1
|
Wang X, Zhi H, Zhang Z, Li J, Guo D. REV-ERBα Mitigates Astrocyte Activation and Protects Dopaminergic Neurons from Damage. J Mol Neurosci 2024; 74:84. [PMID: 39254874 DOI: 10.1007/s12031-024-02264-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024]
Abstract
Parkinson's disease (PD) is characterized by astrocyte activation and disruptions in circadian rhythm. Within the astrocyte population, two distinct reactive states exist: A1 and A2. A1 astrocytes are associated with neurotoxicity and inflammation, while A2 astrocytes exhibit neuroprotective functions. Our investigation focused on the role of REV-ERBα, a member of the nuclear receptor superfamily and a key regulator of the circadian clock, in astrocyte activation. We observed that REV-ERBα expression in A1 astrocytes was reduced to one-third of its normal level. Notably, activation of REV-ERBα prompted a transformation of astrocytes from A1 to A2. Mechanistically, REV-ERBα inhibition was linked to the classical NF-κB pathway, while it concurrently suppressed the STAT3 pathway. Furthermore, astrocytes with low REV-ERBα expression were associated with dopaminergic neurons apoptosis. Intriguingly, the opposite effect was observed when using a REV-ERBα agonist, which mitigated astrocyte activation and reduced dopaminergic neuron damage by 50%. In summary, our study elucidates the pivotal role of REV-ERBα in modulating astrocyte function and its potential implications in PD pathogenesis.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215153, China
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Hui Zhi
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215153, China
| | - Zongqin Zhang
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215153, China
| | - Jingwei Li
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215153, China.
| | - Dongkai Guo
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215153, China.
| |
Collapse
|
2
|
Palomba M, Vecchio D, Allavena G, Capaccio V, De Mei C, Scarpelli R, Grimaldi B. Identification of a Dual Autophagy and REV-ERB Inhibitor with in Vivo Anticancer Efficacy. J Med Chem 2024; 67:349-379. [PMID: 38117953 PMCID: PMC10788905 DOI: 10.1021/acs.jmedchem.3c01432] [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: 08/04/2023] [Revised: 10/20/2023] [Accepted: 11/22/2023] [Indexed: 12/22/2023]
Abstract
The autophagy process appears as a promising target for anticancer interventions. Chloroquine (CQ) and its derivative hydroxychloroquine (HCQ) are the only FDA-approved autophagy flux inhibitors. Although diverse anticancer clinical trials are providing encouraging results, several limitations associated with the need of high dosage and long-term administration of these autophagy inhibitors are also emerging. We showed that the inhibition of REV-ERB, a nuclear receptor regulating circadian rhythm and metabolism, enhances CQ-mediated cancer cell death and identified a class of dual inhibitors of autophagy and REV-ERB displaying an in vitro anticancer activity against diverse tumor cells greatly higher than CQ. Herein, we describe our lead optimization strategy that led to the identification of compound 24 as a dual autophagy and REV-ERB inhibitor, showing improved potency in blocking autophagy, enhanced toxicity against cancer cells, optimal drug-like properties, and efficacy in a mouse xenograft model of melanoma as a single anticancer agent.
Collapse
Affiliation(s)
- Martina Palomba
- Molecular
Medicine, Medicinal Chemistry and Technologies for Drug Discovery and Delivery
Facility, Nanomaterials for Biomedical Applications, Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy
| | - Donatella Vecchio
- Molecular
Medicine, Medicinal Chemistry and Technologies for Drug Discovery and Delivery
Facility, Nanomaterials for Biomedical Applications, Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy
| | - Giulia Allavena
- Molecular
Medicine, Medicinal Chemistry and Technologies for Drug Discovery and Delivery
Facility, Nanomaterials for Biomedical Applications, Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy
| | - Vito Capaccio
- Molecular
Medicine, Medicinal Chemistry and Technologies for Drug Discovery and Delivery
Facility, Nanomaterials for Biomedical Applications, Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy
| | - Claudia De Mei
- Molecular
Medicine, Medicinal Chemistry and Technologies for Drug Discovery and Delivery
Facility, Nanomaterials for Biomedical Applications, Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy
| | - Rita Scarpelli
- Molecular
Medicine, Medicinal Chemistry and Technologies for Drug Discovery and Delivery
Facility, Nanomaterials for Biomedical Applications, Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy
| | - Benedetto Grimaldi
- Molecular
Medicine, Medicinal Chemistry and Technologies for Drug Discovery and Delivery
Facility, Nanomaterials for Biomedical Applications, Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy
| |
Collapse
|
3
|
Xiao X, Chen S, Huang Z, Han X, Dou C, Kang J, Wang T, Xie H, Zhang L, Hei Z, Li H, Yao W. SerpinB1 is required for Rev-erbα-mediated protection against acute lung injury induced by lipopolysaccharide-in mice. Br J Pharmacol 2023; 180:3234-3253. [PMID: 37350044 DOI: 10.1111/bph.16175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 05/05/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND AND PURPOSE Acute lung injury (ALI) is a serious, life-threatening inflammation of the lungs that still lacks effective treatment. We previously showed that serine protease inhibitor B1 (SerpinB1) protects against ALI induced by orthotopic autologous liver transplantation. However, the role of SerpinB1 in lipopolysaccharide (LPS)-induced ALI and its regulatory mechanisms are not known. EXPERIMENTAL APPROACH Wild-type (WT) and SerpinB1 knockout (KO) mice were treated with intratracheal LPS stimulation to induce ALI. Some of the WT and KO mice were injected i.p. with melatonin, a rhythm-related protein Rev-erbα agonist. The circadian rhythm in WT mice was disrupted by exposing mice to 24 h of continuous dark or light conditions after intratracheal LPS. Neutrophils were isolated from alveolar lavage fluid of WT and KO mice, and from human peripheral blood. Neutrophils were treated with LPS and melatonin. KEY RESULTS Disruption of circadian rhythm by either 24-h dark or light conditions exacerbated LPS-induced ALI and decreased expression of Rev-erbα and SerpinB1 protein in lung, whereas melatonin treatment increased SerpinB1 expression and attenuated LPS-induced ALI in WT mice, but not in KO mice. In isolated neutrophils, Rev-erbα was co-localized with SerpinB1 and bound to its promoter to trigger SerpinB1 transcription. Furthermore, LPS stimulation increased formation of neutrophil extracellular traps, which was reversed by melatonin treatment in neutrophils from WT mice, but not from KO mice. CONCLUSION AND IMPLICATIONS In mice, SerpinB1 is rhythmically regulated by Rev-erbα, and its down-regulation exacerbates LPS-induced ALI by inducing formation of neutrophil extracellular traps.
Collapse
Affiliation(s)
- Xue Xiao
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sufang Chen
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ziyan Huang
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xue Han
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chaoxun Dou
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiayi Kang
- Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tienan Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hanbin Xie
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Linan Zhang
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ziqing Hei
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Haobo Li
- Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Weifeng Yao
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Abenza JF, Rossetti L, Mouelhi M, Burgués J, Andreu I, Kennedy K, Roca-Cusachs P, Marco S, García-Ojalvo J, Trepat X. Mechanical control of the mammalian circadian clock via YAP/TAZ and TEAD. J Cell Biol 2023; 222:e202209120. [PMID: 37378613 PMCID: PMC10308087 DOI: 10.1083/jcb.202209120] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 04/13/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Autonomous circadian clocks exist in nearly every mammalian cell type. These cellular clocks are subjected to a multilayered regulation sensitive to the mechanochemical cell microenvironment. Whereas the biochemical signaling that controls the cellular circadian clock is increasingly well understood, mechanisms underlying regulation by mechanical cues are largely unknown. Here we show that the fibroblast circadian clock is mechanically regulated through YAP/TAZ nuclear levels. We use high-throughput analysis of single-cell circadian rhythms and apply controlled mechanical, biochemical, and genetic perturbations to study the expression of the clock gene Rev-erbα. We observe that Rev-erbα circadian oscillations are disrupted with YAP/TAZ nuclear translocation. By targeted mutations and overexpression of YAP/TAZ, we show that this mechanobiological regulation, which also impacts core components of the clock such as Bmal1 and Cry1, depends on the binding of YAP/TAZ to the transcriptional effector TEAD. This mechanism could explain the impairment of circadian rhythms observed when YAP/TAZ activity is upregulated, as in cancer and aging.
Collapse
Affiliation(s)
- Juan F. Abenza
- Institute for Bioengineering of Catalonia, The Barcelona Institute for Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Barcelona, Spain
| | - Leone Rossetti
- Institute for Bioengineering of Catalonia, The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Malèke Mouelhi
- Institute for Bioengineering of Catalonia, The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Javier Burgués
- Institute for Bioengineering of Catalonia, The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Ion Andreu
- Institute for Bioengineering of Catalonia, The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Keith Kennedy
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia, The Barcelona Institute for Science and Technology, Barcelona, Spain
- Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Santiago Marco
- Institute for Bioengineering of Catalonia, The Barcelona Institute for Science and Technology, Barcelona, Spain
- Department of Electronics and Biomedical Engineering, Universitat de Barcelona, Barcelona, Spain
| | - Jordi García-Ojalvo
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia, The Barcelona Institute for Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Barcelona, Spain
- Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| |
Collapse
|
5
|
Kawamata Y, Ryu KA, Hermann GN, Sandahl A, Vantourout JC, Olow AK, Adams LTA, Rivera-Chao E, Roberts LR, Gnaim S, Nassir M, Oslund RC, Fadeyi OO, Baran PS. An electroaffinity labelling platform for chemoproteomic-based target identification. Nat Chem 2023; 15:1267-1275. [PMID: 37322100 DOI: 10.1038/s41557-023-01240-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/12/2023] [Indexed: 06/17/2023]
Abstract
Target identification involves deconvoluting the protein target of a pharmacologically active, small-molecule ligand, a process that is critical for early drug discovery yet technically challenging. Photoaffinity labelling strategies have become the benchmark for small-molecule target deconvolution, but covalent protein capture requires the use of high-energy ultraviolet light, which can complicate downstream target identification. Thus, there is a strong demand for alternative technologies that allow for controlled activation of chemical probes to covalently label their protein target. Here we introduce an electroaffinity labelling platform that leverages the use of a small, redox-active diazetidinone functional group to enable chemoproteomic-based target identification of pharmacophores within live cell environments. The underlying discovery to enable this platform is that the diazetidinone can be electrochemically oxidized to reveal a reactive intermediate useful for covalent modification of proteins. This work demonstrates the electrochemical platform to be a functional tool for drug-target identification.
Collapse
Affiliation(s)
- Yu Kawamata
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Keun Ah Ryu
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, USA
| | - Gary N Hermann
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Alexander Sandahl
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Aleksandra K Olow
- Genetics and Pharmacogenomics, Merck & Co., Inc., San Francisco, CA, USA
| | | | - Eva Rivera-Chao
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Lee R Roberts
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, USA
| | - Samer Gnaim
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Molhm Nassir
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Rob C Oslund
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, USA.
- InduPro Therapeutics, Cambridge, MA, USA.
| | - Olugbeminiyi O Fadeyi
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, USA.
- InduPro Therapeutics, Cambridge, MA, USA.
| | - Phil S Baran
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
6
|
Gomatou G, Karachaliou A, Veloudiou OZ, Karvela A, Syrigos N, Kotteas E. The Role of REV-ERB Receptors in Cancer Pathogenesis. Int J Mol Sci 2023; 24:ijms24108980. [PMID: 37240325 DOI: 10.3390/ijms24108980] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
REV-ERB receptors are members of the nuclear receptor superfamily of proteins, which act as both intracellular receptors and transcription factors, therefore modulating the expression of target genes. REV-ERBs act as transcription repressors because of their unique structure. Their predominant role involves the control of peripheral circadian rhythmicity by participating in a transcription-translation feedback loop with other major clock genes. Regarding their role in cancer pathogenesis, recent studies in various cancerous tissues have revealed that their expression was downregulated in the majority of the cases. Dysregulation of their expression was also implicated in cancer-associated cachexia. The pharmacological restoration of their effects is feasible with synthetic agonists, which have been explored in preclinical studies but with scarce data. There is a need for further investigation, primarily with mechanistic studies, on the effect of the REV-ERB-induced circadian rhythm deregulation in carcinogenesis and cancer-related systemic effects, such as cachexia, in order to address the potential of relevant therapeutic implications.
Collapse
Affiliation(s)
- Georgia Gomatou
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Anastasia Karachaliou
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Orsalia-Zoi Veloudiou
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Alexandra Karvela
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Nikolaos Syrigos
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Elias Kotteas
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| |
Collapse
|
7
|
Adlanmerini M, Lazar MA. The REV-ERB Nuclear Receptors: Timekeepers for the Core Clock Period and Metabolism. Endocrinology 2023; 164:bqad069. [PMID: 37149727 PMCID: PMC10413432 DOI: 10.1210/endocr/bqad069] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/20/2023] [Accepted: 05/03/2023] [Indexed: 05/08/2023]
Abstract
REV-ERB nuclear receptors are potent transcriptional repressors that play an important role in the core mammalian molecular clock and metabolism. Deletion of both REV-ERBα and its largely redundant isoform REV-ERBβ in a murine tissue-specific manner have shed light on their specific functions in clock mechanisms and circadian metabolism. This review highlights recent findings that establish REV-ERBs as crucial circadian timekeepers in a variety of tissues, regulating overlapping and distinct processes that maintain normal physiology and protect from metabolic dysfunction.
Collapse
Affiliation(s)
- Marine Adlanmerini
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, University of Toulouse 3, Toulouse, France
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
8
|
Albrecht U. The circadian system and mood related behavior in mice. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 137:269-291. [PMID: 37709379 DOI: 10.1016/bs.apcsb.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Most organisms on earth have evolved an internal clock in order to predict daily recurring events. This clock called circadian clock has a period of about 24 h and allows organisms to organize biochemical and physiological processes over one day. Changes in lighting conditions as they occur naturally over seasons, or man made by jet lag or shift work, advance or delay clock phase in order to synchronize an organism's physiology to the environment. A misalignment of the clock to its environment results in sleep disturbances and mood disorders. Although there are strong associations between the circadian clock and mood disorders such as depression, the underlying molecular mechanisms are not well understood. This review describes the currently known molecular links between circadian clock components and mood related behaviors in mice, which will help to understand the causal links between the clock and mood in humans in the future.
Collapse
Affiliation(s)
- U Albrecht
- Department of Biology, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
9
|
Rasmussen ES, Takahashi JS, Green CB. Time to target the circadian clock for drug discovery. Trends Biochem Sci 2022; 47:745-758. [PMID: 35577675 PMCID: PMC9378619 DOI: 10.1016/j.tibs.2022.04.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/01/2022] [Accepted: 04/18/2022] [Indexed: 11/30/2022]
Abstract
The circadian clock is an intracellular timekeeping device that drives daily rhythms in diverse and extensive processes throughout the body. The clock mechanism comprises a core transcription/translation negative feedback loop that is modulated by a complex set of additional interlocking feedback loops. Pharmacological manipulation of the clock may be valuable for treating many maladies including jet lag, shift work and related sleep disorders, various metabolic diseases, and cancer. We review recent identification of small-molecule clock modulators and discuss the biochemical features of the core clock that may be amenable to future drug discovery.
Collapse
Affiliation(s)
- Emil Sjulstok Rasmussen
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph S Takahashi
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Carla B Green
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
10
|
Zheng J, Zhang L, Tan Z, Zhao Q, Wei X, Yang Y, Li R. Bmal1- and Per2-mediated regulation of the osteogenic differentiation and proliferation of mouse bone marrow mesenchymal stem cells by modulating the Wnt/β-catenin pathway. Mol Biol Rep 2022; 49:4485-4501. [PMID: 35386071 DOI: 10.1007/s11033-022-07292-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/22/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Bmal1 and Per2 are the core components of the circadian clock genes (CCGs). Bmal1-/- mice exhibit premature aging, as indicated by hypotrichosis and osteoporosis, with a loss of proliferation ability. The same occurs in Per2-/- mice, albeit to a less severe degree. However, whether the effects of Bmal1 and Per2 on proliferation and osteogenic differentiation are synergistic or antagonistic remains unclear. Thus, our study aimed to explore the effects and specific mechanism. METHODS AND RESULTS Lentiviral and adenoviral vectors were constructed to silence or overexpress Bmal1 or Per2 and MTT, flow cytometry, RT-qPCR, WB, immunohistochemistry, alizarin red staining and ChIP-Seq analyses were applied to identify the possible mechanism. The successful knockdown and overexpression of Bmal1/Per2 were detected by fluorescence microcopy. Flow cytometry found out that Bmal1 or Per2 knockdown resulted in G1-phase cell cycle arrest. RT-qPCR showed the different expression levels of Wnt-3a, c-myc1 and axin2 in the Wnt/β-catenin signaling pathway as well as the gene expression change of Rorα and Rev-erbα. Meanwhile, related proteins such as β-catenin, TCF-1, and P-GSK-3β were detected. ALP activity and the amount of mineral nodules were compared. ChIP-Seq results showed the possible mechanism. CONCLUSIONS Bmal1 and Per2, as primary canonical clock genes, showed synergistic effects on the proliferation and differentiation of BMSCs. They would inhibit the Wnt/β-catenin signaling pathway by downregulating Rorα expression or upregulating Rev-erbα expression, both of which were also key elements of CCGs. And this may be the mechanism by which they negatively regulate the osteogenic differentiation of BMSCs. Bmal1 and Per2 show synergistic effects in the proliferation of BMSCs. In addition, they play a synergistic role in negatively regulating the osteogenic differentiation ability of BMSCs. Bmal1 and Per2 may regulate the aging of BMSCs by altering cell proliferation and osteogenic differentiation through Rorα and Rev-erbα to affect Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Jiawen Zheng
- Orthodontic Centre, West China College of Stomatology, Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | - Lanxin Zhang
- Orthodontic Centre, West China College of Stomatology, Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | - Zhen Tan
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China.
- Oral Implant Centre, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Qing Zhao
- Orthodontic Centre, West China College of Stomatology, Sichuan University, Chengdu, China.
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China.
| | - Xiaoyu Wei
- Orthodontic Centre, West China College of Stomatology, Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | - Yuqing Yang
- Orthodontic Centre, West China College of Stomatology, Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | - Rong Li
- Orthodontic Centre, West China College of Stomatology, Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Liu H, Liu Y, Hai R, Liao W, Luo X. The role of circadian clocks in cancer: Mechanisms and clinical implications. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
12
|
Griffett K, Hayes ME, Boeckman MP, Burris TP. The role of REV-ERB in NASH. Acta Pharmacol Sin 2022; 43:1133-1140. [PMID: 35217816 PMCID: PMC9061770 DOI: 10.1038/s41401-022-00883-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/28/2022] [Indexed: 02/07/2023] Open
Abstract
REV-ERBs are atypical nuclear receptors as they function as ligand-regulated transcriptional repressors. The natural ligand for the REV-ERBs (REV-ERBα and REV-ERBβ) is heme, and heme-binding results in recruitment of transcriptional corepressor proteins such as N-CoR that mediates repression of REV-ERB target genes. These two receptors regulate a large range of physiological processes including several important in the pathophysiology of non-alcoholic steatohepatitis (NASH). These include carbohydrate and lipid metabolism as well as inflammatory pathways. A number of synthetic REV-ERB agonists have been developed as chemical tools and they show efficacy in animal models of NASH. Here, we will review the functions of REV-ERB with regard to their relevance to NASH as well as the potential to target REV-ERB for treatment of this disease.
Collapse
Affiliation(s)
- Kristine Griffett
- Center for Clinical Pharmacology, Washington University in St. Louis and University of Health Sciences & Pharmacy, St. Louis, MO, 63110, USA
| | - Matthew E Hayes
- University of Florida Genetics Institute, Gainesville, FL, 32610, USA
| | - Michael P Boeckman
- Center for Clinical Pharmacology, Washington University in St. Louis and University of Health Sciences & Pharmacy, St. Louis, MO, 63110, USA
| | - Thomas P Burris
- University of Florida Genetics Institute, Gainesville, FL, 32610, USA.
| |
Collapse
|
13
|
Moravcová S, Spišská V, Pačesová D, Hrubcová L, Kubištová A, Novotný J, Bendová Z. Circadian control of kynurenine pathway enzymes in the rat pineal gland, liver, and heart and tissue- and enzyme-specific responses to lipopolysaccharide. Arch Biochem Biophys 2022; 722:109213. [DOI: 10.1016/j.abb.2022.109213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/10/2022] [Accepted: 04/07/2022] [Indexed: 11/26/2022]
|
14
|
Zhou L, Zhang Z, Nice E, Huang C, Zhang W, Tang Y. Circadian rhythms and cancers: the intrinsic links and therapeutic potentials. J Hematol Oncol 2022; 15:21. [PMID: 35246220 PMCID: PMC8896306 DOI: 10.1186/s13045-022-01238-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
The circadian rhythm is an evolutionarily conserved time-keeping system that comprises a wide variety of processes including sleep-wake cycles, eating-fasting cycles, and activity-rest cycles, coordinating the behavior and physiology of all organs for whole-body homeostasis. Acute disruption of circadian rhythm may lead to transient discomfort, whereas long-term irregular circadian rhythm will result in the dysfunction of the organism, therefore increasing the risks of numerous diseases especially cancers. Indeed, both epidemiological and experimental evidence has demonstrated the intrinsic link between dysregulated circadian rhythm and cancer. Accordingly, a rapidly increasing understanding of the molecular mechanisms of circadian rhythms is opening new options for cancer therapy, possibly by modulating the circadian clock. In this review, we first describe the general regulators of circadian rhythms and their functions on cancer. In addition, we provide insights into the mechanisms underlying how several types of disruption of the circadian rhythm (including sleep-wake, eating-fasting, and activity-rest) can drive cancer progression, which may expand our understanding of cancer development from the clock perspective. Moreover, we also summarize the potential applications of modulating circadian rhythms for cancer treatment, which may provide an optional therapeutic strategy for cancer patients.
Collapse
Affiliation(s)
- Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Edouard Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Wei Zhang
- Mental Health Center and Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yong Tang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Acupuncture and Chronobiology Laboratory of Sichuan Province, Chengdu, 610075, China.
| |
Collapse
|
15
|
Wang XL, Li L. Circadian Clock Regulates Inflammation and the Development of Neurodegeneration. Front Cell Infect Microbiol 2021; 11:696554. [PMID: 34595127 PMCID: PMC8476957 DOI: 10.3389/fcimb.2021.696554] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022] Open
Abstract
The circadian clock regulates numerous key physiological processes and maintains cellular, tissue, and systemic homeostasis. Disruption of circadian clock machinery influences key activities involved in immune response and brain function. Moreover, Immune activation has been closely linked to neurodegeneration. Here, we review the molecular clock machinery and the diurnal variation of immune activity. We summarize the circadian control of immunity in both central and peripheral immune cells, as well as the circadian regulation of brain cells that are implicated in neurodegeneration. We explore the important role of systemic inflammation on neurodegeneration. The circadian clock modulates cellular metabolism, which could be a mechanism underlying circadian control. We also discuss the circadian interventions implicated in inflammation and neurodegeneration. Targeting circadian clocks could be a potential strategy for the prevention and treatment of inflammation and neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiao-Lan Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lianjian Li
- Department of Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
16
|
Willems S, Zaienne D, Merk D. Targeting Nuclear Receptors in Neurodegeneration and Neuroinflammation. J Med Chem 2021; 64:9592-9638. [PMID: 34251209 DOI: 10.1021/acs.jmedchem.1c00186] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nuclear receptors, also known as ligand-activated transcription factors, regulate gene expression upon ligand signals and present as attractive therapeutic targets especially in chronic diseases. Despite the therapeutic relevance of some nuclear receptors in various pathologies, their potential in neurodegeneration and neuroinflammation is insufficiently established. This perspective gathers preclinical and clinical data for a potential role of individual nuclear receptors as future targets in Alzheimer's disease, Parkinson's disease, and multiple sclerosis, and concomitantly evaluates the level of medicinal chemistry targeting these proteins. Considerable evidence suggests the high promise of ligand-activated transcription factors to counteract neurodegenerative diseases with a particularly high potential of several orphan nuclear receptors. However, potent tools are lacking for orphan receptors, and limited central nervous system exposure or insufficient selectivity also compromises the suitability of well-studied nuclear receptor ligands for functional studies. Medicinal chemistry efforts are needed to develop dedicated high-quality tool compounds for the therapeutic validation of nuclear receptors in neurodegenerative pathologies.
Collapse
Affiliation(s)
- Sabine Willems
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| | - Daniel Zaienne
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| |
Collapse
|