1
|
Almarza C, Villalobos-Nova K, Toro MA, González M, Niechi I, Brown-Brown DA, López-Muñoz RA, Silva-Pavez E, Gaete-Ramírez B, Varas-Godoy M, Burzio VA, Jara L, Aguayo F, Tapia JC. Cisplatin-resistance and aggressiveness are enhanced by a highly stable endothelin-converting enzyme-1c in lung cancer cells. Biol Res 2024; 57:74. [PMID: 39443981 PMCID: PMC11515556 DOI: 10.1186/s40659-024-00551-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Lung cancer constitutes the leading cause of cancer mortality. High levels of endothelin-1 (ET-1), its cognate receptor ETAR and its activating enzyme, the endothelin-converting enzyme-1 (ECE-1), have been reported in several cancer types, including lung cancer. ECE-1 comprises four isoforms, which only differ in their cytoplasmic N-terminus. Protein kinase CK2 phosphorylates the N-terminus of isoform ECE-1c, increasing its stability and leading to enhanced invasiveness in glioblastoma and colorectal cancer cells, which is believed to be mediated by the amino acid residue Lys-6, a conserved putative ubiquitination site neighboring the CK2-phosphorylated residues Ser-18 and Ser-20. Whether Lys-6 is linked to the acquisition of a cancer stem cell (CSC)-like phenotype and aggressiveness in human non-small cell lung cancer (NSCLC) cells has not been studied. METHODS In order to establish the role of Lys-6 in the stability of ECE-1c and its involvement in lung cancer aggressiveness, we mutated this residue to a non-ubiquitinable arginine and constitutively expressed the wild-type (ECE-1cWT) and mutant (ECE-1cK6R) proteins in A549 and H1299 human NSCLC cells by lentiviral transduction. We determined the protein stability of these clones alone or in the presence of the CK2 inhibitor silmitasertib, compared to ECE-1cWT and mock-transduced cells. In addition, the concentration of secreted ET-1 in the growth media was determined by ELISA. Expression of stemness genes were determined by Western blot and RT-qPCR. Chemoresistance to cisplatin was studied by MTS viability assay. Migration and invasion were measured through transwell and Matrigel assays, respectively, and the side-population was determined using flow cytometry. RESULTS ECE-1cK6R displayed higher stability in NSCLC cells compared to ECE-1cWT-expressing cells, but ET-1 secreted levels showed no difference up to 48 h. Most importantly, ECE-1cK6R promoted expression of the stemness genes c-Myc, Sox-2, Oct-4, CD44 and CD133, which enhance cellular self-renewal capability. Also, the ECE-1cK6R-expressing cells showed higher cisplatin chemoresistance, correlating with an augmented side-population abundance due to the increased expression of the ABCG2 efflux pump. Finally, the ECE-1cK6R-expressing cells showed enhanced invasiveness, which correlated with the regulated expression of known EMT markers. CONCLUSIONS Our findings suggest an important role of ECE-1c in lung cancer. ECE-1c is key in a non-canonical ET-1-independent mechanism which triggers a CSC-like phenotype, leading to enhanced lung cancer aggressiveness. Underlying this mechanism, ECE-1c is stabilized upon phosphorylation by CK2, which is upregulated in many cancers. Thus, phospho-ECE-1c may be considered as a novel prognostic biomarker of recurrence, as well as the CK2 inhibitor silmitasertib as a potential therapy for lung cancer patients.
Collapse
Affiliation(s)
- Cristopher Almarza
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Karla Villalobos-Nova
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - María A Toro
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Manuel González
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ignacio Niechi
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - David A Brown-Brown
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Rodrigo A López-Muñoz
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Eduardo Silva-Pavez
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Santiago, Chile
| | - Belén Gaete-Ramírez
- Centro de Biología Celular y Biomedicina, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Manuel Varas-Godoy
- Centro de Biología Celular y Biomedicina, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Verónica A Burzio
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
| | - Lilian Jara
- Programa de Genética, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Francisco Aguayo
- Departamento de Biomedicina, Facultad de Medicina, Universidad de Tarapacá, Arica, Chile
| | - Julio C Tapia
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
- Laboratorio de Transformación Celular, Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Santiago, 8380453, Chile.
| |
Collapse
|
2
|
Roychowdhury T, McNutt SW, Pasala C, Nguyen HT, Thornton DT, Sharma S, Botticelli L, Digwal CS, Joshi S, Yang N, Panchal P, Chakrabarty S, Bay S, Markov V, Kwong C, Lisanti J, Chung SY, Ginsberg SD, Yan P, De Stanchina E, Corben A, Modi S, Alpaugh ML, Colombo G, Erdjument-Bromage H, Neubert TA, Chalkley RJ, Baker PR, Burlingame AL, Rodina A, Chiosis G, Chu F. Phosphorylation-driven epichaperome assembly is a regulator of cellular adaptability and proliferation. Nat Commun 2024; 15:8912. [PMID: 39414766 PMCID: PMC11484706 DOI: 10.1038/s41467-024-53178-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 10/04/2024] [Indexed: 10/18/2024] Open
Abstract
The intricate network of protein-chaperone interactions is crucial for maintaining cellular function. Recent discoveries have unveiled the existence of specialized chaperone assemblies, known as epichaperomes, which serve as scaffolding platforms that orchestrate the reconfiguration of protein-protein interaction networks, thereby enhancing cellular adaptability and proliferation. This study explores the structural and regulatory aspects of epichaperomes, with a particular focus on the role of post-translational modifications (PTMs) in their formation and function. A key finding is the identification of specific PTMs on HSP90, particularly at residues Ser226 and Ser255 within an intrinsically disordered region, as critical determinants of epichaperome assembly. Our data demonstrate that phosphorylation of these serine residues enhances HSP90's interactions with other chaperones and co-chaperones, creating a microenvironment conducive to epichaperome formation. Moreover, we establish a direct link between epichaperome function and cellular physiology, particularly in contexts where robust proliferation and adaptive behavior are essential, such as in cancer and pluripotent stem cell maintenance. These findings not only provide mechanistic insights but also hold promise for the development of novel therapeutic strategies targeting chaperone assemblies in diseases characterized by epichaperome dysregulation, thereby bridging the gap between fundamental research and precision medicine.
Collapse
Affiliation(s)
- Tanaya Roychowdhury
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Seth W McNutt
- Department of Molecular, Cellular & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Chiranjeevi Pasala
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hieu T Nguyen
- Department of Molecular, Cellular & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Daniel T Thornton
- Department of Molecular, Cellular & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Sahil Sharma
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Luke Botticelli
- Department of Molecular, Cellular & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Chander S Digwal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Suhasini Joshi
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nan Yang
- Department of Molecular, Cellular & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Palak Panchal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Souparna Chakrabarty
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sadik Bay
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vladimir Markov
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charlene Kwong
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeanine Lisanti
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sun Young Chung
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stephen D Ginsberg
- Departments of Psychiatry, Neuroscience & Physiology & the NYU Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
| | - Pengrong Yan
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elisa De Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adriana Corben
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Maimonides Medical Center, Brooklyn, NY, USA
| | - Shanu Modi
- Department of Medicine, Division of Solid Tumors, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mary L Alpaugh
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Rowan University, Glassboro, NJ, USA
| | | | - Hediye Erdjument-Bromage
- Department of Neuroscience and Physiology and Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| | - Thomas A Neubert
- Department of Neuroscience and Physiology and Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| | - Robert J Chalkley
- Mass Spectrometry Facility, University of California, San Francisco, CA, USA
| | - Peter R Baker
- Mass Spectrometry Facility, University of California, San Francisco, CA, USA
| | - Alma L Burlingame
- Mass Spectrometry Facility, University of California, San Francisco, CA, USA
| | - Anna Rodina
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Division of Solid Tumors, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Feixia Chu
- Department of Molecular, Cellular & Biomedical Sciences, University of New Hampshire, Durham, NH, USA.
- Hubbard Center for Genome Studies, University of New Hampshire, Durham, NH, USA.
| |
Collapse
|
3
|
McNutt SW, Roychowdhury T, Pasala C, Nguyen HT, Thornton DT, Sharma S, Botticelli L, Digwal CS, Joshi S, Yang N, Panchal P, Chakrabarty S, Bay S, Markov V, Kwong C, Lisanti J, Chung SY, Ginsberg SD, Yan P, DeStanchina E, Corben A, Modi S, Alpaugh M, Colombo G, Erdjument-Bromage H, Neubert TA, Chalkley RJ, Baker PR, Burlingame AL, Rodina A, Chiosis G, Chu F. Phosphorylation-Driven Epichaperome Assembly: A Critical Regulator of Cellular Adaptability and Proliferation. RESEARCH SQUARE 2024:rs.3.rs-4114038. [PMID: 38645031 PMCID: PMC11030525 DOI: 10.21203/rs.3.rs-4114038/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The intricate protein-chaperone network is vital for cellular function. Recent discoveries have unveiled the existence of specialized chaperone complexes called epichaperomes, protein assemblies orchestrating the reconfiguration of protein-protein interaction networks, enhancing cellular adaptability and proliferation. This study delves into the structural and regulatory aspects of epichaperomes, with a particular emphasis on the significance of post-translational modifications in shaping their formation and function. A central finding of this investigation is the identification of specific PTMs on HSP90, particularly at residues Ser226 and Ser255 situated within an intrinsically disordered region, as critical determinants in epichaperome assembly. Our data demonstrate that the phosphorylation of these serine residues enhances HSP90's interaction with other chaperones and co-chaperones, creating a microenvironment conducive to epichaperome formation. Furthermore, this study establishes a direct link between epichaperome function and cellular physiology, especially in contexts where robust proliferation and adaptive behavior are essential, such as cancer and stem cell maintenance. These findings not only provide mechanistic insights but also hold promise for the development of novel therapeutic strategies targeting chaperone complexes in diseases characterized by epichaperome dysregulation, bridging the gap between fundamental research and precision medicine.
Collapse
Affiliation(s)
- Seth W McNutt
- Department of Molecular, Cellular & Biomedical Sciences, University of New Hampshire, Durham, NH 03824, USA
- co-first author, equally contributed to the work
| | - Tanaya Roychowdhury
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- co-first author, equally contributed to the work
| | - Chiranjeevi Pasala
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hieu T Nguyen
- Department of Molecular, Cellular & Biomedical Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Daniel T Thornton
- Department of Molecular, Cellular & Biomedical Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Sahil Sharma
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Luke Botticelli
- Department of Molecular, Cellular & Biomedical Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Chander S Digwal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Suhasini Joshi
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nan Yang
- Department of Molecular, Cellular & Biomedical Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Palak Panchal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Souparna Chakrabarty
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sadik Bay
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vladimir Markov
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Charlene Kwong
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jeanine Lisanti
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sun Young Chung
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephen D Ginsberg
- Departments of Psychiatry, Neuroscience & Physiology & the NYU Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
| | - Pengrong Yan
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elisa DeStanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Adriana Corben
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Shanu Modi
- Department of Medicine, Division of Solid Tumors, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mary Alpaugh
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Giorgio Colombo
- Department of Chemistry, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Hediye Erdjument-Bromage
- Department of Neuroscience and Physiology and Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Thomas A Neubert
- Department of Neuroscience and Physiology and Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Robert J Chalkley
- Mass Spectrometry Facility, University of California, San Francisco, California 94143, USA
| | - Peter R Baker
- Mass Spectrometry Facility, University of California, San Francisco, California 94143, USA
| | - Alma L Burlingame
- Mass Spectrometry Facility, University of California, San Francisco, California 94143, USA
| | - Anna Rodina
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medicine, Division of Solid Tumors, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- These authors jointly supervised this work: Feixia Chu, Gabriela Chiosis
| | - Feixia Chu
- Department of Molecular, Cellular & Biomedical Sciences, University of New Hampshire, Durham, NH 03824, USA
- Hubbard Center for Genome Studies, University of New Hampshire, Durham, NH 03824, USA
- These authors jointly supervised this work: Feixia Chu, Gabriela Chiosis
| |
Collapse
|
4
|
Florke Gee RR, Huber AD, Chen T. Regulation of PXR in drug metabolism: chemical and structural perspectives. Expert Opin Drug Metab Toxicol 2024; 20:9-23. [PMID: 38251638 PMCID: PMC10939797 DOI: 10.1080/17425255.2024.2309212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/19/2024] [Indexed: 01/23/2024]
Abstract
INTRODUCTION Pregnane X receptor (PXR) is a master xenobiotic sensor that transcriptionally controls drug metabolism and disposition pathways. PXR activation by pharmaceutical drugs, natural products, environmental toxins, etc. may decrease drug efficacy and increase drug-drug interactions and drug toxicity, indicating a therapeutic value for PXR antagonists. However, PXR's functions in physiological events, such as intestinal inflammation, indicate that PXR activators may be useful in certain disease contexts. AREAS COVERED We review the reported roles of PXR in various physiological and pathological processes including drug metabolism, cancer, inflammation, energy metabolism, and endobiotic homeostasis. We then highlight specific cellular and chemical routes that modulate PXR activity and discuss the functional consequences. Databases searched and inclusive dates: PubMed, 1 January 1980 to 10 January 2024. EXPERT OPINION Knowledge of PXR's drug metabolism function has helped drug developers produce small molecules without PXR-mediated metabolic liabilities, and further understanding of PXR's cellular functions may offer drug development opportunities in multiple disease settings.
Collapse
Affiliation(s)
- Rebecca R. Florke Gee
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Andrew D. Huber
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
5
|
Embryonic organizer formation disorder leads to multiorgan dysplasia in Down syndrome. Cell Death Dis 2022; 13:1054. [PMID: 36535930 PMCID: PMC9763398 DOI: 10.1038/s41419-022-05517-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Despite the high prevalence of Down syndrome (DS) and early identification of the cause (trisomy 21), its molecular pathogenesis has been poorly understood and specific treatments have consequently been practically unavailable. A number of medical conditions throughout the body associated with DS have prompted us to investigate its molecular etiology from the viewpoint of the embryonic organizer, which can steer the development of surrounding cells into specific organs and tissues. We established a DS zebrafish model by overexpressing the human DYRK1A gene, a highly haploinsufficient gene located at the "critical region" within 21q22. We found that both embryonic organizer and body axis were significantly impaired during early embryogenesis, producing abnormalities of the nervous, heart, visceral, and blood systems, similar to those observed with DS. Quantitative phosphoproteome analysis and related assays demonstrated that the DYRK1A-overexpressed zebrafish embryos had anomalous phosphorylation of β-catenin and Hsp90ab1, resulting in Wnt signaling enhancement and TGF-β inhibition. We found an uncovered ectopic molecular mechanism present in amniocytes from fetuses diagnosed with DS and isolated hematopoietic stem cells (HSCs) of DS patients. Importantly, the abnormal proliferation of DS HSCs could be recovered by switching the balance between Wnt and TGF-β signaling in vitro. Our findings provide a novel molecular pathogenic mechanism in which ectopic Wnt and TGF-β lead to DS physical dysplasia, suggesting potential targeted therapies for DS.
Collapse
|
6
|
Maiti S, Picard D. Cytosolic Hsp90 Isoform-Specific Functions and Clinical Significance. Biomolecules 2022; 12:1166. [PMID: 36139005 PMCID: PMC9496497 DOI: 10.3390/biom12091166] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/17/2022] Open
Abstract
The heat shock protein 90 (Hsp90) is a molecular chaperone and a key regulator of proteostasis under both physiological and stress conditions. In mammals, there are two cytosolic Hsp90 isoforms: Hsp90α and Hsp90β. These two isoforms are 85% identical and encoded by two different genes. Hsp90β is constitutively expressed and essential for early mouse development, while Hsp90α is stress-inducible and not necessary for survivability. These two isoforms are known to have largely overlapping functions and to interact with a large fraction of the proteome. To what extent there are isoform-specific functions at the protein level has only relatively recently begun to emerge. There are studies indicating that one isoform is more involved in the functionality of a specific tissue or cell type. Moreover, in many diseases, functionally altered cells appear to be more dependent on one particular isoform. This leaves space for designing therapeutic strategies in an isoform-specific way, which may overcome the unfavorable outcome of pan-Hsp90 inhibition encountered in previous clinical trials. For this to succeed, isoform-specific functions must be understood in more detail. In this review, we summarize the available information on isoform-specific functions of mammalian Hsp90 and connect it to possible clinical applications.
Collapse
Affiliation(s)
| | - Didier Picard
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, CH-1211 Geneve, Switzerland
| |
Collapse
|
7
|
BRCA1 mutations in high-grade serous ovarian cancer are associated with proteomic changes in DNA repair, splicing, transcription regulation and signaling. Sci Rep 2022; 12:4445. [PMID: 35292711 PMCID: PMC8924168 DOI: 10.1038/s41598-022-08461-0] [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: 05/10/2021] [Accepted: 02/23/2022] [Indexed: 11/08/2022] Open
Abstract
Despite recent advances in the management of BRCA1 mutated high-grade serous ovarian cancer (HGSC), the physiology of these tumors remains poorly understood. Here we provide a comprehensive molecular understanding of the signaling processes that drive HGSC pathogenesis with the addition of valuable ubiquitination profiling, and their dependency on BRCA1 mutation-state directly in patient-derived tissues. Using a multilayered proteomic approach, we show the tight coordination between the ubiquitination and phosphorylation regulatory layers and their role in key cellular processes related to BRCA1-dependent HGSC pathogenesis. In addition, we identify key bridging proteins, kinase activity, and post-translational modifications responsible for molding distinct cancer phenotypes, thus providing new opportunities for therapeutic intervention, and ultimately advance towards a more personalized patient care.
Collapse
|
8
|
Qin M, Xin Y, Bian Y, Yang X, Xi T, Xiong J. Phosphorylation-Induced Ubiquitination and Degradation of PXR through CDK2-TRIM21 Axis. Cells 2022; 11:cells11020264. [PMID: 35053380 PMCID: PMC8773821 DOI: 10.3390/cells11020264] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/22/2021] [Accepted: 01/06/2022] [Indexed: 02/07/2023] Open
Abstract
Pregnane X receptor (PXR) is a member of the nuclear receptor superfamily that is activated by a variety of endogenous metabolites or xenobiotics. Its downstream target genes are involved in metabolism, inflammation and processes closely related to cancer. However, the stability regulation of PXR protein resulting from post-translational modification is still largely undefined. In the present study, primary mouse hepatocytes, hepatoma HepG2 cells and HEK 293T cells were used to investigate gene expression and protein interactions. The role of kinases was evaluated by RNA interference and overexpression constructs with or without PXR phosphorylation site mutations. The activity of CYP3A4 and P-gp was determined by enzymatic and substrate accumulation assays. It was found that E3 ubiquitin ligase TRIM21 mediates the ubiquitination and degradation of PXR and plays an important role in regulating the activity of PXR. On this basis, PXR phosphorylation-associated kinases were evaluated regarding regulation of the stability of PXR. We found cyclin dependent kinase 2 (CDK2) exclusively phosphorylates PXR at Ser350, promotes its disassociation with Hsp90/DNAJC7, and leads to subsequent TRIM21-mediated PXR ubiquitination and degradation. As well-known CDK inhibitors, dinaciclib and kenpaullone stabilize PXR and result in elevated expression and activity of PXR-targeted DMETs, including carboxylesterases, CYP3A4 and P-gp. The suppressed degradation of PXR by CDK2 inhibitors denotes dinaciclib-induced promotion of PXR-targeted genes. The findings of CDK2-mediated PXR degradation indicate a wide range of potential drug–drug interactions during clinical cancer therapy using CDK inhibitors and imply an alternative direction for the development of novel PXR antagonists.
Collapse
Affiliation(s)
- Mengyao Qin
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (M.Q.); (Y.X.); (X.Y.)
| | - Yu Xin
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (M.Q.); (Y.X.); (X.Y.)
| | - Yong Bian
- Laboratory Animal Center, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Xuan Yang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (M.Q.); (Y.X.); (X.Y.)
| | - Tao Xi
- Research Center of Biotechnology, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China;
| | - Jing Xiong
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (M.Q.); (Y.X.); (X.Y.)
- Correspondence:
| |
Collapse
|
9
|
Weidenauer L, Quadroni M. Phosphorylation in the Charged Linker Modulates Interactions and Secretion of Hsp90β. Cells 2021; 10:cells10071701. [PMID: 34359868 PMCID: PMC8304327 DOI: 10.3390/cells10071701] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 12/16/2022] Open
Abstract
Hsp90β is a major chaperone involved in numerous cellular processes. Hundreds of client proteins depend on Hsp90β for proper folding and/or activity. Regulation of Hsp90β is critical to coordinate its tasks and is mediated by several post-translational modifications. Here, we focus on two phosphorylation sites located in the charged linker region of human Hsp90β, Ser226 and Ser255, which have been frequently reported but whose function remains unclear. Targeted measurements by mass spectrometry indicated that intracellular Hsp90β is highly phosphorylated on both sites (>90%). The level of phosphorylation was unaffected by various stresses (e.g., heat shock, inhibition with drugs) that impact Hsp90β activity. Mutating the two serines to alanines increased the amount of proteins interacting with Hsp90β globally and increased the sensitivity to tryptic cleavage in the C-terminal domain. Further investigation revealed that phosphorylation on Ser255 and to a lesser extent on Ser226 is decreased in the conditioned medium of cultured K562 cells, and that a non-phosphorylatable double alanine mutant was secreted more efficiently than the wild type. Overall, our results show that phosphorylation events in the charged linker regulate both the interactions of Hsp90β and its secretion, through changes in the conformation of the chaperone.
Collapse
|
10
|
Stuparu AD, Capri JR, Meyer CAL, Le TM, Evans-Axelsson SL, Current K, Lennox M, Mona CE, Fendler WP, Calais J, Eiber M, Dahlbom M, Czernin J, Radu CG, Lückerath K, Slavik R. Mechanisms of Resistance to Prostate-Specific Membrane Antigen-Targeted Radioligand Therapy in a Mouse Model of Prostate Cancer. J Nucl Med 2021; 62:989-995. [PMID: 33277393 PMCID: PMC8882874 DOI: 10.2967/jnumed.120.256263] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/11/2020] [Indexed: 01/19/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT) is effective against prostate cancer (PCa), but all patients relapse eventually. Poor understanding of the underlying resistance mechanisms represents a key barrier to development of more effective RLT. We investigate the proteome and phosphoproteome in a mouse model of PCa to identify signaling adaptations triggered by PSMA RLT. Methods: Therapeutic efficacy of PSMA RLT was assessed by tumor volume measurements, time to progression, and survival in C4-2 or C4-2 TP53-/- tumor-bearing nonobese diabetic scid γ-mice. Two days after RLT, the proteome and phosphoproteome were analyzed by mass spectrometry. Results: PSMA RLT significantly improved disease control in a dose-dependent manner. Proteome and phosphoproteome datasets revealed activation of genotoxic stress response pathways, including deregulation of DNA damage/replication stress response, TP53, androgen receptor, phosphatidylinositol-3-kinase/AKT, and MYC signaling. C4-2 TP53-/- tumors were less sensitive to PSMA RLT than were parental counterparts, supporting a role for TP53 in mediating RLT responsiveness. Conclusion: We identified signaling alterations that may mediate resistance to PSMA RLT in a PCa mouse model. Our data enable the development of rational synergistic RLT-combination therapies to improve outcomes for PCa patients.
Collapse
Affiliation(s)
| | - Joseph R Capri
- AstraZeneca, Chemical Biology Group, Waltham, Massachusetts
| | - Catherine A L Meyer
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Thuc M Le
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Susan L Evans-Axelsson
- Department of Translational Medicine, Division of Urological Cancers, Skåne University Hospital Malmö, Lund University, Lund, Sweden
| | - Kyle Current
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Mark Lennox
- School of Electronics, Electrical Engineering, and Computer Science, Queen's University Belfast, Belfast, United Kingdom
| | - Christine E Mona
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Department of Urology, Institute of Urologic Oncology, UCLA, Los Angeles, California; and
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium-University Hospital Essen, Essen, Germany
| | - Jeremie Calais
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Department of Urology, Institute of Urologic Oncology, UCLA, Los Angeles, California; and
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Matthias Eiber
- Clinic for Nuclear Medicine, Technical University Munich, Munich, Germany
| | - Magnus Dahlbom
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Johannes Czernin
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Department of Urology, Institute of Urologic Oncology, UCLA, Los Angeles, California; and
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Caius G Radu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Katharina Lückerath
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California;
- Department of Urology, Institute of Urologic Oncology, UCLA, Los Angeles, California; and
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| | - Roger Slavik
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
11
|
Dunphy K, Dowling P, Bazou D, O’Gorman P. Current Methods of Post-Translational Modification Analysis and Their Applications in Blood Cancers. Cancers (Basel) 2021; 13:1930. [PMID: 33923680 PMCID: PMC8072572 DOI: 10.3390/cancers13081930] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/04/2021] [Accepted: 04/14/2021] [Indexed: 12/12/2022] Open
Abstract
Post-translational modifications (PTMs) add a layer of complexity to the proteome through the addition of biochemical moieties to specific residues of proteins, altering their structure, function and/or localization. Mass spectrometry (MS)-based techniques are at the forefront of PTM analysis due to their ability to detect large numbers of modified proteins with a high level of sensitivity and specificity. The low stoichiometry of modified peptides means fractionation and enrichment techniques are often performed prior to MS to improve detection yields. Immuno-based techniques remain popular, with improvements in the quality of commercially available modification-specific antibodies facilitating the detection of modified proteins with high affinity. PTM-focused studies on blood cancers have provided information on altered cellular processes, including cell signaling, apoptosis and transcriptional regulation, that contribute to the malignant phenotype. Furthermore, the mechanism of action of many blood cancer therapies, such as kinase inhibitors, involves inhibiting or modulating protein modifications. Continued optimization of protocols and techniques for PTM analysis in blood cancer will undoubtedly lead to novel insights into mechanisms of malignant transformation, proliferation, and survival, in addition to the identification of novel biomarkers and therapeutic targets. This review discusses techniques used for PTM analysis and their applications in blood cancer research.
Collapse
Affiliation(s)
- Katie Dunphy
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Paul Dowling
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Despina Bazou
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland;
| | - Peter O’Gorman
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland;
| |
Collapse
|
12
|
Alhebbi H, Peer-Zada AA, Al-Hussaini AA, Algubaisi S, Albassami A, AlMasri N, Alrusayni Y, Alruzug IM, Alharby E, Samman MA, Ayoub SZ, Maddirevula S, Peake RWA, Alkuraya FS, Wali S, Almontashiri NAM. New paradigms of USP53 disease: normal GGT cholestasis, BRIC, cholangiopathy, and responsiveness to rifampicin. J Hum Genet 2020; 66:151-159. [PMID: 32759993 DOI: 10.1038/s10038-020-0811-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/15/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
Biallelic variants in the USP53 gene have recently been reported to segregate with normal gamma glutamyltransferase (GGT) cholestasis. Using whole-exome sequencing (WES), we detected two USP53 homozygous variants (c.951delT; p. Phe317fs and c.1744C>T; p. Arg582*) in five additional cases, including an unpublished cousin of a previously described family with intractable itching and normal GGT cholestasis. Three patients, a child and two adults, presented with recurrent episodes of normal GGT cholestasis, consistent with a diagnosis of benign recurrent intrahepatic cholestasis (BRIC). Cholangiopathic changes, possibly autoimmune in origin, were recognized in some patients. Additional phenotypic details in one patient included an enlarged left kidney, and speech/developmental delay. Notably, two patients exhibited a complete response to rifampicin, and one responded to ursodeoxycholic acid (UDCA). Two adult patients were suspected to have autoimmune liver disease and treated with steroids. This report describes new cases of USP53 disease presenting with normal GGT cholestasis or BRIC in three children and two adults. We also describe the novel finding of a dramatic response to rifampicin. The association of cholangiopathy with normal GGT cholestasis provides a diagnostic challenge and remains poorly understood.
Collapse
Affiliation(s)
- Hamoud Alhebbi
- Department of Pediatrics, Division of Gastroenterology and Hepatology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Abdul Ali Peer-Zada
- Molecular Pathology, Pathology and Clinical Laboratory Medicine Administration, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Abdulrahman A Al-Hussaini
- Pediatric Gastroenterology Division, Children's Specialized Hospital at King Fahad Medical City, Riyadh, Saudi Arabia.,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Sara Algubaisi
- Department of Pediatrics, Division of Gastroenterology and Hepatology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Awad Albassami
- Department of Pediatrics, Division of Gastroenterology and Hepatology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Nasser AlMasri
- Department of Adult Gastroenterology and Hepatology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Yasir Alrusayni
- Department of Pathology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Ibrahim M Alruzug
- Department of Adult Gastroenterology and Hepatology, King Saud Medical City, Riyadh, Saudi Arabia
| | - Essa Alharby
- Center for Genetics and Inherited Diseases, Taibah University, Almadinah Almunwarah, Saudi Arabia
| | - Manar A Samman
- Molecular Pathology, Pathology and Clinical Laboratory Medicine Administration, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Syed Zubair Ayoub
- Molecular Pathology, Pathology and Clinical Laboratory Medicine Administration, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Sateesh Maddirevula
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Roy W A Peake
- Department of Laboratory Medicine, Boston Children's Hospital, 300 Longwood Ave., Boston, MA, 02115, USA
| | - Fowzan S Alkuraya
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.,Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Sami Wali
- Department of Pediatrics, Division of Gastroenterology and Hepatology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia.
| | - Naif A M Almontashiri
- Center for Genetics and Inherited Diseases, Taibah University, Almadinah Almunwarah, Saudi Arabia. .,Faculty of Applied Medical Sciences, Taibah University, Almadinah Almunwarah, Saudi Arabia.
| |
Collapse
|
13
|
Backe SJ, Sager RA, Woodford MR, Makedon AM, Mollapour M. Post-translational modifications of Hsp90 and translating the chaperone code. J Biol Chem 2020; 295:11099-11117. [PMID: 32527727 DOI: 10.1074/jbc.rev120.011833] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
Cells have a remarkable ability to synthesize large amounts of protein in a very short period of time. Under these conditions, many hydrophobic surfaces on proteins may be transiently exposed, and the likelihood of deleterious interactions is quite high. To counter this threat to cell viability, molecular chaperones have evolved to help nascent polypeptides fold correctly and multimeric protein complexes assemble productively, while minimizing the danger of protein aggregation. Heat shock protein 90 (Hsp90) is an evolutionarily conserved molecular chaperone that is involved in the stability and activation of at least 300 proteins, also known as clients, under normal cellular conditions. The Hsp90 clients participate in the full breadth of cellular processes, including cell growth and cell cycle control, signal transduction, DNA repair, transcription, and many others. Hsp90 chaperone function is coupled to its ability to bind and hydrolyze ATP, which is tightly regulated both by co-chaperone proteins and post-translational modifications (PTMs). Many reported PTMs of Hsp90 alter chaperone function and consequently affect myriad cellular processes. Here, we review the contributions of PTMs, such as phosphorylation, acetylation, SUMOylation, methylation, O-GlcNAcylation, ubiquitination, and others, toward regulation of Hsp90 function. We also discuss how the Hsp90 modification state affects cellular sensitivity to Hsp90-targeted therapeutics that specifically bind and inhibit its chaperone activity. The ultimate challenge is to decipher the comprehensive and combinatorial array of PTMs that modulate Hsp90 chaperone function, a phenomenon termed the "chaperone code."
Collapse
Affiliation(s)
- Sarah J Backe
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Rebecca A Sager
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, New York, USA.,College of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Mark R Woodford
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Alan M Makedon
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Mehdi Mollapour
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA .,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
14
|
CIGB-300 anticancer peptide regulates the protein kinase CK2-dependent phosphoproteome. Mol Cell Biochem 2020; 470:63-75. [PMID: 32405972 DOI: 10.1007/s11010-020-03747-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/06/2020] [Indexed: 10/24/2022]
Abstract
Casein-kinase CK2 is a Ser/Thr protein kinase that fosters cell survival and proliferation of malignant cells. The CK2 holoenzyme, formed by the association of two catalytic alpha/alpha' (CK2α/CK2α') and two regulatory beta subunits (CK2β), phosphorylates diverse intracellular proteins partaking in key cellular processes. A handful of such CK2 substrates have been identified as targets for the substrate-binding anticancer peptide CIGB-300. However, since CK2β also contains a CK2 phosphorylation consensus motif, this peptide may also directly impinge on CK2 enzymatic activity, thus globally modifying the CK2-dependent phosphoproteome. To address such a possibility, firstly, we evaluated the potential interaction of CIGB-300 with CK2 subunits, both in cell-free assays and cellular lysates, as well as its effect on CK2 enzymatic activity. Then, we performed a phosphoproteomic survey focusing on early inhibitory events triggered by CIGB-300 and identified those CK2 substrates significantly inhibited along with disturbed cellular processes. Altogether, we provided here the first evidence for a direct impairment of CK2 enzymatic activity by CIGB-300. Of note, both CK2-mediated inhibitory mechanisms of this anticancer peptide (i.e., substrate- and enzyme-binding mechanism) may run in parallel in tumor cells and help to explain the different anti-neoplastic effects exerted by CIGB-300 in preclinical cancer models.
Collapse
|
15
|
Abstract
Tuberculosis (TB) is a major issue in global health and affects millions of people each year. Multidrug-resistant tuberculosis (MDR-TB) annually causes many deaths worldwide. Development of a way to diagnose and treat patients with MDR-TB can potentially reduce the incidence of the disease. The current study reviews the risk factors, pattern of progression, mechanism of resistance, and interaction between bacteria and the host immune system, which disrupts the immune response. It also targets the components of Mycobacterium tuberculosis (Mtb) and diagnosis and treatment options that could be available for clinical use in the near future. Mutations play an important role in development of MDR-TB and the selection of appropriate mutations can help to understand the type of resistance in patients to anti-TB drugs. In this way, they can be initially treated with proper and effective therapeutic choices, which can accelerate the course of treatment and improve patient health. Targeting the components and enzymes of Mtb is necessary for understanding bacterial survival and finding a way to destroy the pathogen and allow patients to recover faster and prevent the spread of disease, especially resistant strains.
Collapse
Affiliation(s)
- Majid Faridgohar
- Infectious Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran.,Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
16
|
Borgo C, Ruzzene M. Role of protein kinase CK2 in antitumor drug resistance. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:287. [PMID: 31277672 PMCID: PMC6612148 DOI: 10.1186/s13046-019-1292-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 06/25/2019] [Indexed: 01/21/2023]
Abstract
Drug resistance represents the major reason of pharmacological treatment failure. It is supported by a broad spectrum of mechanisms, whose molecular bases have been frequently correlated to aberrant protein phosphorylation. CK2 is a constitutively active protein kinase which phosphorylates hundreds of substrates; it is expressed in all cells, but its level is commonly found higher in cancer cells, where it plays anti-apoptotic, pro-migration and pro-proliferation functions. Several evidences support a role for CK2 in processes directly responsible of drug resistance, such as drug efflux and DNA repair; moreover, CK2 intervenes in signaling pathways which are crucial to evade drug response (as PI3K/AKT/PTEN, NF-κB, β-catenin, hedgehog signaling, p53), and controls the activity of chaperone machineries fundamental in resistant cells. Interestingly, a panel of specific and effective inhibitors of CK2 is available, and several examples are known of their efficacy in resistant cells, with synergistic effect when used in combination with conventional drugs, also in vivo. Here we analyze and discuss evidences supporting the hypothesis that CK2 targeting represents a valuable strategy to overcome drug resistance.
Collapse
Affiliation(s)
- Christian Borgo
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58b, 35131, Padova, Italy
| | - Maria Ruzzene
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58b, 35131, Padova, Italy.
| |
Collapse
|
17
|
Lee DH, Hasanuzzaman M, Kwon D, Choi HY, Kim SM, Kim DJ, Kang DJ, Hwang TH, Kim HH, Shin HJ, Shin JG, Oh S, Lee S, Kim SW. 10-Phenyltriazoyl Artemisinin is a Novel P-glycoprotein Inhibitor that Suppresses the Overexpression and Function of P-glycoprotein. Curr Pharm Des 2019; 24:5590-5597. [DOI: 10.2174/1381612825666190222155700] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 02/13/2019] [Indexed: 12/11/2022]
Abstract
Background:
The effect of drugs on ATP-binding cassette transporters, especially permeabilityglycoprotein
(P-gp), is an important consideration during new anti-cancer drug development.
Objective:
In this context, the effects of a newly synthesized artemisinin derivative, 10-(4-phenyl-1H-1,2,3-
triazol)-artemisinin (5a), were evaluated on P-gp expression and function.
Methods:
Reverse transcript polymerase chain reaction and immunoblotting techniques were used to determine
the effect of 5a on P-gp expression in LS174T cells. In addition, the ability of 5a to work as either a substrate or
an inhibitor of P-gp was investigated through different methods.
Results:
The results revealed that 5a acts as a novel P-gp inhibitor that dually suppresses the overexpression and
function of P-glycoprotein. Co-treatment of LS174T cell line, human colon adenocarcinoma cell line, with 5a and
paclitaxel recovered the anticancer effect of paclitaxel by controlling the acquired drug resistance pathway. The
overexpression of P-gp induced by rifampin and paclitaxel in a colorectal cell line was suppressed by 5a which
could be a novel inhibitory substrate inhibiting the transport of paclitaxel by P-gp.
Conclusion:
The results revealed that 5a can be classified as a type B P-gp inhibitor (with both substrate and
inhibitor activities) with an additional function of suppressing P-gp overexpression. The results might be clinically
useful in the development of anticancer drugs against cancers with multidrug resistance.
Collapse
Affiliation(s)
- Dong-Hwan Lee
- Hallym Institute for Clinical Medicine, Hallym University Medical Center, Anyang, 14066, Korea
| | - Md. Hasanuzzaman
- Department of Pharmacy, Noakhali Science and Technology University, Sonapur, Noakhali 3814, Bangladesh
| | - Daeho Kwon
- Department of Microbiology, Catholic Kwandong University College of Medicine, Gangneung 25601, Korea
| | - Hye-Young Choi
- Department of Pharmacology, Catholic Kwandong University College of Medicine, Gangneung 25601, Korea
| | - So Myoung Kim
- Department of Pharmacology, Catholic Kwandong University College of Medicine, Gangneung 25601, Korea
| | - Dong Jin Kim
- Approval and Review Team, Medical Device Safety Bureau, Ministry of Food and Drug Safety, Cheongju 28159, Korea
| | - Dong Ju Kang
- Department of Pharmacology, Catholic Kwandong University College of Medicine, Gangneung 25601, Korea
| | - Tae-Ho Hwang
- Gene and Cell Therapy Research Center for Vessel-associated Diseases, Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Hyung-Hoi Kim
- Department of Laboratory Medicine, (Bio) Medical Research Institute, School of Medicine, Pusan National University, Pusan National University Hospital, Busan 4924, Korea
| | - Ho Jung Shin
- SPMED Co., Ltd., 111 Hyoyeol-ro, Buk-gu, Busan 46508, Korea
| | - Jae-Gook Shin
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan 47392, Korea
| | - Sangtae Oh
- Department of Basic Science, Catholic Kwandong University College of Medicine, Gangneung 25601, Korea
| | - Seokjoon Lee
- Department of Pharmacology, Catholic Kwandong University College of Medicine, Gangneung 25601, Korea
| | - So Won Kim
- Department of Pharmacology, Catholic Kwandong University College of Medicine, Gangneung 25601, Korea
| |
Collapse
|
18
|
Rodríguez-Ulloa A, Ramos Y, Sánchez-Puente A, Perera Y, Musacchio-Lasa A, Fernández-de-Cossio J, Padrón G, López LJ, Besada V, Perea SE. The Combination of the CIGB-300 Anticancer Peptide and Cisplatin Modulates Proteins Related to Cell Survival, DNA Repair and Metastasis in a Lung Cancer Cell Line Model. CURR PROTEOMICS 2019. [DOI: 10.2174/1570164616666190126104325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
CIGB-300 is a pro-apoptotic peptide that abrogates CK2-mediated phosphorylation,
and can elicit synergistic interaction in vitro and in vivo when combined with certain anticancer
drugs.
Objective:
The combination of CIGB-300 with cisplatin is studied through data mining and expressionbased
proteomics to reveal the molecular basis of this interaction. Cisplatin resistance-associated proteins,
which have also been reported as CK2 substrates, were first identified by bioinformatic analyses.
Methods:
Data from these analyses suggested that the cisplatin resistance phenotype could be directly
improved by inhibiting CK2 phosphorylation on specific substrates. Furthermore, 157 proteins were
differentially modulated on the NCI-H125 lung cancer cell line in response to CIGB-300, cisplatin or
both drugs as determined by LC-MS/MS.
Results:
The expression of 28 cisplatin resistance-associated proteins was changed when cisplatin was
combined with CIGB-300. Overall, the proteins identified are also related to cell survival, cell proliferation
and metastasis. Furthermore, the CIGB-300 regulated proteome revealed proteins that were initially
involved in the mechanism of action of CIGB-300 and cisplatin as single agents.
Conclusion:
This is the first report describing the protein array modulated by combining CIGB-300
and cisplatin that will support the rationale for future clinical settings based on a multi-target cancer
therapy.
Collapse
Affiliation(s)
| | - Yassel Ramos
- Department of Proteomics, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Aniel Sánchez-Puente
- Department of Proteomics, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Yasser Perera
- Laboratory of Molecular Oncology, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Alexis Musacchio-Lasa
- Department of Bioinformatics, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | | | - Gabriel Padrón
- Department of Proteomics, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Luis J.G. López
- Department of Proteomics, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Vladimir Besada
- Department of Proteomics, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Silvio E. Perea
- Laboratory of Molecular Oncology, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| |
Collapse
|
19
|
Hasanuzzaman M, Yi M, Cho M, Parvez MM, Lee SJ, Shin JG. Rifampin Induces Expression of P-glycoprotein on the THP1 Cell-Derived Macrophages, Causing Decrease Intramacrophage Concentration of Prothionamide. J Pharm Sci 2019; 108:3106-3111. [PMID: 30991038 DOI: 10.1016/j.xphs.2019.04.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 04/05/2019] [Accepted: 04/09/2019] [Indexed: 11/19/2022]
Abstract
Rifampin (RIF) has been widely used for the treatment of bacterial infections, including tuberculosis (TB). Treatment of drug-resistant TB is a global problem because of reduced drug efficacy. The present study determined the effect of RIF on MDR1 gene (P-glycoprotein, P-gp) expression in THP1 macrophages and analyzed the intracellular concentration of the anti-TB drug prothionamide in the presence of RIF. RIF treatment significantly induced MDR1 protein and mRNA levels in phorbol 12-myristate 13-acetate-stimulated THP1 macrophages (p < 0.001 and 0.01, respectively). The pregnane X receptor inhibitors resveratrol and ketoconazole significantly suppressed RIF-induced P-gp expression in THP1 macrophages (p < 0.05). RIF-treated THP1 macrophages also exhibited strong efflux of P-gp substrate, resulting in a reduced intracellular concentration of rhodamine-123 and prothionamide (p < 0.01 and 0.05, respectively). By contrast, the P-gp inhibitor cyclosporine A significantly increased intracellular concentration of rhodamine-123 and prothionamide (p < 0.001 and 0.05, respectively). The present results suggest that the usage of RIF together with P-gp-substrate drugs to treat TB may lead to deteriorated treatment efficacy because of the lower intracellular drug concentration. Further studies would be necessary to know the influence of RIF-induced P-gp induction on the treatment outcome of patients with TB.
Collapse
Affiliation(s)
- Md Hasanuzzaman
- Department of Pharmacology and Clinical Pharmacology, Pharmacogenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea; Department of Pharmacy, Noakhali Science and Technology University, Sonapur, Noakhali 3814, Bangladesh
| | - Myeongjin Yi
- Department of Pharmacology and Clinical Pharmacology, Pharmacogenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| | - Munju Cho
- Department of Pharmacology and Clinical Pharmacology, Pharmacogenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| | - Md Masud Parvez
- Department of Pharmacology and Clinical Pharmacology, Pharmacogenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea; Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Su-Jun Lee
- Department of Pharmacology and Clinical Pharmacology, Pharmacogenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea; Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Jae-Gook Shin
- Department of Pharmacology and Clinical Pharmacology, Pharmacogenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea; Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea.
| |
Collapse
|
20
|
Taneja G, Chu C, Maturu P, Moorthy B, Ghose R. Role of c-Jun-N-Terminal Kinase in Pregnane X Receptor-Mediated Induction of Human Cytochrome P4503A4 In Vitro. Drug Metab Dispos 2018; 46:397-404. [PMID: 29440179 PMCID: PMC5829542 DOI: 10.1124/dmd.117.079160] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/01/2018] [Indexed: 12/17/2022] Open
Abstract
Cytochrome P450 CYP3A4 is the most abundant drug-metabolizing enzyme and is responsible for the metabolism of ∼50% of clinically available drugs. Induction of CYP3A4 impacts the disposition of its substrates and leads to harmful clinical consequences, such as failure of therapy. To prevent such undesirable consequences, the molecular mechanisms of regulation of CYP3A4 need to be fully understood. CYP3A4 induction is regulated primarily by the xenobiotic nuclear receptor pregnane-X receptor (PXR). After ligand binding, PXR is translocated to the nucleus, where it binds to the CYP3A4 promoter and induces its gene expression. PXR function is modulated by phosphorylation(s) by multiple kinases. In this study, we determined the role of the c-Jun N-terminal kinase (JNK) in PXR-mediated induction of CYP3A4 enzyme in vitro. Human liver carcinoma cells (HepG2) were transfected with CYP3A4 luciferase and PXR plasmids, followed by treatment with JNK inhibitor (SP600125; SP) and PXR activators rifampicin (RIF) or hyperforin. Our results indicate that SP treatment significantly attenuated PXR-mediated induction of CYP3A4 reporter activity, as well as gene expression and enzyme activity. JNK knockdown by siRNA (targeting both JNK 1 and 2) also attenuated CYP3A4 induction by RIF. Interestingly, SP treatment attenuated JNK activation by RIF. Furthermore, treatment with RIF increased PXR nuclear levels and binding to the CYP3A4 promoter; SP attenuated these effects. This study shows that JNK is a novel mechanistic regulator of CYP3A4 induction by PXR.
Collapse
Affiliation(s)
- Guncha Taneja
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston (G.T., R.G.), and Department of Pediatrics, Baylor College of Medicine (C.C., P.M., B.M.), Houston, Texas
| | - Chun Chu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston (G.T., R.G.), and Department of Pediatrics, Baylor College of Medicine (C.C., P.M., B.M.), Houston, Texas
| | - Paramahamsa Maturu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston (G.T., R.G.), and Department of Pediatrics, Baylor College of Medicine (C.C., P.M., B.M.), Houston, Texas
| | - Bhagavatula Moorthy
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston (G.T., R.G.), and Department of Pediatrics, Baylor College of Medicine (C.C., P.M., B.M.), Houston, Texas
| | - Romi Ghose
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston (G.T., R.G.), and Department of Pediatrics, Baylor College of Medicine (C.C., P.M., B.M.), Houston, Texas
| |
Collapse
|
21
|
Kim SW, Md Hasanuzzaman, Cho M, Kim NH, Choi HY, Han JW, Park HJ, Oh JW, Shin JG. Role of 14-3-3 sigma in over-expression of P-gp by rifampin and paclitaxel stimulation through interaction with PXR. Cell Signal 2017; 31:124-134. [PMID: 28077325 DOI: 10.1016/j.cellsig.2017.01.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 01/01/2017] [Accepted: 01/03/2017] [Indexed: 01/04/2023]
Abstract
In this study, we presented the role of 14-3-3σ to activate CK2-Hsp90β-PXR-MDR1 pathway on rifampin and paclitaxel treated LS174T cells and in vivo LS174T cell-xenografted nude mouse model. Following several in vitro and in vivo experiments, rifampin and paclitaxel were found to be stimulated the CK2-Hsp90β-PXR-MDR1 pathway. Of the proteins in this pathway, Pregnane X receptor (PXR) is a representative transcription factor of multidrug resistance protein 1 (MDR1). We constructed FLAG-PXR-LS174T stable cell lines and discovered 22 proteins that interacted with PXR on rifampin treatment. Among them, Hsp90β and 14-3-3σ were isolated for further study. Both the proteins were found to be localized in cytoplasm on rifampin treatment by using confocal microscopy. On the other hand, PXR was found to be localized in nucleus after rifampin and paclitaxel treatment by using cell fractionation assay. In Western blot analysis, rifampin did not influence the expression of 14-3-3σ protein. Transient transfection of 14-3-3σ into LS174T cells induced overexpression of PXR; however, P-glycoprotein (P-gp) was not changed significantly. P-gp overexpression was induced only when 14-3-3σ transfected LS174T cells were treated with rifampin and paclitaxel, whereas 14-3-3σ inhibition by nonpeptidic inhibitor, BV02 and 14-3-3σ siRNA reduced rifampin induced PXR and P-gp expression. Cell survival rates were much higher at 14-3-3σ-LS174T stable cell lines than LS174T cells following paclitaxel and vincristine treatment. This data indicates that 14-3-3σ contributes to P-gp overexpression through interaction with PXR with rifampin and paclitaxel treatment.
Collapse
Affiliation(s)
- So Won Kim
- Department of Pharmacology, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea; The Institute for Clinical and Translational Research, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea.
| | - Md Hasanuzzaman
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea; Department of Pharmacy, Noakhali Science and Technology University, Sonapur, Noakhali 3814, Bangladesh
| | - Munju Cho
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Nam Hyun Kim
- Department of Pharmacology, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea
| | - Hye-Young Choi
- Department of Pharmacology, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea
| | - Jung Woo Han
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyun June Park
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Ji Won Oh
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Bio-Medical Research Institute, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Jae-Gook Shin
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea; Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan 47392, Republic of Korea.
| |
Collapse
|
22
|
Woolsey SJ, Beaton MD, Mansell SE, Leon-Ponte M, Yu J, Pin CL, Adams PC, Kim RB, Tirona RG. A Fibroblast Growth Factor 21-Pregnane X Receptor Pathway Downregulates Hepatic CYP3A4 in Nonalcoholic Fatty Liver Disease. Mol Pharmacol 2016; 90:437-46. [PMID: 27482056 DOI: 10.1124/mol.116.104687] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/28/2016] [Indexed: 12/28/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) alters drug response. We previously reported that NAFLD is associated with reduced in vivo CYP3A drug-metabolism activity and hepatic CYP3A4 expression in humans as well as mouse and human hepatoma models of the disease. Here, we investigated the role of the lipid- and glucose-modulating hormone fibroblast growth factor 21 (FGF21) in the molecular mechanism regulating CYP3A4 expression in NAFLD. In human subjects, mouse and cellular NAFLD models with lower CYP3A4 expression, circulating FGF21, or hepatic FGF21 mRNA levels were elevated. Administration of recombinant FGF21 or transient hepatic overexpression of FGF21 resulted in reduced liver CYP3A4 luciferase reporter activity in mice and decreased CYP3A4 mRNA expression and activity in cultured Huh7 hepatoma cells. Blocking canonical FGF21 signaling by pharmacological inhibition of MEK1 kinase in Huh7 cells caused de-repression of CYP3A4 mRNA expression with FGF21 treatment. Mice with high-fat diet-induced simple hepatic steatosis and lipid-loaded Huh7 cells had reduced nuclear localization of the pregnane X receptor (PXR), a key transcriptional regulator of CYP3A4 Furthermore, decreased nuclear PXR was observed in mouse liver and Huh7 cells after FGF21 treatment or FGF21 overexpression. Decreased PXR binding to the CYP3A4 proximal promoter was found in FGF21-treated Huh7 cells. An FGF21-PXR signaling pathway may be involved in decreased hepatic CYP3A4 metabolic activity in NAFLD.
Collapse
Affiliation(s)
- Sarah J Woolsey
- Department of Physiology and Pharmacology (S.J.W., J.Y., C.L.P., R.B.K., R.G.T), Division of Gastroenterology, Department of Medicine (M.D.B., P.C.A.), Division of Clinical Pharmacology, Department of Medicine (S.J.W., S.E.M., M.L.-P., J.Y., R.B.K., R.G.T.), Department of Paediatrics (C.L.P.), and Department of Oncology (C.L.P., R.B.K.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Melanie D Beaton
- Department of Physiology and Pharmacology (S.J.W., J.Y., C.L.P., R.B.K., R.G.T), Division of Gastroenterology, Department of Medicine (M.D.B., P.C.A.), Division of Clinical Pharmacology, Department of Medicine (S.J.W., S.E.M., M.L.-P., J.Y., R.B.K., R.G.T.), Department of Paediatrics (C.L.P.), and Department of Oncology (C.L.P., R.B.K.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Sara E Mansell
- Department of Physiology and Pharmacology (S.J.W., J.Y., C.L.P., R.B.K., R.G.T), Division of Gastroenterology, Department of Medicine (M.D.B., P.C.A.), Division of Clinical Pharmacology, Department of Medicine (S.J.W., S.E.M., M.L.-P., J.Y., R.B.K., R.G.T.), Department of Paediatrics (C.L.P.), and Department of Oncology (C.L.P., R.B.K.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Matilde Leon-Ponte
- Department of Physiology and Pharmacology (S.J.W., J.Y., C.L.P., R.B.K., R.G.T), Division of Gastroenterology, Department of Medicine (M.D.B., P.C.A.), Division of Clinical Pharmacology, Department of Medicine (S.J.W., S.E.M., M.L.-P., J.Y., R.B.K., R.G.T.), Department of Paediatrics (C.L.P.), and Department of Oncology (C.L.P., R.B.K.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Janice Yu
- Department of Physiology and Pharmacology (S.J.W., J.Y., C.L.P., R.B.K., R.G.T), Division of Gastroenterology, Department of Medicine (M.D.B., P.C.A.), Division of Clinical Pharmacology, Department of Medicine (S.J.W., S.E.M., M.L.-P., J.Y., R.B.K., R.G.T.), Department of Paediatrics (C.L.P.), and Department of Oncology (C.L.P., R.B.K.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Christopher L Pin
- Department of Physiology and Pharmacology (S.J.W., J.Y., C.L.P., R.B.K., R.G.T), Division of Gastroenterology, Department of Medicine (M.D.B., P.C.A.), Division of Clinical Pharmacology, Department of Medicine (S.J.W., S.E.M., M.L.-P., J.Y., R.B.K., R.G.T.), Department of Paediatrics (C.L.P.), and Department of Oncology (C.L.P., R.B.K.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Paul C Adams
- Department of Physiology and Pharmacology (S.J.W., J.Y., C.L.P., R.B.K., R.G.T), Division of Gastroenterology, Department of Medicine (M.D.B., P.C.A.), Division of Clinical Pharmacology, Department of Medicine (S.J.W., S.E.M., M.L.-P., J.Y., R.B.K., R.G.T.), Department of Paediatrics (C.L.P.), and Department of Oncology (C.L.P., R.B.K.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Richard B Kim
- Department of Physiology and Pharmacology (S.J.W., J.Y., C.L.P., R.B.K., R.G.T), Division of Gastroenterology, Department of Medicine (M.D.B., P.C.A.), Division of Clinical Pharmacology, Department of Medicine (S.J.W., S.E.M., M.L.-P., J.Y., R.B.K., R.G.T.), Department of Paediatrics (C.L.P.), and Department of Oncology (C.L.P., R.B.K.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Rommel G Tirona
- Department of Physiology and Pharmacology (S.J.W., J.Y., C.L.P., R.B.K., R.G.T), Division of Gastroenterology, Department of Medicine (M.D.B., P.C.A.), Division of Clinical Pharmacology, Department of Medicine (S.J.W., S.E.M., M.L.-P., J.Y., R.B.K., R.G.T.), Department of Paediatrics (C.L.P.), and Department of Oncology (C.L.P., R.B.K.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
23
|
Oladimeji P, Cui H, Zhang C, Chen T. Regulation of PXR and CAR by protein-protein interaction and signaling crosstalk. Expert Opin Drug Metab Toxicol 2016; 12:997-1010. [PMID: 27295009 DOI: 10.1080/17425255.2016.1201069] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Protein-protein interaction and signaling crosstalk contribute to the regulation of pregnane X receptor (PXR) and constitutive androstane receptor (CAR) and broaden their cellular function. AREA COVERED This review covers key historic discoveries and recent advances in our understanding of the broad function of PXR and CAR and their regulation by protein-protein interaction and signaling crosstalk. EXPERT OPINION PXR and CAR were first discovered as xenobiotic receptors; however, it is clear that PXR and CAR perform a much broader range of cellular functions through protein-protein interaction and signaling crosstalk, which typically mutually affect the function of all the partners involved. Future research on PXR and CAR should, therefore, look beyond their xenobiotic function.
Collapse
Affiliation(s)
- Peter Oladimeji
- a Department of Chemical Biology and Therapeutics , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Hongmei Cui
- a Department of Chemical Biology and Therapeutics , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Chen Zhang
- a Department of Chemical Biology and Therapeutics , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Taosheng Chen
- a Department of Chemical Biology and Therapeutics , St. Jude Children's Research Hospital , Memphis , TN , USA
| |
Collapse
|
24
|
Renaud HJ, Klaassen CD, Csanaky IL. Calorie Restriction Increases P-Glycoprotein and Decreases Intestinal Absorption of Digoxin in Mice. Drug Metab Dispos 2016; 44:366-9. [PMID: 26744253 PMCID: PMC4767382 DOI: 10.1124/dmd.115.064766] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 01/06/2016] [Indexed: 02/05/2023] Open
Abstract
There is wide variation in how patients respond to therapeutics. Factors that contribute to pharmacokinetic variations include disease, genetics, drugs, age, and diet. The purpose of this study was to determine the effect of calorie restriction on the expression of Abcb1a in the intestine and whether calorie restriction can alter the absorption of an Abcb1a substrate (i.e., digoxin) in mice. Ten-week-old C57BL/6 mice were given either an ad libitum diet or a 25% calorie-restricted diet for 3 weeks. To determine digoxin absorption, mice were administered [(3)H]-labeled digoxin by oral gavage. Blood and intestine with contents were collected at 1, 2, 4, and 12 hours after digoxin administration. Concentrations of [(3)H]-digoxin in plasma and tissues were determined by liquid scintillation. Calorie restriction decreased plasma digoxin concentrations (about 60%) at 1, 2, and 4 hours after administration. Additionally, digoxin concentrations in the small intestine of calorie-restricted mice were elevated at 4 and 12 hours after administration. Furthermore, calorie restriction increased Abcb1a transcripts in the duodenum (4.5-fold) and jejunum (12.5-fold). To confirm a role of Abcb1a in the altered digoxin pharmacokinetics induced by calorie restriction, the experiment was repeated in Abcb1a/b-null mice 4 hours after drug administration. No difference in intestine or plasma digoxin concentrations were observed between ad libitum-fed and calorie-restricted Abcb1a/b-null mice. Thus, these findings support the hypothesis that calorie restriction increases intestinal Abcb1a expression, leading to decreased absorption of digoxin in mice. Because Abcb1a transports a wide variety of therapeutics, these results may be of important clinical significance.
Collapse
Affiliation(s)
- Helen J Renaud
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Curtis D Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Iván L Csanaky
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
25
|
Acetylation of lysine 109 modulates pregnane X receptor DNA binding and transcriptional activity. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1155-1169. [PMID: 26855179 DOI: 10.1016/j.bbagrm.2016.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 01/19/2016] [Accepted: 01/26/2016] [Indexed: 12/31/2022]
Abstract
Pregnane X receptor (PXR) is a major transcriptional regulator of xenobiotic metabolism and transport pathways in the liver and intestines, which are critical for protecting organisms against potentially harmful xenobiotic and endobiotic compounds. Inadvertent activation of drug metabolism pathways through PXR is known to contribute to drug resistance, adverse drug-drug interactions, and drug toxicity in humans. In both humans and rodents, PXR has been implicated in non-alcoholic fatty liver disease, diabetes, obesity, inflammatory bowel disease, and cancer. Because of PXR's important functions, it has been a therapeutic target of interest for a long time. More recent mechanistic studies have shown that PXR is modulated by multiple PTMs. Herein we provide the first investigation of the role of acetylation in modulating PXR activity. Through LC-MS/MS analysis, we identified lysine 109 (K109) in the hinge as PXR's major acetylation site. Using various biochemical and cell-based assays, we show that PXR's acetylation status and transcriptional activity are modulated by E1A binding protein (p300) and sirtuin 1 (SIRT1). Based on analysis of acetylation site mutants, we found that acetylation at K109 represses PXR transcriptional activity. The mechanism involves loss of RXRα dimerization and reduced binding to cognate DNA response elements. This mechanism may represent a promising therapeutic target using modulators of PXR acetylation levels. This article is part of a Special Issue entitled: Xenobiotic nuclear receptors: New Tricks for An Old Dog, edited by Dr. Wen Xie.
Collapse
|
26
|
Faya N, Penkler DL, Tastan Bishop Ö. Human, vector and parasite Hsp90 proteins: A comparative bioinformatics analysis. FEBS Open Bio 2015; 5:916-27. [PMID: 26793431 PMCID: PMC4688443 DOI: 10.1016/j.fob.2015.11.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/23/2015] [Accepted: 11/09/2015] [Indexed: 12/27/2022] Open
Abstract
Cytosolic Hsp90s are more conserved than those from mitochondrial and ER. Cell environment plays a role in the overall physicochemical properties of Hsp90s. Serine and tyrosine are favored phosphorylated residues of Hsp90s. Mitochondrial and ER Hsp90s have motifs unique to specific organisms.
The treatment of protozoan parasitic diseases is challenging, and thus identification and analysis of new drug targets is important. Parasites survive within host organisms, and some need intermediate hosts to complete their life cycle. Changing host environment puts stress on parasites, and often adaptation is accompanied by the expression of large amounts of heat shock proteins (Hsps). Among Hsps, Hsp90 proteins play an important role in stress environments. Yet, there has been little computational research on Hsp90 proteins to analyze them comparatively as potential parasitic drug targets. Here, an attempt was made to gain detailed insights into the differences between host, vector and parasitic Hsp90 proteins by large-scale bioinformatics analysis. A total of 104 Hsp90 sequences were divided into three groups based on their cellular localizations; namely cytosolic, mitochondrial and endoplasmic reticulum (ER). Further, the parasitic proteins were divided according to the type of parasite (protozoa, helminth and ectoparasite). Primary sequence analysis, phylogenetic tree calculations, motif analysis and physicochemical properties of Hsp90 proteins suggested that despite the overall structural conservation of these proteins, parasitic Hsp90 proteins have unique features which differentiate them from human ones, thus encouraging the idea that protozoan Hsp90 proteins should be further analyzed as potential drug targets.
Collapse
|