment. The increase in the intracellular temperature due to stimulation with a mitochondrial uncoupler was higher in matured brown adipocytes than in precursor cells. Stimulation with a β-adrenergic receptor (β-AR) agonist, norepinephrine, raised the intracellular temperature of matured brown adipocytes to a level comparable to that observed after stimulation with a β3-AR-specific agonist, CL316.243. In contrast, neither β-AR agonist induced an intracellular temperature increase in precursor cells. Further, pretreatment of brown adipocytes with a β3-AR antagonist inhibited the norepinephrine-stimulated elevation of temperature. These results demonstrate that our novel method successfully determined the difference in intracellular temperature increase between matured brown adipocytes and precursor cells in response to stimulation by an uncoupler and β-AR agonists.
Collapse
|
49
|
Hattori K, Ishikawa H, Sakauchi C, Takayanagi S, Naguro I, Ichijo H. Cold stress-induced ferroptosis involves the ASK1-p38 pathway. EMBO Rep 2017; 18:2067-2078. [PMID: 28887319 DOI: 10.15252/embr.201744228] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/19/2017] [Accepted: 07/31/2017] [Indexed: 01/22/2023] Open
Abstract
A wide variety of cell death mechanisms, such as ferroptosis, have been proposed in mammalian cells, and the classification of cell death attracts global attention because each type of cell death has the potential to play causative roles in specific diseases. However, the precise molecular mechanisms leading to cell death are poorly understood, particularly in ferroptosis. Here, we show that continuous severe cold stress induces ferroptosis and the ASK1-p38 MAPK pathway in multiple cell lines. The activation of the ASK1-p38 pathway is mediated by critical determinants of ferroptosis: MEK activity, iron ions, and lipid peroxide. The chemical compound erastin, a potent ferroptosis inducer, also activates the ASK1-p38 axis downstream of lipid peroxide accumulation and leads to ASK1-dependent cell death in a cell type-specific manner. These lines of evidence provide mechanistic insight into ferroptosis, a type of regulated necrosis.
Collapse
Affiliation(s)
- Kazuki Hattori
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Ishikawa
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Chihiro Sakauchi
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Saki Takayanagi
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Isao Naguro
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hidenori Ichijo
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
50
|
Singh SP, Grant I, Meissner A, Kappas A, Abraham NG. Ablation of adipose-HO-1 expression increases white fat over beige fat through inhibition of mitochondrial fusion and of PGC1α in female mice. Horm Mol Biol Clin Investig 2017; 31:hmbci-2017-0027. [PMID: 28763300 DOI: 10.1515/hmbci-2017-0027] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/23/2017] [Indexed: 12/15/2022]
Abstract
Background Hmox1 plays an important role in the regulation of mitochondrial bioenergetics and function by regulating cellular heme-derived CO and bilirubin. Previous studies have demonstrated that global disruption of HO-1 in humans and mice resulted in severe organ dysfunction. Methods We investigated the potential role of adipose-specific-HO-1 genetic ablation on adipose tissue function, mitochondrial quality control and energy expenditure by generating an adipo-HO-1 knockout mouse model (Adipo-HO-1-/-) and, in vitro, adipocyte cells in which HO activity was inhibited. Adiposity, signaling proteins, fasting glucose and oxygen consumption were determined and compared to adipocyte cultures with depressed levels of both HO-1/HO-2. Results Adipo-HO-1-/- female mice exhibited increased adipocyte size, and decreases in the mitochondrial fusion to fission ratio, PGC1, and SIRT3. Importantly, ablation of HO-1 in adipose tissue resulted in fat acquiring many properties of visceral fat such as decreases in thermogenic genes including pAMPK and PRDM16. Deletion of HO-1 in mouse adipose tissue led to complete metabolic dysfunction, an increase in white adipose tissue, a reduction of beige fat and associated increases in FAS, aP2 and hyperglycemia. Mechanistically, genetic deletion of HO-1 in adipose tissues decreased the mitochondrial fusion to fission ratio; disrupted the activity of the PGC1 transcriptional axis and thermogenic genes both in vitro and in vivo. Conclusion Ablation of adipose tissue-HO-1 abridged PGC1 expression promoted mitochondrial dysfunction and contributed to an increase of pro-inflammatory visceral fat and abrogated beige-cell like phenotype.
Collapse
Affiliation(s)
| | - Ilana Grant
- Department of Medicine, New York Medical College, NY, USA
| | - Aliza Meissner
- Department of Medicine, New York Medical College, NY, USA
| | - Attallah Kappas
- The Rockefeller University, New York, NY 10065, USA, Phone: 212-327-8494, Fax: 212-327-8690
| | - Nader G Abraham
- Department of Pharmacology, New York Medical College, NY, USA
- New York Medical College, Valhalla, NY 10595, USA, Phone: +914-594-3121, Fax: +914-347-4956
| |
Collapse
|