1
|
Miro C, Cicatiello AG, Nappi A, Sagliocchi S, Acampora L, Restolfer F, Cuomo O, de Alteris G, Pugliese G, Torabinejad S, Maritato R, Murolo M, Di Cicco E, Velotti N, Capuano M, La Civita E, Terracciano D, Ciampaglia R, Stornaiuolo M, Musella M, Aprea G, Pignataro G, Savastano S, Dentice M. Leptin enhances the intracellular thyroid hormone activation in skeletal muscle to boost energy balance. Cell Metab 2025; 37:936-953.e7. [PMID: 39986272 DOI: 10.1016/j.cmet.2025.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 10/15/2024] [Accepted: 01/27/2025] [Indexed: 02/24/2025]
Abstract
Thyroid hormones (THs) are key modulators of energy metabolism and cross-talk with other endocrine and metabolic factors. Notably, leptin can increase hypothalamic control of TH synthesis as an adaptive metabolic response regulating body weight. In this study, we found that the TH signal is heightened in overweight humans and is lost with obesity. In mice, systemic and intracerebroventricular leptin injection induces the expression of type 2 deiodinase (D2), the TH-activating enzyme, in skeletal muscle. Mechanistically, leptin enhances the transcription of D2 by a STAT3- and α-melanocyte-stimulating hormone (α-MSH)/cyclic AMP (cAMP)-dependent regulation. Notably, mice lacking D2 or with a mutation in the TH receptor do not exhibit the metabolic effects of leptin, such as increased insulin sensitivity and oxygen consumption, indicating that leptin's peripheral metabolic effects in skeletal muscle are mediated by TH. These findings underscore the critical role of leptin in integrating the TH-induced metabolic activation, while also contributing to appetite suppression in response to perceived fat stores.
Collapse
Affiliation(s)
- Caterina Miro
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | | | - Annarita Nappi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Serena Sagliocchi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Lucia Acampora
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Federica Restolfer
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Ornella Cuomo
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II," 80131 Naples, Italy
| | - Giulia de Alteris
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Gabriella Pugliese
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Sepehr Torabinejad
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Rosa Maritato
- Department of Translational Medical Sciences, University of Naples "Federico II," 80131 Naples, Italy
| | - Melania Murolo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Emery Di Cicco
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Nunzio Velotti
- Department of Advanced Biomedical Sciences, University of Naples "Federico II," 80131 Naples, Italy
| | - Marianna Capuano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Evelina La Civita
- Department of Translational Medical Sciences, University of Naples "Federico II," 80131 Naples, Italy
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University of Naples "Federico II," 80131 Naples, Italy
| | - Roberto Ciampaglia
- Department of Pharmacy, University of Naples "Federico II," 80149 Naples, Italy
| | - Mariano Stornaiuolo
- Department of Pharmacy, University of Naples "Federico II," 80149 Naples, Italy
| | - Mario Musella
- Department of Advanced Biomedical Sciences, University of Naples "Federico II," 80131 Naples, Italy
| | - Giovanni Aprea
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II," 80131 Naples, Italy
| | - Silvia Savastano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," 80131 Naples, Italy; CEINGE - Biotecnologie Avanzate S.c.a.r.l., 80131 Naples, Italy.
| |
Collapse
|
2
|
Enders JD, Prodoehl EK, Penn SM, Sriram A, Stucky CL. Episodic pain in Fabry disease is mediated by a heat shock protein-TRPA1 axis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.20.639340. [PMID: 40060522 PMCID: PMC11888165 DOI: 10.1101/2025.02.20.639340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Two-thirds of patients with Fabry disease suffer debilitating pain attacks triggered by exercise, fever, and exposure to environmental heat. These patients face an even greater risk of heat-related episodic pain in the face of global climate change. Almost nothing is known about the biological mechanisms underlying heat-induced pain crises in Fabry disease, and there is no preclinical model available to study Fabry crises. Here, we established the first model of heat-induced pain attacks in Fabry disease by exposing transgenic Fabry rats to environmental heat. Heat exposure precipitated robust mechanical hypersensitivity, closely matching temporal features reported by patients with Fabry disease. At the cellular level, heat exposure sensitized Fabry dorsal root ganglia (DRG) neurons to agonists for transient receptor potential cation channel A1 (TRPA1), but not TRPV1. The heat shock response, which normally confers heat-resilience, was impaired in Fabry disease, and we demonstrated that heat shock proteins (HSP70 and HSP90) regulate TRPA1. Strikingly, pharmacologically inhibiting HSP90 completely prevented cellular and behavioral sensitization by environmental heat in Fabry disease. Together, this work establishes the first model of episodic pain in Fabry disease, implicates the heat shock response in heat-evoked pain episodes, and identifies a novel heat shock protein-TRPA1 regulatory axis.
Collapse
Affiliation(s)
- Jonathan D Enders
- Department of Cell Biology, Neurobiology, and Anatomy; Medical College of Wisconsin, Milwaukee, WI
| | - Eve K Prodoehl
- Department of Cell Biology, Neurobiology, and Anatomy; Medical College of Wisconsin, Milwaukee, WI
| | - Signe M Penn
- Department of Cell Biology, Neurobiology, and Anatomy; Medical College of Wisconsin, Milwaukee, WI
| | - Anvitha Sriram
- Department of Cell Biology, Neurobiology, and Anatomy; Medical College of Wisconsin, Milwaukee, WI
| | - Cheryl L Stucky
- Department of Cell Biology, Neurobiology, and Anatomy; Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
3
|
Qian W, Zhang X, Yuan D, Wu Y, Li H, Wei L, Li Z, Dai Z, Song P, Sun Q, Zhou Z, Xia Q, Cheng D. USP8 and Hsp70 regulate endoreplication by synergistically promoting Fzr deubiquitination and stabilization. SCIENCE ADVANCES 2025; 11:eadq9111. [PMID: 40106570 PMCID: PMC11922063 DOI: 10.1126/sciadv.adq9111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 02/11/2025] [Indexed: 03/22/2025]
Abstract
Endoreplication is characterized by multiple rounds of DNA replication without cell division and determines the growth and final size of endoreplicating cells and tissues in eukaryotes. The cyclic ubiquitination and degradation of several cell cycle regulators are required for endoreplication progression. However, the deubiquitinase that deubiquitinates and stabilizes key factors to modulate endoreplication remains unknown. Here, we found in the endoreplicating Drosophila salivary gland and Bombyx silk gland that the depletion of ubiquitin-specific peptidase 8 (USP8) led to endoreplication arrest and a decrease in gland size. Mechanistically, we showed that USP8 interacted with the Fizzy-related (Fzr) protein, a conserved master regulator of endoreplication, thereby deubiquitinating and stabilizing Fzr to modulate endoreplication. Moreover, the molecular chaperone heat shock protein 70 (Hsp70) mediated proper folding of Fzr and increased the interaction between Fzr and USP8, thereby promoting the deubiquitination and stabilization of Fzr. Together, our study demonstrates that USP8 and Hsp70 regulate endoreplication by synergistically maintaining Fzr stability though deubiquitination.
Collapse
Affiliation(s)
- Wenliang Qian
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Xing Zhang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Dongqin Yuan
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Yuting Wu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Hao Li
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Ling Wei
- School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Zheng Li
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Zongcai Dai
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Pei Song
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Qiaoling Sun
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Zizhang Zhou
- College of Life Sciences, Shandong Agricultural University, Tai’an 271018, China
| | - Qingyou Xia
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Daojun Cheng
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400715, China
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| |
Collapse
|
4
|
Huang S, Piao C, Zhao Z, Beuschel CB, Turrel O, Toppe D, Sigrist SJ. Enhanced memory despite severe sleep loss in Drosophila insomniac mutants. PLoS Biol 2025; 23:e3003076. [PMID: 40111981 DOI: 10.1371/journal.pbio.3003076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Sleep is crucial for cognitive functions and life span across species. While sleep homeostasis and cognitive processes are linked through cellular and synaptic plasticity, the signaling pathways connecting them remain unclear. Here, we show that Drosophila insomniac (inc) short sleep mutants, which lack an adaptor protein for the autism-associated Cullin-3 ubiquitin ligase, exhibited enhanced Pavlovian aversive olfactory learning and memory, unlike other sleep mutants with normal or reduced memory. Through a genetic modifier screen, we found that a mild reduction of Protein Kinase A (PKA) signaling specifically rescued the sleep and longevity phenotypes of inc mutants. However, this reduction further increased their excessive memory and mushroom body overgrowth. Since inc mutants displayed higher PKA signaling, we propose that inc loss-of-function suppresses sleep via increased PKA activity, which also constrains the excessive memory of inc mutants. Our data identify a signaling cascade for balancing sleep and memory functions, and provide a plausible explanation for the sleep phenotypes of inc mutants, suggesting that memory hyperfunction can provoke sleep deficits.
Collapse
Affiliation(s)
- Sheng Huang
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Chengji Piao
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Zhiying Zhao
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Christine B Beuschel
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Oriane Turrel
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - David Toppe
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| |
Collapse
|
5
|
Hao X, Hu Z, Li M, Zhang S, Tang M, Hao C, Qi S, Liang Y, Almeida MF, Smith K, Zuo C, Feng Y, Guo M, Ma D, Li S, Wang Z, Sun Y, Deng Z, Mao C, Xia Z, Jiang Y, Gao Y, Xu Y, Schisler JC, Shi C. E3 ubiquitin ligase CHIP facilitates cAMP and cGMP signalling cross-talk by polyubiquitinating PDE9A. EMBO J 2025; 44:1249-1273. [PMID: 39806097 PMCID: PMC11833080 DOI: 10.1038/s44318-024-00351-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025] Open
Abstract
The carboxyl terminus of Hsc70-interacting protein (CHIP) is pivotal for managing misfolded and aggregated proteins via chaperone networks and degradation pathways. In a preclinical rodent model of CHIP-related ataxia, we observed that CHIP mutations lead to increased levels of phosphodiesterase 9A (PDE9A), whose role in this context remains poorly understood. Here, we investigated the molecular mechanisms underlying the role of PDE9A in CHIP-related ataxia and demonstrated that CHIP binds to PDE9A, facilitating its polyubiquitination and autophagic degradation. Conversely, dysfunctional CHIP disrupts this process, resulting in PDE9A accumulation, increased cGMP hydrolysis, and impaired PKG phosphorylation of CHIP at serine 19. This cascade further amplifies PDE9A accumulation, ultimately disrupting mitophagy and triggering neuronal apoptosis. Elevated PKA levels inhibit PDE9A degradation, further exacerbating this neuronal dysfunction. Notably, pharmacological inhibition of PDE9A via Bay 73-6691 or virus-mediated CHIP expression restored the balance of cGMP/cAMP signalling. These interventions protect against cerebellar neuropathologies, particularly Purkinje neuron mitophagy dysfunction. Thus, PDE9A upregulation considerably exacerbates ataxia associated with CHIP mutations, and targeting the interaction between PDE9A and CHIP is an innovative therapeutic strategy for CHIP-related ataxia.
Collapse
Affiliation(s)
- Xiaoyan Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Zhengwei Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Mengjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Shuo Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Mibo Tang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Chenwei Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Shasha Qi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yuanyuan Liang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Michael F Almeida
- McAllister Heart Institute and the Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Kaitlan Smith
- McAllister Heart Institute and the Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chunyan Zuo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yanmei Feng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Mengnan Guo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Dongrui Ma
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Shuangjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Zhiyun Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yuemeng Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Zhifen Deng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Chengyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Zongping Xia
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yong Jiang
- State Key Laboratory of Antiviral Drugs, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450000, China
- Henan Key Laboratory of Critical Care Medicine, Department of Emergency Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450000, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, 450000, China
| | - Yanxia Gao
- State Key Laboratory of Antiviral Drugs, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450000, China
- Henan Key Laboratory of Critical Care Medicine, Department of Emergency Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450000, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, 450000, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Medical Key Laboratory of Poisoning Diseases of Henan Province, Zhengzhou, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Jonathan C Schisler
- McAllister Heart Institute and the Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
6
|
Liu Y, Liu J, Hu N, Li Z, Liu A, Luo R, Du S, Guo D, Li J, Duan J. Classical prescription Daqinjiao decoction inhibit cerebral ischemia/reperfusion induced necroptosis and ferroptosis through multiple mechanisms. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119300. [PMID: 39736347 DOI: 10.1016/j.jep.2024.119300] [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: 09/26/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Daqinjiao decoction (DQJT), a classical prescription, has been utilized for millennia in stroke management, yet its underlying mechanisms remained obscure. AIM OF THE STUDY The aim of this study was to elucidate the mechanisms through which DQJT mitigates cerebral ischemia/reperfusion injury (CI/RI). MATERIALS AND METHODS The quantification of DQJT's primary components were performed by HPLC. Pharmacological assessments were then conducted to ascertain DQJT's efficacy in a Middle Cerebral Artery Occlusion/Reperfusion (MCAO/R) model. Following this, untargeted metabolomics, lipidomics and network pharmacology analyses were undertaken to unveil potential mechanisms, which were subsequently validated. UPLC-Q-TOF/MS was utilized to detect DQJT-derived chemicals in brain tissue, and molecular docking techniques were employed to investigate the bioactive compounds. RESULTS DQJT treatment reduced brain damage induced by MCAO/R, as evidenced by decreased infarct sizes, enhanced behavioral function scores, and diminished neuronal damages. Untargeted metabolomics and lipidomics revealed that DQJT improved metabolism of unsaturated fatty acids. According to network pharmacology, lipid metabolism, cAMP signaling pathway and toll-like receptor signaling pathway pathways were notably affected, with HSP90AA1, TLR4, and PKA identified as potential targets of DQJT. Immunofluorescence and Western blot analyses further demonstrated that DQJT counteracted necroptosis and ferroptosis by inhibiting the HSP90AA1 and TLR4 pathways and enhancing the PKA pathway. Molecular docking results supported that the possible pharmacodynamic substances of DQJT in protecting against CI/RI. CONCLUSION This research established that DQJT attenuates brain injury induced by MCAO/R through the modulation of necroptosis and ferroptosis via pathways including HSP90AA1, TLR4, and PKA. It shed light on the potential mechanisms and effective constituents of DQJT in stroke treatment, paving the way for further exploration of this ancient formula.
Collapse
Affiliation(s)
- Yuwen Liu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi, 710072, China
| | - Jiping Liu
- The Key Laboratory of Basic and New Drug Research of Traditional Chinese Medicine, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi, China
| | - Naping Hu
- General Hospital of Xinjiang Production and Construction Corps, Department of Pharmacy, 232 Qingnian Road, Tianshan District, Xinjiang Uygur Autonomous Region, Urumqi City, 830092, China
| | - Zhengrong Li
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi, 710072, China
| | - Anqi Liu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi, 710072, China
| | - Ruyue Luo
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi, 710072, China
| | - Siyu Du
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi, 710072, China
| | - Dongyan Guo
- The Key Laboratory of Basic and New Drug Research of Traditional Chinese Medicine, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi, China.
| | - Jiankang Li
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi, 710072, China.
| | - Jialin Duan
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi, 710072, China.
| |
Collapse
|
7
|
Laabs BH, Lohmann K, Vollstedt EJ, Reinberger T, Nuxoll LM, Kilic-Berkmen G, Perlmutter JS, Loens S, Cruchaga C, Franke A, Dobricic V, Hinrichs F, Grözinger A, Altenmüller E, Bellows S, Boesch S, Bressman SB, Duque KR, Espay AJ, Ferbert A, Feuerstein JS, Frank S, Gasser T, Haslinger B, Jech R, Kaiser F, Kamm C, Kollewe K, Kühn AA, LeDoux MS, Lohmann E, Mahajan A, Münchau A, Multhaupt-Buell T, Pantelyat A, Richardson SEP, Raymond D, Reich SG, Pullman RS, Schormair B, Sharma N, Sichani AH, Simonyan K, Volkmann J, Shukla AW, Winkelmann J, Wright LJ, Zech M, Zeuner KE, Zittel S, Kasten M, Sun YV, Bäumer T, Brüggemann N, Ozelius LJ, Jinnah HA, Klein C, König IR. Genetic Risk Factors in Isolated Dystonia Escape Genome-Wide Association Studies. Mov Disord 2024; 39:2110-2116. [PMID: 39287592 PMCID: PMC11975433 DOI: 10.1002/mds.29968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/19/2024] [Accepted: 07/22/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Despite considerable heritability, previous smaller genome-wide association studies (GWASs) have not identified any robust genetic risk factors for isolated dystonia. OBJECTIVE The objective of this study was to perform a large-scale GWAS in a well-characterized, multicenter sample of >6000 individuals to identify genetic risk factors for isolated dystonia. METHODS Array-based GWASs were performed on autosomes for 4303 dystonia participants and 2362 healthy control subjects of European ancestry with subgroup analysis based on age at onset, affected body regions, and a newly developed clinical score. Another 736 individuals were used for validation. RESULTS This GWAS identified no common genome-wide significant loci that could be replicated despite sufficient power to detect meaningful effects. Power analyses imply that the effects of individual variants are likely very small. CONCLUSIONS Moderate single-nucleotide polymorphism-based heritability indicates that common variants do not contribute to isolated dystonia in this cohort. Sequence-based GWASs (eg, by whole-genome sequencing) might help to better understand the genetic basis. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Björn-Hergen Laabs
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | | | | | - Lisa-Marie Nuxoll
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | | | - Joel S. Perlmutter
- Department of Neurology, Radiology and Neuroscience, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sebastian Loens
- Institute of Systems Motor Science, CBBM, University of Lübeck, Lübeck, Germany
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Valerija Dobricic
- Lübeck Interdisciplinary Platform for Genome Analysis, University of Lübeck, Lübeck, Germany
| | - Frauke Hinrichs
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Anne Grözinger
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Eckart Altenmüller
- Institute of Music Physiology and Musician’s Medicine, Hanover University of Music, Drama and Media, Hanover, Germany
| | - Steven Bellows
- Parkinson’s Disease Center and Movement Disorder Clinic, Baylor College of Medicine, Houston, Texas, USA
| | - Sylvia Boesch
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Susan B. Bressman
- Department of Neurology, Mount Sinai Beth Israel Medical Center, New York, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kevin R. Duque
- James J. and Joan A. Gardner Family Center for Parkinson’s Disease and Movement Disorders Neurology and Rehabilitation Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Alberto J. Espay
- James J. and Joan A. Gardner Family Center for Parkinson’s Disease and Movement Disorders Neurology and Rehabilitation Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Andreas Ferbert
- Department of Neurology, Kassel School of Medicine, Klinikum Kassel, Kassel, Germany
| | - Jeanne S. Feuerstein
- Department of Neurology, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Samuel Frank
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas Gasser
- Department of Neurology, University of Tübingen, Tübingen, Germany
- Hertie Institute for Clinical Brain Research and DZNE, University of Tübingen, Tübingen, Germany
| | - Bernhard Haslinger
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Robert Jech
- Department of Neurology, Charles University in Prague, 1st Faculty of Medicine and General University Hospital in Prague, Prague, Czech Republic
| | - Frank Kaiser
- Institute of Human Genetics, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Essener Zentrum für Seltene Erkrankungen, University Hospital Essen, Essen, Germany
| | - Christoph Kamm
- Department of Neurology, University Medical Centre Rostock, Rostock, Germany
| | - Katja Kollewe
- Clinic for Neurology, Hannover Medical School, Hannover, Germany
| | - Andrea A. Kühn
- Department of Neurology and Experimental Neurology, Charité–University Medicine, Berlin, Germany
| | - Mark S. LeDoux
- Veracity Neuroscience LLC, Memphis, Tennessee, USA
- Department of Psychology, University of Memphis, Memphis, Tennessee, USA
| | - Ebba Lohmann
- Department of Neurology, University of Tübingen, Tübingen, Germany
- Hertie Institute for Clinical Brain Research and DZNE, University of Tübingen, Tübingen, Germany
| | - Abhimanyu Mahajan
- Department of Neurological Sciences, RUSH University, Chicago, Illinois, USA
| | - Alexander Münchau
- Institute of Systems Motor Science, CBBM, University of Lübeck, Lübeck, Germany
| | - Trisha Multhaupt-Buell
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Deborah Raymond
- Department of Neurology, Mount Sinai Beth Israel Medical Center, New York, New York, USA
| | - Stephen G. Reich
- University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Rachel Saunders Pullman
- Department of Neurology, Mount Sinai Beth Israel Medical Center, New York, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Barbara Schormair
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Nutan Sharma
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Azadeh Hamzehei Sichani
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Otolaryngology–Head and Neck Surgery, Massachusetts Eye and Ear and Harvard Medical School, Boston, Massachusetts, USA
| | - Kristina Simonyan
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Neurological Sciences, RUSH University, Chicago, Illinois, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Otolaryngology–Head and Neck Surgery, Massachusetts Eye and Ear and Harvard Medical School, Boston, Massachusetts, USA
| | - Jens Volkmann
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | | | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology, SyNergy, Munich, Germany
| | - Laura J. Wright
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael Zech
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | | | - Simone Zittel
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Meike Kasten
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| | - Yan V. Sun
- Department of Epidemiology, Emory University Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Tobias Bäumer
- Institute of Systems Motor Science, CBBM, University of Lübeck, Lübeck, Germany
| | | | - Laurie J. Ozelius
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Hyder A. Jinnah
- Department of Neurology, Emory University, Atlanta, Georgia, USA
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Inke R. König
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
8
|
Zhou H, Patel V, Rice R, Lee R, Kim HW, Weintraub NL, Su H, Chen W. Neddylation and Its Target Cullin 3 Are Essential for Adipocyte Differentiation. Cells 2024; 13:1654. [PMID: 39404417 PMCID: PMC11475318 DOI: 10.3390/cells13191654] [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: 08/13/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
The ongoing obesity epidemic has raised awareness of the complex physiology of adipose tissue. Abnormal adipocyte differentiation results in the development of systemic metabolic disorders such as insulin resistance and diabetes. The conjugation of NEDD8 (neural precursor cell expressed, developmentally downregulated 8) to target protein, termed neddylation, has been shown to mediate adipogenesis. However, much remains unknown about its role in adipogenesis. Here, we demonstrated that neddylation and its targets, the cullin (CUL) family members, are differentially regulated during mouse and human adipogenesis. Inhibition of neddylation by MLN4924 significantly reduced adipogenesis of 3T3-L1 and human stromal vascular cells. Deletion of NAE1, a subunit of the only NEDD8 E1 enzyme, suppressed neddylation and impaired adipogenesis. Neddylation deficiency did not affect mitotic cell expansion. Instead, it disrupted CREB/CEBPβ/PPARγ signaling, essential for adipogenesis. Interestingly, among the neddylation-targeted CUL family members, deletion of CUL3, but not CUL1, CUL2, or CUL4A, largely replicated the adipogenic defects observed with neddylation deficiency. A PPARγ agonist minimally rescued the adipogenic defects caused by the deletion of NAE1 and CUL3. In conclusion, our study demonstrates that neddylation and its targeted CUL3 are crucial for adipogenesis. These findings provide potential targets for therapeutic intervention in obesity and metabolic disorders.
Collapse
Affiliation(s)
- Hongyi Zhou
- Departments of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Vijay Patel
- Department of Surgery, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (V.P.); (R.R.); (R.L.)
| | - Robert Rice
- Department of Surgery, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (V.P.); (R.R.); (R.L.)
| | - Richard Lee
- Department of Surgery, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (V.P.); (R.R.); (R.L.)
| | - Ha Won Kim
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (H.W.K.); (N.L.W.)
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Neal L. Weintraub
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (H.W.K.); (N.L.W.)
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Huabo Su
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Weiqin Chen
- Departments of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
9
|
Feichtner A, Enzler F, Kugler V, Hoppe K, Mair S, Kremser L, Lindner H, Huber RG, Stelzl U, Stefan E, Torres-Quesada O. Phosphorylation of the compartmentalized PKA substrate TAF15 regulates RNA-protein interactions. Cell Mol Life Sci 2024; 81:162. [PMID: 38568213 PMCID: PMC10991009 DOI: 10.1007/s00018-024-05204-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 03/08/2024] [Accepted: 03/09/2024] [Indexed: 04/05/2024]
Abstract
Spatiotemporal-controlled second messengers alter molecular interactions of central signaling nodes for ensuring physiological signal transmission. One prototypical second messenger molecule which modulates kinase signal transmission is the cyclic-adenosine monophosphate (cAMP). The main proteinogenic cellular effectors of cAMP are compartmentalized protein kinase A (PKA) complexes. Their cell-type specific compositions precisely coordinate substrate phosphorylation and proper signal propagation which is indispensable for numerous cell-type specific functions. Here we present evidence that TAF15, which is implicated in the etiology of amyotrophic lateral sclerosis, represents a novel nuclear PKA substrate. In cross-linking and immunoprecipitation experiments (iCLIP) we showed that TAF15 phosphorylation alters the binding to target transcripts related to mRNA maturation, splicing and protein-binding related functions. TAF15 appears to be one of multiple PKA substrates that undergo RNA-binding dynamics upon phosphorylation. We observed that the activation of the cAMP-PKA signaling axis caused a change in the composition of a collection of RNA species that interact with TAF15. This observation appears to be a broader principle in the regulation of molecular interactions, as we identified a significant enrichment of RNA-binding proteins within endogenous PKA complexes. We assume that phosphorylation of RNA-binding domains adds another layer of regulation to binary protein-RNAs interactions with consequences to RNA features including binding specificities, localization, abundance and composition.
Collapse
Affiliation(s)
- Andreas Feichtner
- Tyrolean Cancer Research Institute (TKFI), Innrain 66, 6020, Innsbruck, Austria
- Institute of Molecular Biology and Center for Molecular Biosciences, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria
| | - Florian Enzler
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innrain 66/66a, 6020, Innsbruck, Austria
| | - Valentina Kugler
- Tyrolean Cancer Research Institute (TKFI), Innrain 66, 6020, Innsbruck, Austria
- Institute of Molecular Biology and Center for Molecular Biosciences, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria
| | - Katharina Hoppe
- Institute of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innrain 80/82, 6020, Innsbruck, Austria
| | - Sophia Mair
- Department of Cardiac Surgery, Medical University of Innsbruck, Innrain 66/66a, 6020, Innsbruck, Austria
- Vascage, Center of Clinical Stroke Research, 6020, Innsbruck, Austria
| | - Leopold Kremser
- Division of Clinical Biochemistry, Biocenter, Medical University of Innsbruck, Innrain 80/82, 6020, Innsbruck, Austria
| | - Herbert Lindner
- Division of Clinical Biochemistry, Biocenter, Medical University of Innsbruck, Innrain 80/82, 6020, Innsbruck, Austria
| | - Roland G Huber
- Bioinformatics Institute, Agency for Science Technology and Research, Singapore, 138671, Singapore
| | - Ulrich Stelzl
- Institute of Pharmaceutical Sciences, University of Graz, Schubertstrasse 1, 8010, Graz, Austria
| | - Eduard Stefan
- Tyrolean Cancer Research Institute (TKFI), Innrain 66, 6020, Innsbruck, Austria.
- Institute of Molecular Biology and Center for Molecular Biosciences, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria.
| | - Omar Torres-Quesada
- Tyrolean Cancer Research Institute (TKFI), Innrain 66, 6020, Innsbruck, Austria.
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Innrain 80/82, 6020, Innsbruck, Austria.
| |
Collapse
|
10
|
Lv M, Mu J, Xing Y, Zhou X, Ge J, Gong D, Geng T, Zhao M. Glucose inhibits the inflammatory response in goose fatty liver by increasing the ubiquitination level of PKA. J Anim Sci 2024; 102:skae239. [PMID: 39158360 PMCID: PMC11375046 DOI: 10.1093/jas/skae239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024] Open
Abstract
Protein kinase A (PKA) plays an important role in cellular life activities. Recently, PKA was found to bind to the inhibitor of nuclear factor-kappaB (IκB), a key protein in the nuclear factor-kappaB (NF-κB) pathway, to form a complex involved in the regulation of inflammatory response. However, the role of PKA in the anti-inflammatory of goose fatty liver is still unclear. A total of 14 healthy 70-d-old male Lander geese were randomly divided into a control group and an overfeeding group. Inflammation level was analyzed by histopathological method in the liver. The mRNA and protein abundance of PKA and tumor necrosis factor-alpha (TNFα), as well as the ubiquitination level of PKA, were detected. Moreover, goose primary hepatocytes were cotreated with glucose, harringtonine, and carbobenzoxy-l-leucyl-l-leucyl-l-leucinal (MG132). Finally, the co-immunoprecipitated samples of PKA from the control and overfeeding group were used for protein mass spectrometry. The results showed that no difference in PKA mRNA expression was observed (P > 0.05), while the PKA protein level in the overfed group was significantly reduced (P < 0.05) when compared with the control group. The ubiquitination level of PKA was higher than that of the control group in fatty liver. The mRNA expression of PKA was elevated but protein abundance was reduced in goose primary hepatocytes with 200 mmol/L glucose treatment (P < 0.05). The PKA protein abundance was dramatically reduced in hepatocytes treated with harringtonine (P < 0.01) when compared with the glucose-supplemented group. Nevertheless, MG132 tended to alleviate the inhibitory effect of harringtonine on PKA protein abundance (P = 0.081). There was no significant difference in TNFα protein level among glucose-treated groups and control (P > 0.05). Protein mass spectrometry analysis showed that 29 and 76 interacting proteins of PKA were screened in goose normal and fatty liver, respectively. Validation showed that PKA interacted with the E3 ubiquitination ligases ring finger protein 135 (RNF135) and potassium channel modulatory factor 1 (KCMF1). In summary, glucose may inhibit the inflammatory response in goose fatty liver by increasing the ubiquitination level of PKA. Additionally, RNF135 and KCMF1 may be involved in the regulation of PKA ubiquitination level as E3 ubiquitination ligases.
Collapse
Affiliation(s)
- Mengqing Lv
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
| | - Ji'an Mu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
| | - Ya Xing
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
| | - Xiaoyi Zhou
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
| | - Jing Ge
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
| | - Daoqing Gong
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
| | - Tuoyu Geng
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
| | - Minmeng Zhao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
| |
Collapse
|
11
|
Yin P, Jian X, Liu Y, Liu Y, Lv L, Cui H, Zhang L. Elucidating cellular interactome of chikungunya virus identifies host dependency factors. Virol Sin 2023; 38:497-507. [PMID: 37182691 PMCID: PMC10436055 DOI: 10.1016/j.virs.2023.05.007] [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: 11/29/2022] [Accepted: 05/11/2023] [Indexed: 05/16/2023] Open
Abstract
Chikungunya virus (CHIKV) is a re-emerging mosquito-transmitted RNA virus causing joint and muscle pain. To better understand how CHIKV rewires the host cell and usurps host cell functions, we generated a systematic CHIKV-human protein-protein interaction map and revealed several novel connections that will inform further mechanistic studies. One of these novel interactions, between the viral protein E1 and STIP1 homology and U-box containing protein 1 (STUB1), was found to mediate ubiquitination of E1 and degrade E1 through the proteasome. Capsid associated with G3BP1, G3BP2 and AAA+ ATPase valosin-containing protein (VCP). Furthermore, VCP inhibitors blocked CHIKV infection, suggesting VCP could serve as a therapeutic target. Further work is required to fully understand the functional consequences of these interactions. Given that CHIKV proteins are conserved across alphaviruses, many virus-host protein-protein interactions identified in this study might also exist in other alphaviruses. Construction of interactome of CHIKV provides the basis for further studying the function of alphavirus biology.
Collapse
Affiliation(s)
- Peiqi Yin
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250013, China; NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100176, China
| | - Xia Jian
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100176, China
| | - Yihan Liu
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Yuwen Liu
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Lu Lv
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Haoran Cui
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Leiliang Zhang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250013, China; Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
12
|
Liu D, Bai J, Chen Q, Tan R, An Z, Xiao J, Qu Y, Xu Y. Brain metastases: It takes two factors for a primary cancer to metastasize to brain. Front Oncol 2022; 12:1003715. [PMID: 36248975 PMCID: PMC9554149 DOI: 10.3389/fonc.2022.1003715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Brain metastasis of a cancer is a malignant disease with high mortality, but the cause and the molecular mechanism remain largely unknown. Using the samples of primary tumors of 22 cancer types in the TCGA database, we have performed a computational study of their transcriptomic data to investigate the drivers of brain metastases at the basic physics and chemistry level. Our main discoveries are: (i) the physical characteristics, namely electric charge, molecular weight, and the hydrophobicity of the extracellular structures of the expressed transmembrane proteins largely affect a primary cancer cell’s ability to cross the blood-brain barrier; and (ii) brain metastasis may require specific functions provided by the activated enzymes in the metastasizing primary cancer cells for survival in the brain micro-environment. Both predictions are supported by published experimental studies. Based on these findings, we have built a classifier to predict if a given primary cancer may have brain metastasis, achieving the accuracy level at AUC = 0.92 on large test sets.
Collapse
Affiliation(s)
- Dingyun Liu
- Center for Cancer Systems Biology, China-Japan Union Hospital of Jilin University, Changchun, China
- College of Computer Science and Technology, Jilin University, Changchun, China
| | - Jun Bai
- Center for Cancer Systems Biology, China-Japan Union Hospital of Jilin University, Changchun, China
- School of Artificial Intelligence, Jilin University, Changchun, China
| | - Qian Chen
- Center for Cancer Systems Biology, China-Japan Union Hospital of Jilin University, Changchun, China
- College of Computer Science and Technology, Jilin University, Changchun, China
| | - Renbo Tan
- Center for Cancer Systems Biology, China-Japan Union Hospital of Jilin University, Changchun, China
- College of Computer Science and Technology, Jilin University, Changchun, China
| | - Zheng An
- Center for Cancer Systems Biology, China-Japan Union Hospital of Jilin University, Changchun, China
- Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, The University of Georgia, Athens, GA, United States
| | - Jun Xiao
- Center for Cancer Systems Biology, China-Japan Union Hospital of Jilin University, Changchun, China
- College of Computer Science and Technology, Jilin University, Changchun, China
| | - Yingwei Qu
- Center for Cancer Systems Biology, China-Japan Union Hospital of Jilin University, Changchun, China
- College of Computer Science and Technology, Jilin University, Changchun, China
| | - Ying Xu
- Center for Cancer Systems Biology, China-Japan Union Hospital of Jilin University, Changchun, China
- Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, The University of Georgia, Athens, GA, United States
- *Correspondence: Ying Xu,
| |
Collapse
|
13
|
Omar MH, Byrne DP, Jones KN, Lakey TM, Collins KB, Lee KS, Daly LA, Forbush KA, Lau HT, Golkowski M, McKnight GS, Breault DT, Lefrançois-Martinez AM, Martinez A, Eyers CE, Baird GS, Ong SE, Smith FD, Eyers PA, Scott JD. Mislocalization of protein kinase A drives pathology in Cushing's syndrome. Cell Rep 2022; 40:111073. [PMID: 35830806 PMCID: PMC9311266 DOI: 10.1016/j.celrep.2022.111073] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/20/2022] [Accepted: 06/17/2022] [Indexed: 01/15/2023] Open
Abstract
Mutations in the catalytic subunit of protein kinase A (PKAc) drive the stress hormone disorder adrenal Cushing's syndrome. We define mechanisms of action for the PKAc-L205R and W196R variants. Proximity proteomic techniques demonstrate that both Cushing's mutants are excluded from A kinase-anchoring protein (AKAP)-signaling islands, whereas live-cell photoactivation microscopy reveals that these kinase mutants indiscriminately diffuse throughout the cell. Only cAMP analog drugs that displace native PKAc from AKAPs enhance cortisol release. Rescue experiments that incorporate PKAc mutants into AKAP complexes abolish cortisol overproduction, indicating that kinase anchoring restores normal endocrine function. Analyses of adrenal-specific PKAc-W196R knockin mice and Cushing's syndrome patient tissue reveal defective signaling mechanisms of the disease. Surprisingly each Cushing's mutant engages a different mitogenic-signaling pathway, with upregulation of YAP/TAZ by PKAc-L205R and ERK kinase activation by PKAc-W196R. Thus, aberrant spatiotemporal regulation of each Cushing's variant promotes the transmission of distinct downstream pathogenic signals.
Collapse
Affiliation(s)
- Mitchell H Omar
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA.
| | - Dominic P Byrne
- Department of Biochemistry & Systems Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Kiana N Jones
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Tyler M Lakey
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Kerrie B Collins
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Kyung-Soon Lee
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Leonard A Daly
- Centre for Proteome Research, Department of Biochemistry and Systems Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Katherine A Forbush
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Ho-Tak Lau
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Martin Golkowski
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - G Stanley McKnight
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Anne-Marie Lefrançois-Martinez
- Génétique, Reproduction et Développement (GReD), CNRS, INSERM, Université Clermont Auvergne, 63001 Clermont-Ferrand, France
| | - Antoine Martinez
- Génétique, Reproduction et Développement (GReD), CNRS, INSERM, Université Clermont Auvergne, 63001 Clermont-Ferrand, France
| | - Claire E Eyers
- Centre for Proteome Research, Department of Biochemistry and Systems Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Geoffrey S Baird
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Shao-En Ong
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - F Donelson Smith
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Patrick A Eyers
- Department of Biochemistry & Systems Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - John D Scott
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
14
|
Senatore E, Iannucci R, Chiuso F, Delle Donne R, Rinaldi L, Feliciello A. Pathophysiology of Primary Cilia: Signaling and Proteostasis Regulation. Front Cell Dev Biol 2022; 10:833086. [PMID: 35646931 PMCID: PMC9130585 DOI: 10.3389/fcell.2022.833086] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/21/2022] [Indexed: 01/29/2023] Open
Abstract
Primary cilia are microtubule-based, non-motile sensory organelles present in most types of growth-arrested eukaryotic cells. They are transduction hubs that receive and transmit external signals to the cells in order to control growth, differentiation and development. Mutations of genes involved in the formation, maintenance or disassembly of ciliary structures cause a wide array of developmental genetic disorders, also known as ciliopathies. The primary cilium is formed during G1 in the cell cycle and disassembles at the G2/M transition. Following the completion of the cell division, the cilium reassembles in G1. This cycle is finely regulated at multiple levels. The ubiquitin-proteasome system (UPS) and the autophagy machinery, two main protein degradative systems in cells, play a fundamental role in cilium dynamics. Evidence indicate that UPS, autophagy and signaling pathways may act in synergy to control the ciliary homeostasis. However, the mechanisms involved and the links between these regulatory systems and cilium biogenesis, dynamics and signaling are not well defined yet. Here, we discuss the reciprocal regulation of signaling pathways and proteolytic machineries in the control of the assembly and disassembly of the primary cilium, and the impact of the derangement of these regulatory networks in human ciliopathies.
Collapse
|
15
|
Carroll EC, Marqusee S. Site-specific ubiquitination: Deconstructing the degradation tag. Curr Opin Struct Biol 2022; 73:102345. [PMID: 35247748 DOI: 10.1016/j.sbi.2022.102345] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/28/2021] [Accepted: 01/17/2022] [Indexed: 02/04/2023]
Abstract
Ubiquitin is a small eukaryotic protein so named for its cellular abundance and originally recognized for its role as the posttranslational modification (PTM) "tag" condemning substrates to degradation by the 26S proteasome. Since its discovery in the 1970s, protein ubiquitination has also been identified as a key regulatory feature in dozens of non-degradative cellular processes. This myriad of roles illustrates the versatility of ubiquitin as a PTM; however, understanding the cellular and molecular factors that enable discrimination between degradative versus non-degradative ubiquitination events has been a persistent challenge. Here, we discuss recent advances in uncovering how site-specificity - the exact residue that gets modified - modulates distinct protein fates and cellular outcomes with an emphasis on how ubiquitination site specificity regulates proteasomal degradation. We explore recent advances in structural biology, biophysics, and cell biology that have enabled a broader understanding of the role of ubiquitination in altering the dynamics of the target protein, including implications for the design of targeted protein degradation therapeutics.
Collapse
Affiliation(s)
- Emma C Carroll
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, 94038, USA.
| | - Susan Marqusee
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA; QB3 Institute for Quantitative Biosciences, University of California Berkeley, Berkeley, CA, 94720, USA; Department of Chemistry, University of California Berkeley, Berkeley, CA, 94720, USA.
| |
Collapse
|
16
|
Ramms DJ, Raimondi F, Arang N, Herberg FW, Taylor SS, Gutkind JS. G αs-Protein Kinase A (PKA) Pathway Signalopathies: The Emerging Genetic Landscape and Therapeutic Potential of Human Diseases Driven by Aberrant G αs-PKA Signaling. Pharmacol Rev 2021; 73:155-197. [PMID: 34663687 PMCID: PMC11060502 DOI: 10.1124/pharmrev.120.000269] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Many of the fundamental concepts of signal transduction and kinase activity are attributed to the discovery and crystallization of cAMP-dependent protein kinase, or protein kinase A. PKA is one of the best-studied kinases in human biology, with emphasis in biochemistry and biophysics, all the way to metabolism, hormone action, and gene expression regulation. It is surprising, however, that our understanding of PKA's role in disease is largely underappreciated. Although genetic mutations in the PKA holoenzyme are known to cause diseases such as Carney complex, Cushing syndrome, and acrodysostosis, the story largely stops there. With the recent explosion of genomic medicine, we can finally appreciate the broader role of the Gαs-PKA pathway in disease, with contributions from aberrant functioning G proteins and G protein-coupled receptors, as well as multiple alterations in other pathway components and negative regulators. Together, these represent a broad family of diseases we term the Gαs-PKA pathway signalopathies. The Gαs-PKA pathway signalopathies encompass diseases caused by germline, postzygotic, and somatic mutations in the Gαs-PKA pathway, with largely endocrine and neoplastic phenotypes. Here, we present a signaling-centric review of Gαs-PKA-driven pathophysiology and integrate computational and structural analysis to identify mutational themes commonly exploited by the Gαs-PKA pathway signalopathies. Major mutational themes include hotspot activating mutations in Gαs, encoded by GNAS, and mutations that destabilize the PKA holoenzyme. With this review, we hope to incite further study and ultimately the development of new therapeutic strategies in the treatment of a wide range of human diseases. SIGNIFICANCE STATEMENT: Little recognition is given to the causative role of Gαs-PKA pathway dysregulation in disease, with effects ranging from infectious disease, endocrine syndromes, and many cancers, yet these disparate diseases can all be understood by common genetic themes and biochemical signaling connections. By highlighting these common pathogenic mechanisms and bridging multiple disciplines, important progress can be made toward therapeutic advances in treating Gαs-PKA pathway-driven disease.
Collapse
Affiliation(s)
- Dana J Ramms
- Department of Pharmacology (D.J.R., N.A., J.S.G.), Department of Chemistry and Biochemistry (S.S.T.), and Moores Cancer Center (D.J.R., N.A., J.S.G.), University of California, San Diego, La Jolla, California; Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Pisa, Italy (F.R.); and Department of Biochemistry, University of Kassel, Kassel, Germany (F.W.H.)
| | - Francesco Raimondi
- Department of Pharmacology (D.J.R., N.A., J.S.G.), Department of Chemistry and Biochemistry (S.S.T.), and Moores Cancer Center (D.J.R., N.A., J.S.G.), University of California, San Diego, La Jolla, California; Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Pisa, Italy (F.R.); and Department of Biochemistry, University of Kassel, Kassel, Germany (F.W.H.)
| | - Nadia Arang
- Department of Pharmacology (D.J.R., N.A., J.S.G.), Department of Chemistry and Biochemistry (S.S.T.), and Moores Cancer Center (D.J.R., N.A., J.S.G.), University of California, San Diego, La Jolla, California; Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Pisa, Italy (F.R.); and Department of Biochemistry, University of Kassel, Kassel, Germany (F.W.H.)
| | - Friedrich W Herberg
- Department of Pharmacology (D.J.R., N.A., J.S.G.), Department of Chemistry and Biochemistry (S.S.T.), and Moores Cancer Center (D.J.R., N.A., J.S.G.), University of California, San Diego, La Jolla, California; Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Pisa, Italy (F.R.); and Department of Biochemistry, University of Kassel, Kassel, Germany (F.W.H.)
| | - Susan S Taylor
- Department of Pharmacology (D.J.R., N.A., J.S.G.), Department of Chemistry and Biochemistry (S.S.T.), and Moores Cancer Center (D.J.R., N.A., J.S.G.), University of California, San Diego, La Jolla, California; Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Pisa, Italy (F.R.); and Department of Biochemistry, University of Kassel, Kassel, Germany (F.W.H.)
| | - J Silvio Gutkind
- Department of Pharmacology (D.J.R., N.A., J.S.G.), Department of Chemistry and Biochemistry (S.S.T.), and Moores Cancer Center (D.J.R., N.A., J.S.G.), University of California, San Diego, La Jolla, California; Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Pisa, Italy (F.R.); and Department of Biochemistry, University of Kassel, Kassel, Germany (F.W.H.)
| |
Collapse
|
17
|
Xu R, Guo Y, Peng S, Liu J, Li P, Jia W, Zhao J. Molecular Targets and Biological Functions of cAMP Signaling in Arabidopsis. Biomolecules 2021; 11:biom11050688. [PMID: 34063698 PMCID: PMC8147800 DOI: 10.3390/biom11050688] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 01/11/2023] Open
Abstract
Cyclic AMP (cAMP) is a pivotal signaling molecule existing in almost all living organisms. However, the mechanism of cAMP signaling in plants remains very poorly understood. Here, we employ the engineered activity of soluble adenylate cyclase to induce cellular cAMP elevation in Arabidopsis thaliana plants and identify 427 cAMP-responsive genes (CRGs) through RNA-seq analysis. Induction of cellular cAMP elevation inhibits seed germination, disturbs phytohormone contents, promotes leaf senescence, impairs ethylene response, and compromises salt stress tolerance and pathogen resistance. A set of 62 transcription factors are among the CRGs, supporting a prominent role of cAMP in transcriptional regulation. The CRGs are significantly overrepresented in the pathways of plant hormone signal transduction, MAPK signaling, and diterpenoid biosynthesis, but they are also implicated in lipid, sugar, K+, nitrate signaling, and beyond. Our results provide a basic framework of cAMP signaling for the community to explore. The regulatory roles of cAMP signaling in plant plasticity are discussed.
Collapse
Affiliation(s)
- Ruqiang Xu
- School of Agricultural Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.G.); (S.P.); (J.L.); (P.L.); (W.J.); (J.Z.)
- Zhengzhou Research Base, State Key Laboratory of Cotton Biology, Zhengzhou University, Zhengzhou 450001, China
- Correspondence: ; Tel.: +86-0371-6778-5095
| | - Yanhui Guo
- School of Agricultural Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.G.); (S.P.); (J.L.); (P.L.); (W.J.); (J.Z.)
| | - Song Peng
- School of Agricultural Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.G.); (S.P.); (J.L.); (P.L.); (W.J.); (J.Z.)
| | - Jinrui Liu
- School of Agricultural Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.G.); (S.P.); (J.L.); (P.L.); (W.J.); (J.Z.)
| | - Panyu Li
- School of Agricultural Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.G.); (S.P.); (J.L.); (P.L.); (W.J.); (J.Z.)
| | - Wenjing Jia
- School of Agricultural Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.G.); (S.P.); (J.L.); (P.L.); (W.J.); (J.Z.)
| | - Junheng Zhao
- School of Agricultural Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.G.); (S.P.); (J.L.); (P.L.); (W.J.); (J.Z.)
| |
Collapse
|
18
|
Senatore E, Chiuso F, Rinaldi L, Intartaglia D, Delle Donne R, Pedone E, Catalanotti B, Pirone L, Fiorillo B, Moraca F, Giamundo G, Scala G, Raffeiner A, Torres-Quesada O, Stefan E, Kwiatkowski M, van Pijkeren A, Morleo M, Franco B, Garbi C, Conte I, Feliciello A. The TBC1D31/praja2 complex controls primary ciliogenesis through PKA-directed OFD1 ubiquitylation. EMBO J 2021; 40:e106503. [PMID: 33934390 PMCID: PMC8126939 DOI: 10.15252/embj.2020106503] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/03/2021] [Accepted: 02/11/2021] [Indexed: 12/16/2022] Open
Abstract
The primary cilium is a microtubule‐based sensory organelle that dynamically links signalling pathways to cell differentiation, growth, and development. Genetic defects of primary cilia are responsible for genetic disorders known as ciliopathies. Orofacial digital type I syndrome (OFDI) is an X‐linked congenital ciliopathy caused by mutations in the OFD1 gene and characterized by malformations of the face, oral cavity, digits and, in the majority of cases, polycystic kidney disease. OFD1 plays a key role in cilium biogenesis. However, the impact of signalling pathways and the role of the ubiquitin‐proteasome system (UPS) in the control of OFD1 stability remain unknown. Here, we identify a novel complex assembled at centrosomes by TBC1D31, including the E3 ubiquitin ligase praja2, protein kinase A (PKA), and OFD1. We show that TBC1D31 is essential for ciliogenesis. Mechanistically, upon G‐protein‐coupled receptor (GPCR)‐cAMP stimulation, PKA phosphorylates OFD1 at ser735, thus promoting OFD1 proteolysis through the praja2‐UPS circuitry. This pathway is essential for ciliogenesis. In addition, a non‐phosphorylatable OFD1 mutant dramatically affects cilium morphology and dynamics. Consistent with a role of the TBC1D31/praja2/OFD1 axis in ciliogenesis, alteration of this molecular network impairs ciliogenesis in vivo in Medaka fish, resulting in developmental defects. Our findings reveal a multifunctional transduction unit at the centrosome that links GPCR signalling to ubiquitylation and proteolysis of the ciliopathy protein OFD1, with important implications on cilium biology and development. Derangement of this control mechanism may underpin human genetic disorders.
Collapse
Affiliation(s)
- Emanuela Senatore
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| | - Francesco Chiuso
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| | - Laura Rinaldi
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| | | | - Rossella Delle Donne
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| | - Emilia Pedone
- Institute of Biostructures and Bioimaging, CNR, Naples, Italy
| | | | - Luciano Pirone
- Institute of Biostructures and Bioimaging, CNR, Naples, Italy
| | - Bianca Fiorillo
- Department of Pharmacy, University Federico II, Naples, Italy
| | - Federica Moraca
- Department of Pharmacy, University Federico II, Naples, Italy.,Net4Science srl, University "Magna Graecia", Catanzaro, Italy
| | | | - Giovanni Scala
- Department of Biology, University Federico II, Naples, Italy
| | - Andrea Raffeiner
- Institute of Biochemistry, University of Innsbruck, Innsbruck, Austria.,Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Omar Torres-Quesada
- Institute of Biochemistry, University of Innsbruck, Innsbruck, Austria.,Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Eduard Stefan
- Institute of Biochemistry, University of Innsbruck, Innsbruck, Austria.,Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| | | | | | - Manuela Morleo
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Brunella Franco
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Translational Medical Science, University Federico II, Naples, Italy
| | - Corrado Garbi
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| | - Ivan Conte
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Biology, University Federico II, Naples, Italy
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| |
Collapse
|
19
|
Jiang Y, Hao N. Memorizing environmental signals through feedback and feedforward loops. Curr Opin Cell Biol 2021; 69:96-102. [PMID: 33549848 PMCID: PMC8058236 DOI: 10.1016/j.ceb.2020.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022]
Abstract
Cells in diverse organisms can store the information of previous environmental conditions for long periods of time. This form of cellular memory adjusts the cell's responses to future challenges, providing fitness advantages in fluctuating environments. Many biological functions, including cellular memory, are mediated by specific recurring patterns of interactions among proteins and genes, known as 'network motifs.' In this review, we focus on three well-characterized network motifs - negative feedback loops, positive feedback loops, and feedforward loops, which underlie different types of cellular memories. We describe the latest studies identifying these motifs in various molecular processes and discuss how the topologies and dynamics of these motifs can enable memory encoding and storage.
Collapse
Affiliation(s)
- Yanfei Jiang
- Section of Molecular Biology, Division of Biological Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Nan Hao
- Section of Molecular Biology, Division of Biological Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
20
|
Patwardhan A, Cheng N, Trejo J. Post-Translational Modifications of G Protein-Coupled Receptors Control Cellular Signaling Dynamics in Space and Time. Pharmacol Rev 2021; 73:120-151. [PMID: 33268549 PMCID: PMC7736832 DOI: 10.1124/pharmrev.120.000082] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are a large family comprising >800 signaling receptors that regulate numerous cellular and physiologic responses. GPCRs have been implicated in numerous diseases and represent the largest class of drug targets. Although advances in GPCR structure and pharmacology have improved drug discovery, the regulation of GPCR function by diverse post-translational modifications (PTMs) has received minimal attention. Over 200 PTMs are known to exist in mammalian cells, yet only a few have been reported for GPCRs. Early studies revealed phosphorylation as a major regulator of GPCR signaling, whereas later reports implicated a function for ubiquitination, glycosylation, and palmitoylation in GPCR biology. Although our knowledge of GPCR phosphorylation is extensive, our knowledge of the modifying enzymes, regulation, and function of other GPCR PTMs is limited. In this review we provide a comprehensive overview of GPCR post-translational modifications with a greater focus on new discoveries. We discuss the subcellular location and regulatory mechanisms that control post-translational modifications of GPCRs. The functional implications of newly discovered GPCR PTMs on receptor folding, biosynthesis, endocytic trafficking, dimerization, compartmentalized signaling, and biased signaling are also provided. Methods to detect and study GPCR PTMs as well as PTM crosstalk are further highlighted. Finally, we conclude with a discussion of the implications of GPCR PTMs in human disease and their importance for drug discovery. SIGNIFICANCE STATEMENT: Post-translational modification of G protein-coupled receptors (GPCRs) controls all aspects of receptor function; however, the detection and study of diverse types of GPCR modifications are limited. A thorough understanding of the role and mechanisms by which diverse post-translational modifications regulate GPCR signaling and trafficking is essential for understanding dysregulated mechanisms in disease and for improving and refining drug development for GPCRs.
Collapse
Affiliation(s)
- Anand Patwardhan
- Department of Pharmacology and the Biomedical Sciences Graduate Program, School of Medicine, University of California, San Diego, La Jolla, California
| | - Norton Cheng
- Department of Pharmacology and the Biomedical Sciences Graduate Program, School of Medicine, University of California, San Diego, La Jolla, California
| | - JoAnn Trejo
- Department of Pharmacology and the Biomedical Sciences Graduate Program, School of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
21
|
Laufkötter C, Zscheischler J, Frölicher TL. High-impact marine heatwaves attributable to human-induced global warming. Science 2020; 369:1621-1625. [PMID: 32973027 DOI: 10.1126/science.aba0690] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 08/04/2020] [Indexed: 01/01/2023]
Abstract
Marine heatwaves (MHWs)-periods of extremely high ocean temperatures in specific regions-have occurred in all of Earth's ocean basins over the past two decades, with severe negative impacts on marine organisms and ecosystems. However, for most individual MHWs, it is unclear to what extent they have been altered by human-induced climate change. We show that the occurrence probabilities of the duration, intensity, and cumulative intensity of most documented, large, and impactful MHWs have increased more than 20-fold as a result of anthropogenic climate change. MHWs that occurred only once every hundreds to thousands of years in the preindustrial climate are projected to become decadal to centennial events under 1.5°C warming conditions and annual to decadal events under 3°C warming conditions. Thus, ambitious climate targets are indispensable to reduce the risks of substantial MHW impacts.
Collapse
Affiliation(s)
- Charlotte Laufkötter
- Climate and Environmental Physics, Physics Institute, University of Bern, 3012 Bern, Switzerland. .,Oeschger Centre for Climate Change Research, University of Bern, 3012 Bern, Switzerland
| | - Jakob Zscheischler
- Climate and Environmental Physics, Physics Institute, University of Bern, 3012 Bern, Switzerland.,Oeschger Centre for Climate Change Research, University of Bern, 3012 Bern, Switzerland
| | - Thomas L Frölicher
- Climate and Environmental Physics, Physics Institute, University of Bern, 3012 Bern, Switzerland.,Oeschger Centre for Climate Change Research, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
22
|
Baro Graf C, Ritagliati C, Stival C, Luque GM, Gentile I, Buffone MG, Krapf D. Everything you ever wanted to know about PKA regulation and its involvement in mammalian sperm capacitation. Mol Cell Endocrinol 2020; 518:110992. [PMID: 32853743 DOI: 10.1016/j.mce.2020.110992] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/29/2022]
Abstract
The 3', 5'-cyclic adenosine monophosphate (cAMP) dependent protein kinase (PKA) is a tetrameric holoenzyme comprising a set of two regulatory subunits (PKA-R) and two catalytic (PKA-C) subunits. The PKA-R subunits act as sensors of cAMP and allow PKA-C activity. One of the first signaling events observed during mammalian sperm capacitation is PKA activation. Thus, understanding how PKA activity is restricted in space and time is crucial to decipher the critical steps of sperm capacitation. It is widely accepted that PKA specificity depends on several levels of regulation. Anchoring proteins play a pivotal role in achieving proper localization signaling, subcellular targeting and cAMP microdomains. These multi-factorial regulation steps are necessary for a precise spatio-temporal activation of PKA. Here we discuss recent understanding of regulatory mechanisms of PKA in mammalian sperm, such as post-translational modifications, in the context of its role as the master orchestrator of molecular events conducive to capacitation.
Collapse
Affiliation(s)
- Carolina Baro Graf
- Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET-UNR, Rosario, Argentina; Laboratorio de Medicina Reproductiva (LMR), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Carla Ritagliati
- Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET-UNR, Rosario, Argentina
| | - Cintia Stival
- Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET-UNR, Rosario, Argentina
| | - Guillermina M Luque
- Laboratory of Cellular and Molecular Reproductive Biology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Iñaki Gentile
- Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET-UNR, Rosario, Argentina
| | - Mariano G Buffone
- Laboratory of Cellular and Molecular Reproductive Biology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Dario Krapf
- Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET-UNR, Rosario, Argentina; Laboratorio de Medicina Reproductiva (LMR), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina.
| |
Collapse
|
23
|
Abdjan MI, Aminah NS, Siswanto I, Thant TM, Kristanti AN, Takaya Y. In silico approach: biological prediction of nordentatin derivatives as anticancer agent inhibitors in the cAMP pathway. RSC Adv 2020; 10:42733-42743. [PMID: 35514899 PMCID: PMC9058016 DOI: 10.1039/d0ra07838g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 11/06/2020] [Indexed: 11/23/2022] Open
Abstract
A combination of computational techniques has been carried out to predict the binding of nordentatin derivatives based on pyranocoumarin semi-synthesis with the target protein from the expression of the PDE4B gene. The inhibition of the cAMP pathway is the main target of anti-cancer drugs, which is responsible for uncontrolled cell division in cancer. Modeling was done using a combination of semi-empirical methods and the density functional theory (PM3-DFT/6-31G*/B3LYP) to obtain the optimal structure of a small ligand that could be modeled. Studies on the interaction of the ligands and amino acid residues on protein targets were carried out using a combination of molecular docking and molecular dynamic simulation. Molecular docking based on functional grid scores showed a very good native ligand pose with an RMSD of 0.93 Å in determining the initial coordinates of the ligand-receptor interactions. Furthermore, the amino acid residues responsible for interaction through H-bonds were Tyr103, His104, His177, Met217, and Gln313. The binding free energy (kcal mol-1) results of the candidates were PS-1 (-36.84 ± 0.31), PS-2 (-35.34 ± 0.28), PS-3 (-26.65 ± 0.30), PS-5 (-42.66 ± 0.26), PS-7 (-35.33 ± 0.23), and PS-9 (-32.57 ± 0.20), which are smaller than that of the native ligand Z72 (-24.20 ± 0.19), and thus these have good potential as drugs that can inhibit the cAMP pathway. These results provide theoretical information for the efficient inhibition of the cAMP pathway in the future.
Collapse
Affiliation(s)
- Muhammad Ikhlas Abdjan
- Departement of Chemistry, Faculty of Science and Technology, Universitas Airlangga Surabaya 60115 Indonesia
| | - Nanik Siti Aminah
- Departement of Chemistry, Faculty of Science and Technology, Universitas Airlangga Surabaya 60115 Indonesia
- Biotechnology of Tropical Medicinal Plants Research Group, Universitas Airlangga Indonesia
| | - Imam Siswanto
- Departement of Chemistry, Faculty of Science and Technology, Universitas Airlangga Surabaya 60115 Indonesia
| | - Tin Myo Thant
- Department of Chemistry, Faculty of Science and Technology, Universitas Airlangga, Komplek Kampus C Jl. Mulyorejo Surabaya Indonesia 60115
- Department of Chemistry, Mandalar Degree College Mandalay Myanmar
| | - Alfinda Novi Kristanti
- Departement of Chemistry, Faculty of Science and Technology, Universitas Airlangga Surabaya 60115 Indonesia
- Biotechnology of Tropical Medicinal Plants Research Group, Universitas Airlangga Indonesia
| | - Yoshiaki Takaya
- Faculty of Pharmacy, Meijo University 150 Yagotoyama, Tempaku Nagoya 468-8503 Japan
| |
Collapse
|
24
|
Schuster S, Heuten E, Velic A, Admard J, Synofzik M, Ossowski S, Macek B, Hauser S, Schöls L. CHIP mutations affect the heat shock response differently in human fibroblasts and iPSC-derived neurons. Dis Model Mech 2020; 13:13/10/dmm045096. [PMID: 33097556 PMCID: PMC7578354 DOI: 10.1242/dmm.045096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 08/10/2020] [Indexed: 01/09/2023] Open
Abstract
C-terminus of HSC70-interacting protein (CHIP) encoded by the gene STUB1 is a co-chaperone and E3 ligase that acts as a key regulator of cellular protein homeostasis. Mutations in STUB1 cause autosomal recessive spinocerebellar ataxia type 16 (SCAR16) with widespread neurodegeneration manifesting as spastic-ataxic gait disorder, dementia and epilepsy. CHIP-/- mice display severe cerebellar atrophy, show high perinatal lethality and impaired heat stress tolerance. To decipher the pathomechanism underlying SCAR16, we investigated the heat shock response (HSR) in primary fibroblasts of three SCAR16 patients. We found impaired HSR induction and recovery compared to healthy controls. HSPA1A/B transcript levels (coding for HSP70) were reduced upon heat shock but HSP70 remained higher upon recovery in patient- compared to control-fibroblasts. As SCAR16 primarily affects the central nervous system we next investigated the HSR in cortical neurons (CNs) derived from induced pluripotent stem cells of SCAR16 patients. We found CNs of patients and controls to be surprisingly resistant to heat stress with high basal levels of HSP70 compared to fibroblasts. Although heat stress resulted in strong transcript level increases of many HSPs, this did not translate into higher HSP70 protein levels upon heat shock, independent of STUB1 mutations. Furthermore, STUB1(-/-) neurons generated by CRISPR/Cas9-mediated genome editing from an isogenic healthy control line showed a similar HSR to patients. Proteomic analysis of CNs showed dysfunctional protein (re)folding and higher basal oxidative stress levels in patients. Our results question the role of impaired HSR in SCAR16 neuropathology and highlight the need for careful selection of proper cell types for modeling human diseases.
Collapse
Affiliation(s)
- S Schuster
- Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany.,Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany.,Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - E Heuten
- Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - A Velic
- Proteome Center Tübingen, University of Tübingen, 72076 Tübingen, Germany
| | - J Admard
- Institute for Medical Genetics and Applied Genomics, University of Tübingen, 72076 Tübingen, Germany
| | - M Synofzik
- Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - S Ossowski
- Institute for Medical Genetics and Applied Genomics, University of Tübingen, 72076 Tübingen, Germany
| | - B Macek
- Proteome Center Tübingen, University of Tübingen, 72076 Tübingen, Germany
| | - S Hauser
- Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany .,Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - L Schöls
- Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany .,Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| |
Collapse
|
25
|
Dhyani V, Gare S, Gupta RK, Swain S, Venkatesh K, Giri L. GPCR mediated control of calcium dynamics: A systems perspective. Cell Signal 2020; 74:109717. [PMID: 32711109 PMCID: PMC7375278 DOI: 10.1016/j.cellsig.2020.109717] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 02/09/2023]
Abstract
G-protein coupled receptor (GPCR) mediated calcium (Ca2+)-signaling transduction remains crucial in designing drugs for various complex diseases including neurodegeneration, chronic heart failure as well as respiratory diseases. Although there are several reviews detailing various aspects of Ca2+-signaling such as the role of IP3 receptors and Ca2+-induced-Ca2+-release, none of them provide an integrated view of the mathematical descriptions of GPCR signal transduction and investigations on dose-response curves. This article is the first study in reviewing the network structures underlying GPCR signal transduction that control downstream [Cac2+]-oscillations. The central theme of this paper is to present the biochemical pathways, as well as molecular mechanisms underlying the GPCR-mediated Ca2+-dynamics in order to facilitate a better understanding of how agonist concentration is encoded in Ca2+-signals for Gαq, Gαs, and Gαi/o signaling pathways. Moreover, we present the GPCR targeting drugs that are relevant for treating cardiac, respiratory, and neuro-diseases. The current paper presents the ODE formulation for various models along with the detailed schematics of signaling networks. To provide a systems perspective, we present the network motifs that can provide readers an insight into the complex and intriguing science of agonist-mediated Ca2+-dynamics. One of the features of this review is to pinpoint the interplay between positive and negative feedback loops that are involved in controlling intracellular [Cac2+]-oscillations. Furthermore, we review several examples of dose-response curves obtained from [Cac2+]-spiking for various GPCR pathways. This paper is expected to be useful for pharmacologists and computational biologists for designing clinical applications of GPCR targeting drugs through modulation of Ca2+-dynamics.
Collapse
Affiliation(s)
- Vaibhav Dhyani
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
| | - Suman Gare
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
| | - Rishikesh Kumar Gupta
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
| | - Sarpras Swain
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
| | - K.V. Venkatesh
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, India
| | - Lopamudra Giri
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India.
| |
Collapse
|
26
|
CHIP as a therapeutic target for neurological diseases. Cell Death Dis 2020; 11:727. [PMID: 32908122 PMCID: PMC7481199 DOI: 10.1038/s41419-020-02953-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 08/16/2020] [Accepted: 08/27/2020] [Indexed: 12/12/2022]
Abstract
Carboxy-terminus of Hsc70-interacting protein (CHIP) functions both as a molecular co-chaperone and ubiquitin E3 ligase playing a critical role in modulating the degradation of numerous chaperone-bound proteins. To date, it has been implicated in the regulation of numerous biological functions, including misfolded-protein refolding, autophagy, immunity, and necroptosis. Moreover, the ubiquitous expression of CHIP in the central nervous system suggests that it may be implicated in a wide range of functions in neurological diseases. Several recent studies of our laboratory and other groups have highlighted the beneficial role of CHIP in the pathogenesis of several neurological diseases. The objective of this review is to discuss the possible molecular mechanisms that contribute to the pathogenesis of neurological diseases in which CHIP has a pivotal role, such as stroke, intracerebral hemorrhage, Alzheimer's disease, Parkinson's disease, and polyglutamine diseases; furthermore, CHIP mutations could also cause neurodegenerative diseases. Based on the available literature, CHIP overexpression could serve as a promising therapeutic target for several neurological diseases.
Collapse
|
27
|
The Proteasome Governs Fungal Morphogenesis via Functional Connections with Hsp90 and cAMP-Protein Kinase A Signaling. mBio 2020; 11:mBio.00290-20. [PMID: 32317319 PMCID: PMC7175089 DOI: 10.1128/mbio.00290-20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Protein homeostasis is critical for proliferation and viability of all organisms. For Candida albicans, protein homeostasis also modulates the transition between yeast and filamentous forms, which is critical for virulence. A key regulator of morphogenesis is the molecular chaperone Hsp90, which mediates proteostasis under physiological and stress conditions. Hsp90 regulates morphogenesis by repressing cyclic AMP-protein kinase A (cAMP-PKA) signaling, such that inhibition of Hsp90 causes filamentation in the absence of an inducing cue. We explored the effect of perturbation of another facet of protein homeostasis and discovered that morphogenesis is also regulated by the proteasome, a large 33-subunit protein complex consisting of a 20S catalytic core and two 19S regulatory particles, which controls degradation of intracellular proteins. We identified a conserved role of the proteasome in morphogenesis as pharmacological inhibition of the proteasome induced filamentation of C. albicans and the related species Candida dubliniensis, Candida tropicalis, Candida krusei, and Candida parapsilosis For C. albicans, genetic depletion of any of 29 subunits of the 19S or 20S particle induced filamentation. Filaments induced by inhibition of either the proteasome or Hsp90 have shared structural characteristics, such as aberrant nuclear content, and shared genetic dependencies, such as intact cAMP-PKA signaling. Consistent with a functional connection between these facets of protein homeostasis that modulate morphogenesis, we observed that proteasome inhibition results in an accumulation of ubiquitinated proteins that overwhelm Hsp90 function, relieving Hsp90-mediated repression of morphogenesis. Together, our findings provide a mechanism whereby interconnected facets of proteostasis regulate C. albicans morphogenesis.IMPORTANCE Fungi cause life-threatening infections and pose a serious threat to human health as there are very few effective antifungal drugs. Candida albicans is a major human fungal pathogen and cause of morbidity and mortality in immunocompromised individuals. A key trait that enables C. albicans virulence is its ability to transition between yeast and filamentous forms. Understanding the mechanisms regulating this virulence trait can facilitate the development of much-needed, novel therapeutic strategies. A key regulator of morphogenesis is the molecular chaperone Hsp90, which is crucial for proteostasis. Here, we expanded our understanding of how proteostasis regulates fungal morphogenesis and identified the proteasome as a repressor of filamentation in C. albicans and related species. Our work suggests that proteasome inhibition overwhelms Hsp90 function, thereby inducing morphogenesis. This work provides a foundation for understanding the role of the proteasome in fungal virulence and offers potential for targeting the proteasome to disarm fungal pathogens.
Collapse
|
28
|
Bustamante HA, Cereceda K, González AE, Valenzuela GE, Cheuquemilla Y, Hernández S, Arias-Muñoz E, Cerda-Troncoso C, Bandau S, Soza A, Kausel G, Kerr B, Mardones GA, Cancino J, Hay RT, Rojas-Fernandez A, Burgos PV. The Proteasomal Deubiquitinating Enzyme PSMD14 Regulates Macroautophagy by Controlling Golgi-to-ER Retrograde Transport. Cells 2020; 9:E777. [PMID: 32210007 PMCID: PMC7140897 DOI: 10.3390/cells9030777] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/16/2020] [Accepted: 03/19/2020] [Indexed: 12/15/2022] Open
Abstract
Ubiquitination regulates several biological processes, however the role of specific members of the ubiquitinome on intracellular membrane trafficking is not yet fully understood. Here, we search for ubiquitin-related genes implicated in protein membrane trafficking performing a High-Content siRNA Screening including 1187 genes of the human "ubiquitinome" using amyloid precursor protein (APP) as a reporter. We identified the deubiquitinating enzyme PSMD14, a subunit of the 19S regulatory particle of the proteasome, specific for K63-Ub chains in cells, as a novel regulator of Golgi-to-endoplasmic reticulum (ER) retrograde transport. Silencing or pharmacological inhibition of PSMD14 with Capzimin (CZM) caused a robust increase in APP levels at the Golgi apparatus and the swelling of this organelle. We showed that this phenotype is the result of rapid inhibition of Golgi-to-ER retrograde transport, a pathway implicated in the early steps of the autophagosomal formation. Indeed, we observed that inhibition of PSMD14 with CZM acts as a potent blocker of macroautophagy by a mechanism related to the retention of Atg9A and Rab1A at the Golgi apparatus. As pharmacological inhibition of the proteolytic core of the 20S proteasome did not recapitulate these effects, we concluded that PSMD14, and the K63-Ub chains, act as a crucial regulatory factor for macroautophagy by controlling Golgi-to-ER retrograde transport.
Collapse
Affiliation(s)
- Hianara A Bustamante
- Instituto de Fisiología, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5110566, Chile; (H.A.B.); (A.E.G.); (G.A.M.)
| | - Karina Cereceda
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile; (K.C.); (S.H.); (E.A.-M.); (C.C.-T.); (A.S.); (B.K.); (J.C.)
| | - Alexis E González
- Instituto de Fisiología, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5110566, Chile; (H.A.B.); (A.E.G.); (G.A.M.)
| | - Guillermo E Valenzuela
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile; (G.E.V.); (G.K.)
- Instituto de Medicina & Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia 5110566, Chile;
| | - Yorka Cheuquemilla
- Instituto de Medicina & Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia 5110566, Chile;
| | - Sergio Hernández
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile; (K.C.); (S.H.); (E.A.-M.); (C.C.-T.); (A.S.); (B.K.); (J.C.)
| | - Eloisa Arias-Muñoz
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile; (K.C.); (S.H.); (E.A.-M.); (C.C.-T.); (A.S.); (B.K.); (J.C.)
| | - Cristóbal Cerda-Troncoso
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile; (K.C.); (S.H.); (E.A.-M.); (C.C.-T.); (A.S.); (B.K.); (J.C.)
| | - Susanne Bandau
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, DD1 4HN, Dundee DD1 4HN UK; (S.B.); (R.T.H.)
| | - Andrea Soza
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile; (K.C.); (S.H.); (E.A.-M.); (C.C.-T.); (A.S.); (B.K.); (J.C.)
| | - Gudrun Kausel
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile; (G.E.V.); (G.K.)
| | - Bredford Kerr
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile; (K.C.); (S.H.); (E.A.-M.); (C.C.-T.); (A.S.); (B.K.); (J.C.)
| | - Gonzalo A Mardones
- Instituto de Fisiología, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5110566, Chile; (H.A.B.); (A.E.G.); (G.A.M.)
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Jorge Cancino
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile; (K.C.); (S.H.); (E.A.-M.); (C.C.-T.); (A.S.); (B.K.); (J.C.)
| | - Ronald T Hay
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, DD1 4HN, Dundee DD1 4HN UK; (S.B.); (R.T.H.)
| | - Alejandro Rojas-Fernandez
- Instituto de Medicina & Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia 5110566, Chile;
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, DD1 4HN, Dundee DD1 4HN UK; (S.B.); (R.T.H.)
| | - Patricia V Burgos
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile; (K.C.); (S.H.); (E.A.-M.); (C.C.-T.); (A.S.); (B.K.); (J.C.)
- Centro de Envejecimiento y Regeneración (CARE-UC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 83330023, Chile
| |
Collapse
|
29
|
STUB1 is targeted by the SUMO-interacting motif of EBNA1 to maintain Epstein-Barr Virus latency. PLoS Pathog 2020; 16:e1008447. [PMID: 32176739 PMCID: PMC7105294 DOI: 10.1371/journal.ppat.1008447] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/30/2020] [Accepted: 03/01/2020] [Indexed: 12/31/2022] Open
Abstract
Latent Epstein-Barr virus (EBV) infection is strongly associated with several malignancies, including B-cell lymphomas and epithelial tumors. EBNA1 is a key antigen expressed in all EBV-associated tumors during latency that is required for maintenance of the EBV episome DNA and the regulation of viral gene transcription. However, the mechanism utilized by EBV to maintain latent infection at the levels of posttranslational regulation remains largely unclear. Here, we report that EBNA1 contains two SUMO-interacting motifs (SIM2 and SIM3), and mutation of SIM2, but not SIM3, dramatically disrupts the EBNA1 dimerization, while SIM3 contributes to the polySUMO2 modification of EBNA1 at lysine 477 in vitro. Proteomic and immunoprecipitation analyses further reveal that the SIM3 motif is required for the EBNA1-mediated inhibitory effects on SUMO2-modified STUB1, SUMO2-mediated degradation of USP7, and SUMO1-modified KAP1. Deletion of the EBNASIM motif leads to functional loss of both EBNA1-mediated viral episome maintenance and lytic gene silencing. Importantly, hypoxic stress induces the SUMO2 modification of EBNA1, and in turn the dissociation of EBNA1 with STUB1, KAP1 and USP7 to increase the SUMO1 modification of both STUB1 and KAP1 for reactivation of lytic replication. Therefore, the EBNA1SIM motif plays an essential role in EBV latency and is a potential therapeutic target against EBV-associated cancers. The Small Ubiquitin-related modifier (SUMO) modification of proteins is a reversible post-translational regulation involved in control of gene transcription, among other functions. Epstein-Barr virus (EBV) infects most people worldwide and contributes to the development of several types of cancers due to its ability to induce cell proliferation and survival. EBNA1 is expressed in all forms of EBV-associated tumors. In this study, we found that EBNA1 contains a SUMO-interacting motif (SIM) named EBNA1SIM, which is required for EBNA1 to exert inhibitory effects on a SUMO2-modified complex (SC2) including STUB1, KAP1 and USP7. Disruption of EBNA1SIM leads to loss of both EBNA1-mediated viral episome maintenance and lytic gene silencing. Importantly, hypoxia-mediated reactivation of viral lytic replication induces the EBNA1 dissociation from STUB1 in the SC2 complex. This discovery not only opens a new insight on the interplay between host and virus, but it also provides a therapeutic target specific against EBV-associated cancers.
Collapse
|
30
|
Cyclin-Dependent Kinase 18 Controls Trafficking of Aquaporin-2 and Its Abundance through Ubiquitin Ligase STUB1, Which Functions as an AKAP. Cells 2020; 9:cells9030673. [PMID: 32164329 PMCID: PMC7140648 DOI: 10.3390/cells9030673] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/27/2020] [Accepted: 03/09/2020] [Indexed: 12/24/2022] Open
Abstract
Arginine-vasopressin (AVP) facilitates water reabsorption in renal collecting duct principal cells through regulation of the water channel aquaporin-2 (AQP2). The hormone binds to vasopressin V2 receptors (V2R) on the surface of the cells and stimulates cAMP synthesis. The cAMP activates protein kinase A (PKA), which initiates signaling that causes an accumulation of AQP2 in the plasma membrane of the cells facilitating water reabsorption from primary urine and fine-tuning of body water homeostasis. AVP-mediated PKA activation also causes an increase in the AQP2 protein abundance through a mechanism that involves dephosphorylation of AQP2 at serine 261 and a decrease in its poly-ubiquitination. However, the signaling downstream of PKA that controls the localization and abundance of AQP2 is incompletely understood. We carried out an siRNA screen targeting 719 kinase-related genes, representing the majority of the kinases of the human genome and analyzed the effect of the knockdown on AQP2 by high-content imaging and biochemical approaches. The screening identified 13 hits whose knockdown inhibited the AQP2 accumulation in the plasma membrane. Amongst the candidates was the so far hardly characterized cyclin-dependent kinase 18 (CDK18). Our further analysis revealed a hitherto unrecognized signalosome comprising CDK18, an E3 ubiquitin ligase, STUB1 (CHIP), PKA and AQP2 that controls the localization and abundance of AQP2. CDK18 controls AQP2 through phosphorylation at serine 261 and STUB1-mediated ubiquitination. STUB1 functions as an A-kinase anchoring protein (AKAP) tethering PKA to the protein complex and bridging AQP2 and CDK18. The modulation of the protein complex may lead to novel concepts for the treatment of disorders which are caused or are associated with dysregulated AQP2 and for which a satisfactory treatment is not available, e.g., hyponatremia, liver cirrhosis, diabetes insipidus, ADPKD or heart failure.
Collapse
|
31
|
Teijeiro JM, Marini PE. Hormone-regulated PKA activity in porcine oviductal epithelial cells. Cell Tissue Res 2020; 380:657-667. [PMID: 32112257 DOI: 10.1007/s00441-020-03180-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/28/2020] [Indexed: 11/24/2022]
Abstract
The oviduct is a dynamic organ that suffers changes during the oestrous cycle and modulates gamete and embryo physiology. We analyse the possible existence of Protein kinase A (PKA)-dependent hormone-regulated pathways in porcine ampulla and primary cell cultures by 2D-electrophoresis/Western blot using anti-phospho PKA substrate antibodies. Differential phosphorylation was observed for ten proteins that were identified by mass spectrometry. The results were validated for five of the proteins: Annexin A5, Calumenin, Glyoxalase I and II and Enolase I. Immunofluorescence analyses show that Calumenin, Glyoxalase II and Enolase I change their localisation in the oviductal epithelium through the oestrus cycle. The results demonstrate the existence of PKA hormone-regulated pathways in the ampulla epithelium during the oestrus cycle.
Collapse
Affiliation(s)
- Juan Manuel Teijeiro
- Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina. .,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rosario, Argentina.
| | - Patricia Estela Marini
- Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina.,Consejo de Investigaciones de la Universidad Nacional de Rosario (CIUNR), Rosario, Argentina.,Instituto de Biología Molecular y Celular de Rosario, IBR-CONICET, Rosario, Argentina
| |
Collapse
|
32
|
Tschaikner P, Enzler F, Torres-Quesada O, Aanstad P, Stefan E. Hedgehog and Gpr161: Regulating cAMP Signaling in the Primary Cilium. Cells 2020; 9:E118. [PMID: 31947770 PMCID: PMC7017137 DOI: 10.3390/cells9010118] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/20/2019] [Accepted: 12/29/2019] [Indexed: 12/14/2022] Open
Abstract
Compartmentalization of diverse types of signaling molecules contributes to the precise coordination of signal propagation. The primary cilium fulfills this function by acting as a spatiotemporally confined sensory signaling platform. For the integrity of ciliary signaling, it is mandatory that the ciliary signaling pathways are constantly attuned by alterations in both oscillating small molecules and the presence or absence of their sensor/effector proteins. In this context, ciliary G protein-coupled receptor (GPCR) pathways participate in coordinating the mobilization of the diffusible second messenger molecule 3',5'-cyclic adenosine monophosphate (cAMP). cAMP fluxes in the cilium are primarily sensed by protein kinase A (PKA) complexes, which are essential for the basal repression of Hedgehog (Hh) signaling. Here, we describe the dynamic properties of underlying signaling circuits, as well as strategies for second messenger compartmentalization. As an example, we summarize how receptor-guided cAMP-effector pathways control the off state of Hh signaling. We discuss the evidence that a macromolecular, ciliary-localized signaling complex, composed of the orphan GPCR Gpr161 and type I PKA holoenzymes, is involved in antagonizing Hh functions. Finally, we outline how ciliary cAMP-linked receptor pathways and cAMP-sensing signalosomes may become targets for more efficient combinatory therapy approaches to counteract dysregulation of Hh signaling.
Collapse
Affiliation(s)
- Philipp Tschaikner
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (P.T.); (F.E.); (O.T.-Q.)
- Institute of Molecular Biology, University of Innsbruck, 6020 Innsbruck, Austria;
| | - Florian Enzler
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (P.T.); (F.E.); (O.T.-Q.)
| | - Omar Torres-Quesada
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (P.T.); (F.E.); (O.T.-Q.)
| | - Pia Aanstad
- Institute of Molecular Biology, University of Innsbruck, 6020 Innsbruck, Austria;
| | - Eduard Stefan
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (P.T.); (F.E.); (O.T.-Q.)
| |
Collapse
|
33
|
Schernthaner-Reiter MH, Trivellin G, Stratakis CA. Chaperones, somatotroph tumors and the cyclic AMP (cAMP)-dependent protein kinase (PKA) pathway. Mol Cell Endocrinol 2020; 499:110607. [PMID: 31586652 DOI: 10.1016/j.mce.2019.110607] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/28/2019] [Accepted: 09/30/2019] [Indexed: 02/08/2023]
Abstract
The cAMP-PKA pathway plays an essential role in the pituitary gland, governing cell differentiation and survival, and maintenance of endocrine function. Somatotroph growth hormone transcription and release as well as cell proliferation are regulated by the cAMP-PKA pathway; cAMP-PKA pathway abnormalities are frequently detected in sporadic as well as in hereditary somatotroph tumors and more rarely in other pituitary tumors. Inactivating variants of the aryl hydrocarbon receptor-interacting protein (AIP)-coding gene are the genetic cause of a subset of familial isolated pituitary adenomas (FIPA). Multiple functional links between the co-chaperone AIP and the cAMP-PKA pathway have been described. This review explores the role of chaperones including AIP in normal pituitary function as well as in somatotroph tumors, and their interaction with the cAMP-PKA pathway.
Collapse
Affiliation(s)
| | - Giampaolo Trivellin
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, USA
| | - Constantine A Stratakis
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, USA
| |
Collapse
|