1
|
Li W, Zhu K, Liu Y, Liu M, Chen Q. Recent advances in PKC inhibitor development: Structural design strategies and therapeutic applications. Eur J Med Chem 2025; 287:117290. [PMID: 39904144 DOI: 10.1016/j.ejmech.2025.117290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Protein kinase C (PKC) isozymes play critical roles in diverse cellular processes and are implicated in numerous diseases, including cancer, diabetes, and autoimmune disorders. Despite extensive research efforts spanning four decades, only one PKC inhibitor has received clinical approval, highlighting the challenges in developing selective and efficacious PKC-targeting therapeutics. Here we review recent advances in the development of small-molecule PKC inhibitors, focusing on structural design strategies, pharmacological activities, and structure-activity relationships. We analyze emerging approaches including fragment-based drug design, allosteric targeting, and natural product derivatization that have yielded promising new scaffold classes. Special attention is given to innovations in achieving isozyme selectivity, particularly for PKCα and PKCβ, which have proven crucial for therapeutic applications. We discuss how integration of computational methods, structural biology insights, and rational design principles has advanced our understanding of PKC inhibition mechanisms. This comprehensive analysis reveals key challenges in PKC drug development, including the need for enhanced selectivity and reduced off-target effects, while highlighting promising directions for future therapeutic development. Our findings provide a framework for designing next-generation PKC inhibitors with improved clinical potential.
Collapse
Affiliation(s)
- Wen Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Kun Zhu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yuyin Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Meixi Liu
- Department of Endocrinology, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, 618000, China
| | - Qiu Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
2
|
Massey S, Kongchan N, Gao Y, Chaudhury A, Olokpa E, Karch J, Malovannaya A, Cheng C, Zhang X, Neilson JR. PKC-mediated phosphorylation governs the stability and function of CELF1 as a driver of EMT in breast epithelial cells. J Biol Chem 2024; 300:107826. [PMID: 39343007 PMCID: PMC11585768 DOI: 10.1016/j.jbc.2024.107826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 10/01/2024] Open
Abstract
Epithelial to mesenchymal transition (EMT) is believed to be a principal factor contributing to cancer metastasis. The post-transcriptional and post-translational mechanisms underlying EMT are comparatively underexplored. We previously demonstrated that the CELF1 RNA binding protein is necessary and sufficient to drive the EMT of breast epithelial cells, and that the relative protein expression of CELF1 in this context was dictated at the post-translational level. Here, we elucidate the mechanism of this regulation. Mass spectrometric analysis of CELF1 isolated from mesenchymal MCF-10A cells identified multiple sites of serine and threonine phosphorylation on the protein, correlating with the increased stability of this protein in this cellular state. Analysis of phosphomimetic and serine/threonine-to-alanine phosphomutant variants of CELF1 revealed that these phosphorylation sites indeed dictate CELF1 stability, ubiquitination state, and function in vitro. Via co-immunoprecipitation and in vitro kinase assays, we identified the protein kinase C alpha and epsilon isozymes as the kinases responsible for CELF1 phosphorylation in a breast cell line. Genetic epistasis experiments confirmed that these PKCs function upstream of CELF1 in this EMT program, and CELF1 phosphorylation impacts tumor metastasis in a xenograft model. This work is the first to formally establish the mechanisms underlying post-translational control of CELF1 expression and function during EMT of breast epithelial cells. Given the broad dysregulation of CELF1 expression in human breast cancer, our results may ultimately provide knowledge that may be leveraged for novel therapeutic interventions in this context.
Collapse
Affiliation(s)
- Shebna Massey
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - Natee Kongchan
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - Yang Gao
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Arindam Chaudhury
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - Emuejevoke Olokpa
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - Jason Karch
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - Anna Malovannaya
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Chonghui Cheng
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Xiang Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; McNair Medical Institute, Baylor College of Medicine, Houston, Texas, USA
| | - Joel R Neilson
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA.
| |
Collapse
|
3
|
Kim HAJ, Zeng PYF, Cecchini M, Shaikh MH, Laxague F, Deng X, Jarycki L, Ryan SEB, Dawson A, Liu MH, Palma DA, Patel K, Mundi N, Barrett JW, Mymryk JS, Boutros PC, Nichols AC. HPV-negative head and neck cancers with adverse pathological features carry specific molecular changes that are associated with survival. Head Neck 2024; 46:353-366. [PMID: 38059331 DOI: 10.1002/hed.27591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 10/21/2023] [Accepted: 11/21/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Adverse pathological features following surgery in head and neck squamous cell carcinoma (HNSCC) are strongly associated with survival and guide adjuvant therapy. We investigated molecular changes associated with these features. METHODS We downloaded data from the Cancer Genome Atlas and Cancer Proteome Atlas HNSCC cohorts. We compared tumors positive versus negative for perineural invasion (PNI), lymphovascular invasion (LVI), extracapsular spread (ECS), and positive margins (PSM), with multivariable analysis. RESULTS All pathological features were associated with poor survival, as were the following molecular changes: low cyclin E1 (HR = 1.7) and high PKC-alpha (HR = 1.8) in tumors with PNI; six of 13 protein abundance changes with LVI; greater tumor hypoxia and high Raptor (HR = 2.0) and Rictor (HR = 1.6) with ECS; and low p38 (HR = 2.3), high fibronectin (HR = 1.6), low annexin A1 (HR = 3.1), and high caspase-9 (HR = 1.6) abundances with PSM. CONCLUSIONS Pathological features in HNSCC carry specific molecular changes that may explain their poor prognostic associations.
Collapse
Affiliation(s)
- Hugh Andrew Jinwook Kim
- Department of Otolaryngology-Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Peter Y F Zeng
- Department of Otolaryngology-Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Matthew Cecchini
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
| | - Mushfiq Hassan Shaikh
- Department of Otolaryngology-Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Francisco Laxague
- Department of Otolaryngology-Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Xiaoxiao Deng
- Department of Otolaryngology-Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Laura Jarycki
- Department of Otolaryngology-Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Sarah Elizabeth Belle Ryan
- Department of Otolaryngology-Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
| | - Alice Dawson
- Department of Otolaryngology-Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
| | - Mu Han Liu
- Department of Otolaryngology-Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - David A Palma
- Department of Otolaryngology-Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
- Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - Krupal Patel
- Department of Otolaryngology-Head & Neck Surgery, Moffitt Cancer Center, Tampa, Florida, USA
| | - Neil Mundi
- Department of Otolaryngology-Head & Neck Surgery, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - John W Barrett
- Department of Otolaryngology-Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
- Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - Joe S Mymryk
- Department of Otolaryngology-Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
- Department of Oncology, University of Western Ontario, London, Ontario, Canada
- Department of Microbiology & Immunology, University of Western Ontario, London, Ontario, Canada
| | - Paul C Boutros
- Department of Human Genetics, University of California, Los Angeles, California, USA
- Department of Urology, University of California, Los Angeles, California, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, California, USA
- Institute for Precision Health, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, California, USA
| | - Anthony C Nichols
- Department of Otolaryngology-Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
- Department of Oncology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
4
|
Wang J, Casimiro-Garcia A, Johnson BG, Duffen J, Cain M, Savary L, Wang S, Nambiar P, Lech M, Zhao S, Xi L, Zhan Y, Olson J, Stejskal JA, Lin H, Zhang B, Martinez RV, Masek-Hammerman K, Schlerman FJ, Dower K. A protein kinase C α and β inhibitor blunts hyperphagia to halt renal function decline and reduces adiposity in a rat model of obesity-driven type 2 diabetes. Sci Rep 2023; 13:16919. [PMID: 37805649 PMCID: PMC10560236 DOI: 10.1038/s41598-023-43759-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 09/28/2023] [Indexed: 10/09/2023] Open
Abstract
Type 2 diabetes (T2D) and its complications can have debilitating, sometimes fatal consequences for afflicted individuals. The disease can be difficult to control, and therapeutic strategies to prevent T2D-induced tissue and organ damage are needed. Here we describe the results of administering a potent and selective inhibitor of Protein Kinase C (PKC) family members PKCα and PKCβ, Cmpd 1, in the ZSF1 obese rat model of hyperphagia-induced, obesity-driven T2D. Although our initial intent was to evaluate the effect of PKCα/β inhibition on renal damage in this model setting, Cmpd 1 unexpectedly caused a marked reduction in the hyperphagic response of ZSF1 obese animals. This halted renal function decline but did so indirectly and indistinguishably from a pair feeding comparator group. However, above and beyond this food intake effect, Cmpd 1 lowered overall animal body weights, reduced liver vacuolation, and reduced inguinal adipose tissue (iWAT) mass, inflammation, and adipocyte size. Taken together, Cmpd 1 had strong effects on multiple disease parameters in this obesity-driven rodent model of T2D. Further evaluation for potential translation of PKCα/β inhibition to T2D and obesity in humans is warranted.
Collapse
Affiliation(s)
- Ju Wang
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA.
| | | | - Bryce G Johnson
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Jennifer Duffen
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Michael Cain
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Mediar Therapeutics, Boston, MA, USA
| | - Leigh Savary
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Instem Life Science Systems Ltd, Mount Ida College, South Hadley, MA, USA
| | - Stephen Wang
- Pharmacokinetics and Drug Metabolism, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Novartis Gene Therapies, Novartis Institute for Biomedical Research, Cambridge, MA, USA
| | - Prashant Nambiar
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Strand Therapeutics, Cambridge, MA, USA
| | - Matthew Lech
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Shanrong Zhao
- Clinical Genetics and Bioinformatics, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Amunix Pharmaceuticals, San Francisco, CA, USA
| | - Li Xi
- Early Clinical Development, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Yutian Zhan
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Jennifer Olson
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, Groton, CT, USA
| | - James A Stejskal
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, Groton, CT, USA
- Charles River Laboratories, Shrewsbury, MA, USA
| | - Hank Lin
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Sunovion Pharmaceuticals Inc., Marlborough, MA, USA
| | - Baohong Zhang
- Clinical Genetics and Bioinformatics, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Data Sciences, Biogen, Cambridge, MA, USA
| | - Robert V Martinez
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Center for Technological Innovation, Pfizer Worldwide Research and Development, San Francisco, CA, USA
| | | | - Franklin J Schlerman
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Ken Dower
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA.
| |
Collapse
|
5
|
Singh RK, Kumar S, Kumar S, Shukla A, Kumar N, Patel AK, Yadav LK, Kaushalendra, Antiwal M, Acharya A. Potential implications of protein kinase Cα in pathophysiological conditions and therapeutic interventions. Life Sci 2023; 330:121999. [PMID: 37536614 DOI: 10.1016/j.lfs.2023.121999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
PKCα is a molecule with many functions that play an important role in cell survival and death to maintain cellular homeostasis. Alteration in the normal functioning of PKCα is responsible for the complicated etiology of many pathologies, including cancer, cardiovascular diseases, kidney complications, neurodegenerative diseases, diabetics, and many others. Several studies have been carried out over the years on this kinase's function, and regulation in normal physiology and pathological conditions. A lot of data with antithetical results have therefore accumulated over time to create a complex framework of physiological implications connected to the PKCα function that needs comprehensive elucidation. In light of this information, we critically analyze the multiple roles played by PKCα in basic cellular processes and their molecular mechanism during various pathological conditions. This review further discusses the current approaches to manipulating PKCα signaling amplitude in the patient's favour and proposed PKCα as a therapeutic target to reverse pathological states.
Collapse
Affiliation(s)
- Rishi Kant Singh
- Lab of Hematopoiesis and Leukemia, KSBS, Indian Institute of Technology, Delhi, New Delhi 110016, India; Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Sanjay Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Sandeep Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Alok Shukla
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Naveen Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Anand Kumar Patel
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Lokesh Kumar Yadav
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Kaushalendra
- Department of Zoology, Pachhunga University College Campus, Mizoram University, Aizawl 796001, India
| | - Meera Antiwal
- Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Arbind Acharya
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
6
|
Wang Q, Chang Y, Yang X, Han Z. Deep sequencing of circulating miRNAs and target mRNAs level in deep venous thrombosis patients. IET Syst Biol 2023; 17:212-227. [PMID: 37466160 PMCID: PMC10439493 DOI: 10.1049/syb2.12071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 06/15/2023] [Accepted: 06/26/2023] [Indexed: 07/20/2023] Open
Abstract
Deep venous thrombosis is one of the most common peripheral vascular diseases that lead to major morbidity and mortality. The authors aimed to identify potential differentially expressed miRNAs and target mRNAs, which were helpful in understanding the potential molecule mechanism of deep venous thrombosis. The plasma samples of patients with deep venous thrombosis were obtained for the RNA sequencing. Differentially expressed miRNAs were identified, followed by miRNA-mRNA target analysis. Enrichment analysis was used to analyze the potential biological function of target mRNAs. GSE19151 and GSE173461 datasets were used for expression validation of mRNAs and miRNAs. 131 target mRNAs of 21 differentially expressed miRNAs were identified. Among which, 8 differentially expressed miRNAs including hsa-miR-150-5p, hsa-miR-326, hsa-miR-144-3p, hsa-miR-199a-5p, hsa-miR-199b-5p, hsa-miR-125a-5p, hsa-let-7e-5p and hsa-miR-381-3p and their target mRNAs (PRKCA, SP1, TP53, SLC27A4, PDE1B, EPHB3, IRS1, HIF1A, MTUS1 and ZNF652) were found associated with deep venous thrombosis for the first time. Interestingly, PDE1B and IRS1 had a potential diagnostic value for patients. Additionally, 3 important signaling pathways including p53, PI3K-Akt and MAPK were identified in the enrichment analysis of target mRNAs (TP53, PRKCA and IRS1). Identified circulating miRNAs and target mRNAs and related signaling pathways may be involved in the process of deep venous thrombosis.
Collapse
Affiliation(s)
- Qingxian Wang
- Department of Orthopedic Trauma, Orthopedic Research Institution of Hebei ProvinceKey Labratory of Biomechanics of Hebei ProvinceThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Yunhe Chang
- Department of Orthopedic Trauma, Orthopedic Research Institution of Hebei ProvinceKey Labratory of Biomechanics of Hebei ProvinceThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Xuqing Yang
- Department of Orthopedic Trauma, Orthopedic Research Institution of Hebei ProvinceKey Labratory of Biomechanics of Hebei ProvinceThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Ziwang Han
- Department of Orthopedic Trauma, Orthopedic Research Institution of Hebei ProvinceKey Labratory of Biomechanics of Hebei ProvinceThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
| |
Collapse
|
7
|
Hu H, Long Y, Song G, Chen S, Xu Z, Li Q, Wu Z. Dysfunction of Prkcaa Links Social Behavior Defects with Disturbed Circadian Rhythm in Zebrafish. Int J Mol Sci 2023; 24:ijms24043849. [PMID: 36835261 PMCID: PMC9961154 DOI: 10.3390/ijms24043849] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/17/2023] Open
Abstract
Protein kinase Cα (PKCα/PRKCA) is a crucial regulator of circadian rhythm and is associated with human mental illnesses such as autism spectrum disorders and schizophrenia. However, the roles of PRKCA in modulating animal social behavior and the underlying mechanisms remain to be explored. Here we report the generation and characterization of prkcaa-deficient zebrafish (Danio rerio). The results of behavioral tests indicate that a deficiency in Prkcaa led to anxiety-like behavior and impaired social preference in zebrafish. RNA-sequencing analyses revealed the significant effects of the prkcaa mutation on the expression of the morning-preferring circadian genes. The representatives are the immediate early genes, including egr2a, egr4, fosaa, fosab and npas4a. The downregulation of these genes at night was attenuated by Prkcaa dysfunction. Consistently, the mutants demonstrated reversed day-night locomotor rhythm, which are more active at night than in the morning. Our data show the roles of PRKCA in regulating animal social interactions and link the social behavior defects with a disturbed circadian rhythm.
Collapse
Affiliation(s)
- Han Hu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Research Center of Fishery Resources and Environment, Southwest University, Chongqing 400715, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Yong Long
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- Correspondence: (Y.L.); (Z.W.); Tel.: +86-27-6878-0100 (Y.L.); +86-23-6836-6018 (Z.W.)
| | - Guili Song
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Shaoxiong Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Zhicheng Xu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Research Center of Fishery Resources and Environment, Southwest University, Chongqing 400715, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Qing Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Zhengli Wu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Research Center of Fishery Resources and Environment, Southwest University, Chongqing 400715, China
- Correspondence: (Y.L.); (Z.W.); Tel.: +86-27-6878-0100 (Y.L.); +86-23-6836-6018 (Z.W.)
| |
Collapse
|
8
|
Chen N, Qi Y, Ma X, Xiao X, Liu Q, Xia T, Xiang J, Zeng J, Tang J. Rediscovery of Traditional Plant Medicine: An Underestimated Anticancer Drug of Chelerythrine. Front Pharmacol 2022; 13:906301. [PMID: 35721116 PMCID: PMC9198297 DOI: 10.3389/fphar.2022.906301] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/26/2022] [Indexed: 12/24/2022] Open
Abstract
In many studies, the extensive and significant anticancer activity of chelerythrine (CHE) was identified, which is the primary natural active compound in four traditional botanical drugs and can be applied as a promising treatment in various solid tumors. So this review aimed to summarize the anticancer capacities and the antitumor mechanism of CHE. The literature searches revolving around CHE have been carried out on PubMed, Web of Science, ScienceDirect, and MEDLINE databases. Increasing evidence indicates that CHE, as a benzophenanthridine alkaloid, exhibits its excellent anticancer activity as CHE can intervene in tumor progression and inhibit tumor growth in multiple ways, such as induction of cancer cell apoptosis, cell cycle arrest, prevention of tumor invasion and metastasis, autophagy-mediated cell death, bind selectively to telomeric G-quadruplex and strongly inhibit the telomerase activity through G-quadruplex stabilization, reactive oxygen species (ROS), mitogen-activated protein kinase (MAPK), and PKC. The role of CHE against diverse types of cancers has been investigated in many studies and has been identified as the main antitumor drug candidate in drug discovery programs. The current complex data suggest the potential value in clinical application and the future direction of CHE as a therapeutic drug in cancer. Furthermore, the limitations and the present problems are also highlighted in this review. Despite the unclearly delineated molecular targets of CHE, extensive research in this area provided continuously fresh data exploitable in the clinic while addressing the present requirement for further studies such as toxicological studies, combination medication, and the development of novel chemical methods or biomaterials to extend the effects of CHE or the development of its derivatives and analogs, contributing to the effective transformation of this underestimated anticancer drug into clinical practice. We believe that this review can provide support for the clinical application of a new anticancer drug in the future.
Collapse
Affiliation(s)
- Nianzhi Chen
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yulin Qi
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaolin Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qingsong Liu
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ting Xia
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Juyi Xiang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- Geriatric Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianyuan Tang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
9
|
Hernández-Rojas R, Jiménez-Arellano C, de la Fuente-Granada M, Ordaz-Rosado D, García-Becerra R, Valencia-Mayoral P, Álvarez-Arellano L, Eguía-Aguilar P, Velasco-Velázquez MA, González-Arenas A. The interplay between estrogen receptor beta and protein kinase C, a crucial collaboration for medulloblastoma cell proliferation and invasion. Cell Signal 2022; 92:110246. [PMID: 35033667 DOI: 10.1016/j.cellsig.2022.110246] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/14/2021] [Accepted: 01/10/2022] [Indexed: 11/03/2022]
Abstract
Medulloblastoma (MB) is the most common and aggressive pediatric intracranial tumor. Estrogen receptor β (ERβ) expression correlates with MB development and its phosphorylation modifies its transcriptional activity in a ligand-dependent or independent manner. Using in silico tools, we have identified several residues in ERβ protein as potential targets of protein kinases C (PKCs) α and δ. Using Daoy cells, we observed that PKCα and PKCδ associate with ERβ and induce its phosphorylation. The activation of ERβ promotes MB cells proliferation and invasion, and PKCs downregulation dysregulates these steroid receptor mediated processes. Our data suggest that these kinases may play a crucial role in the regulation of the ERβ transcriptional activity. Overexpression of both PKCα and PKCδ in MB biopsies samples supports their relevance in MB progression.
Collapse
Affiliation(s)
- Rubí Hernández-Rojas
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510 Ciudad de México, México
| | - Carolina Jiménez-Arellano
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510 Ciudad de México, México
| | - Marisol de la Fuente-Granada
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510 Ciudad de México, México
| | - David Ordaz-Rosado
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, 14080 Ciudad de México, México
| | - Rocío García-Becerra
- Programa de Investigación de Cáncer de Mama y Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510 Ciudad de México, México
| | - Pedro Valencia-Mayoral
- Departamento de Patología, Hospital Infantil de México Federico Gómez, 06720 Ciudad de México, México
| | | | - Pilar Eguía-Aguilar
- Laboratorio de Biología Molecular, Departamento de Patología Clínica y Experimental, Hospital Infantil de México Federico Gómez, México
| | - Marco A Velasco-Velázquez
- Laboratorio de Farmacología Molecular, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 Ciudad de México, México
| | - Aliesha González-Arenas
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510 Ciudad de México, México.
| |
Collapse
|
10
|
Kumar A, Kumar Gaur G, Panigrahi M, V N MA, Priya B, Gupta JP, Gandham RK. Comparative gene expression profile in circulating PBMCs of Bos indicus and crossbred cattle to understand disease tolerance mechanism. Anim Biotechnol 2022:1-9. [PMID: 35244514 DOI: 10.1080/10495398.2022.2043883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
The present investigation was performed to compare the global gene expression profile in peripheral blood mononuclear cells (PBMCs) of Bos indicus and crossbred (Bos taurus × B. indicus) cattle. Previously, several studies revealed the disease tolerance potential of B. indicus cattle but underlying genetic mechanism is still not fully explored. The PBMCs model was used for this investigation as it plays crucial role in the immune system regulation. Transcriptomic analysis revealed total 6767 significantly differentially expressed transcripts (fold change (absolute) >2.0, p < .05). In addition, 4149 transcripts were upregulated, 2618 transcripts were downregulated and fold change (absolute) of differentially expressed transcript varied from -223.32 to 213.63. Functional annotation analysis of differentially expressed genes confirmed their role in various molecular pathways viz. innate immune response, antigen processing and presentation, MHC protein complex, defense response to bacterium, regulation of immune response, positive regulation of JAK-STAT cascade, cytoskeletal protein binding, etc. Protein-protein interaction network analysis provided understanding of inter-relationship of immune genes with differentially expressed genes. In conclusion, this study could provide comprehensive information about the dysregulated genes and biological pathways in PBMCs which might be responsible for disease tolerance in B. indicus cattle.
Collapse
Affiliation(s)
- Amod Kumar
- ICAR - National Bureau of Animal Genetic Resources (NBAGR), Karnal, India
| | | | | | | | - Bhuvana Priya
- ICAR - Indian Veterinary Research Institute, Bareilly, India
| | | | | |
Collapse
|
11
|
Domingo-Fernández D, Gadiya Y, Patel A, Mubeen S, Rivas-Barragan D, Diana CW, Misra BB, Healey D, Rokicki J, Colluru V. Causal reasoning over knowledge graphs leveraging drug-perturbed and disease-specific transcriptomic signatures for drug discovery. PLoS Comput Biol 2022; 18:e1009909. [PMID: 35213534 PMCID: PMC8906585 DOI: 10.1371/journal.pcbi.1009909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/09/2022] [Accepted: 02/09/2022] [Indexed: 12/29/2022] Open
Abstract
Network-based approaches are becoming increasingly popular for drug discovery as they provide a systems-level overview of the mechanisms underlying disease pathophysiology. They have demonstrated significant early promise over other methods of biological data representation, such as in target discovery, side effect prediction and drug repurposing. In parallel, an explosion of -omics data for the deep characterization of biological systems routinely uncovers molecular signatures of disease for similar applications. Here, we present RPath, a novel algorithm that prioritizes drugs for a given disease by reasoning over causal paths in a knowledge graph (KG), guided by both drug-perturbed as well as disease-specific transcriptomic signatures. First, our approach identifies the causal paths that connect a drug to a particular disease. Next, it reasons over these paths to identify those that correlate with the transcriptional signatures observed in a drug-perturbation experiment, and anti-correlate to signatures observed in the disease of interest. The paths which match this signature profile are then proposed to represent the mechanism of action of the drug. We demonstrate how RPath consistently prioritizes clinically investigated drug-disease pairs on multiple datasets and KGs, achieving better performance over other similar methodologies. Furthermore, we present two case studies showing how one can deconvolute the predictions made by RPath as well as predict novel targets.
Collapse
Affiliation(s)
| | - Yojana Gadiya
- Enveda Biosciences, Boulder, Colorado, United States of America
| | - Abhishek Patel
- Enveda Biosciences, Boulder, Colorado, United States of America
| | - Sarah Mubeen
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | | | - Chris W. Diana
- Enveda Biosciences, Boulder, Colorado, United States of America
| | | | - David Healey
- Enveda Biosciences, Boulder, Colorado, United States of America
| | - Joe Rokicki
- Enveda Biosciences, Boulder, Colorado, United States of America
| | - Viswa Colluru
- Enveda Biosciences, Boulder, Colorado, United States of America
| |
Collapse
|
12
|
Gou X, Hu T, Gou Y, Li C, Yi M, Jia M. Specific protein kinase C isoform exerts chronic inhibition on the slowly activating delayed-rectifier potassium current by affecting channel trafficking. Channels (Austin) 2021; 15:262-272. [PMID: 33535882 PMCID: PMC7872027 DOI: 10.1080/19336950.2021.1882112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/22/2021] [Accepted: 01/22/2021] [Indexed: 11/04/2022] Open
Abstract
The slowly activating delayed rectifier K+ current (IKs) plays a key role in the repolarization of ventricular action potential in the human heart and is formed by the pore-forming α-subunit encoded by KCNQ1 (Kv7.1) and β-subunit encoded by KCNE1. Evidence suggested that IKs was regulated through protein kinase C (PKC) pathway, but the mechanism is controversial. This study was designed to identify the specific PKC isoform involved in the long-term regulation of IKs current. The IKs current was recorded using whole-cell patch-clamp technique in human embryonic kidney (HEK) 293B cell co-transfected with human KCNQ1/KCNE1 genes. The results revealed that both chronic activation of Ang II and PMA reduced the IKs current in a long-term regulation (about 24 hours). Further evidence showed that PKCε knockdown by siRNA antagonized the AngII-induced chronic inhibition on the IKs current, whereas knockdown of cPKC (PKCα and PKCβ) attenuated the inhibition effect of PMA on the current. Moreover, the forward transport inhibition of the channel with brefeldin A alleviated the Ang II-induced chronic inhibition on IKs current, while the channel endocytosis inhibition with dynasore alleviated both Ang II and PMA-induced chronic inhibition on IKs current. The above results showed that PKCε activation promoted the channel endocytosis and inhibited the channel forward transport to the plasma membrane, while cPKC activation only promoted the channel endocytosis, which both down regulated the channel current.
Collapse
Affiliation(s)
- Xiangbo Gou
- Tianjin Key Labortory of Drug Targeting and Bioimaging, Tianjin University of Technology, Tianjin, China
- School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin, China
| | - Tingting Hu
- Department of Orthopaedic Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
- Department of Neurobiology, School of Basic Medical Science, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yu Gou
- Department of Orthopaedic Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Chaoqi Li
- School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin, China
| | - Ming Yi
- School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin, China
| | - Mengran Jia
- School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin, China
| |
Collapse
|
13
|
Singh RK, Kumar S, Tomar MS, Verma PK, Kumar A, Kumar S, Kumar N, Singh JP, Acharya A. Putative role of natural products as Protein Kinase C modulator in different disease conditions. ACTA ACUST UNITED AC 2021; 29:397-414. [PMID: 34216003 DOI: 10.1007/s40199-021-00401-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 05/25/2021] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Protein kinase C (PKC) is a promising drug target for various therapeutic areas. Natural products derived from plants, animals, microorganisms, and marine organisms have been used by humans as medicine from prehistoric times. Recently, several compounds derived from plants have been found to modulate PKC activities through competitive binding with ATP binding site, and other allosteric regions of PKC. As a result fresh race has been started in academia and pharmaceutical companies to develop an effective naturally derived small-molecule inhibitor to target PKC activities. Herein, in this review, we have discussed several natural products and their derivatives, which are reported to have an impact on PKC signaling cascade. METHODS All information presented in this review article regarding the regulation of PKC by natural products has been acquired by a systematic search of various electronic databases, including ScienceDirect, Scopus, Google Scholar, Web of science, ResearchGate, and PubMed. The keywords PKC, natural products, curcumin, rottlerin, quercetin, ellagic acid, epigallocatechin-3 gallate, ingenol 3 angelate, resveratrol, protocatechuic acid, tannic acid, PKC modulators from marine organism, bryostatin, staurosporine, midostaurin, sangivamycin, and other relevant key words were explored. RESULTS The natural products and their derivatives including curcumin, rottlerin, quercetin, ellagic acid, epigallocatechin-3 gallate, ingenol 3 angelate, resveratrol, bryostatin, staurosporine, and midostaurin play a major role in the management of PKC activity during various disease progression. CONCLUSION Based on the comprehensive literature survey, it could be concluded that various natural products can regulate PKC activity during disease progression. However, extensive research is needed to circumvent the challenge of isoform specific regulation of PKC by natural products.
Collapse
Affiliation(s)
- Rishi Kant Singh
- Department of Zoology, Institute of Science, BHU, Varanasi, 221005, India
| | | | - Munendra Singh Tomar
- Department of Pharmaceutical Science, School of Pharmacy, University of Colorado, Denver, USA
| | | | - Amit Kumar
- Department of Zoology, Institute of Science, BHU, Varanasi, 221005, India
| | - Sandeep Kumar
- Department of Zoology, Institute of Science, BHU, Varanasi, 221005, India
| | - Naveen Kumar
- Department of Zoology, Institute of Science, BHU, Varanasi, 221005, India
| | - Jai Prakash Singh
- Department of Panchkarma, Institute of Medical Science, BHU, Varanasi, India, 221005
| | - Arbind Acharya
- Department of Zoology, Institute of Science, BHU, Varanasi, 221005, India.
| |
Collapse
|
14
|
Qian XH, Song XX, Liu XL, Chen SD, Tang HD. Inflammatory pathways in Alzheimer's disease mediated by gut microbiota. Ageing Res Rev 2021; 68:101317. [PMID: 33711509 DOI: 10.1016/j.arr.2021.101317] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/25/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022]
Abstract
In the past decade, numerous studies have demonstrated the close relationship between gut microbiota and the occurrence and development of Alzheimer's disease (AD). However, the specific mechanism is still unclear. Both the neuroinflammation and systemic inflammation serve as the key hubs to accelerate the process of AD by promoting pathology and damaging neuron. What's more, the gut microbiota is also crucial for the regulation of inflammation. Therefore, this review focused on the role of gut microbiota in AD through inflammatory pathways. Firstly, this review summarized the relationship and interaction among gut microbiota, inflammation, and AD. Secondly, the direct and indirect regulatory effects of gut microbiota on AD through inflammatory pathways were described. These effects were mainly mediated by the component of the gut microbiota (lipopolysaccharides (LPS) and amyloid peptides), the metabolites of bacteria (short-chain fatty acids, branched amino acids, and neurotransmitters) and functional by-products (bile acids). In addition, potential treatments (fecal microbiota transplantation, antibiotics, probiotics, prebiotics, and dietary interventions) for AD were also discussed through these mechanisms. Finally, according to the current research status, the key problems to be solved in the future studies were proposed.
Collapse
Affiliation(s)
- Xiao-Hang Qian
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xiao-Xuan Song
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xiao-Li Liu
- Department of Neurology, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, 201406, China.
| | - Sheng-di Chen
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hui-Dong Tang
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
15
|
Kim CW, Toita R, Kang JH, Mori T, Kishimura A, Katayama Y. Protein Kinase C α-Responsive Gene Carrier for Cancer-Specific Transgene Expression and Cancer Therapy. ACS Biomater Sci Eng 2021; 7:2530-2537. [PMID: 33890761 DOI: 10.1021/acsbiomaterials.1c00213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The presence of intracellular signal transduction and its abnormal activities in many cancers has potential for medical and pharmaceutical applications. We recently developed a protein kinase C α (PKCα)-responsive gene carrier for cancer-specific gene delivery. Here, we demonstrate an in-depth analysis of cellular signal-responsive gene carrier and the impact of its selective transgene expression in response to malfunctioning intracellular signaling in cancer cells. We prepared a novel gene carrier consisting of a linear polyethylenimine (LPEI) main chain grafted to a cationic PKCα-specific substrate (FKKQGSFAKKK-NH2). The LPEI-peptide conjugate formed a nanosized polyplex with pDNA and mediated efficient cellular uptake and endosomal escape. This polyplex also led to successful transgene expression which responded to the target PKCα in various cancer cells and exhibited a 10-100-fold higher efficiency compared to the control group. In xenograft tumor models, the LPEI-peptide conjugate promoted transgene expression showing a clear-cut response to PKCα. Furthermore, when a plasmid containing a therapeutic gene, human caspase-8 (pcDNA-hcasp8), was used, the LPEI-peptide conjugate had significant cancer-suppressive effects and extended animal survival. Collectively, these results reveal that our method has great potential for cancer-specific gene delivery and therapy.
Collapse
Affiliation(s)
- Chan Woo Kim
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Riki Toita
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan.,AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, AIST, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Jeong-Hun Kang
- Division of Biopharmaceutics and Pharmacokinetics, National Cerebral and Cardiovascular Center Research Institute, 6-1 Shinmachi, Kishibe, Suita, Osaka 564-8565, Japan
| | - Takeshi Mori
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Akihiro Kishimura
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,International Research Center for Molecular Systems, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yoshiki Katayama
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,International Research Center for Molecular Systems, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.,Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| |
Collapse
|
16
|
Lin HY, Wang X, Tseng TS, Kao YH, Fang Z, Molina PE, Cheng CH, Berglund AE, Eeles RA, Muir KR, Pashayan N, Haiman CA, Brenner H, Consortium TP, Park JY. Alcohol Intake and Alcohol-SNP Interactions Associated with Prostate Cancer Aggressiveness. J Clin Med 2021; 10:553. [PMID: 33540941 PMCID: PMC7867322 DOI: 10.3390/jcm10030553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/25/2021] [Accepted: 01/28/2021] [Indexed: 12/24/2022] Open
Abstract
Excessive alcohol intake is a well-known modifiable risk factor for many cancers. It is still unclear whether genetic variants or single nucleotide polymorphisms (SNPs) can modify alcohol intake's impact on prostate cancer (PCa) aggressiveness. The objective is to test the alcohol-SNP interactions of the 7501 SNPs in the four pathways (angiogenesis, mitochondria, miRNA, and androgen metabolism-related pathways) associated with PCa aggressiveness. We evaluated the impacts of three excessive alcohol intake behaviors in 3306 PCa patients with European ancestry from the PCa Consortium. We tested the alcohol-SNP interactions using logistic models with the discovery-validation study design. All three excessive alcohol intake behaviors were not significantly associated with PCa aggressiveness. However, the interactions of excessive alcohol intake and three SNPs (rs13107662 [CAMK2D, p = 6.2 × 10-6], rs9907521 [PRKCA, p = 7.1 × 10-5], and rs11925452 [ROBO1, p = 8.2 × 10-4]) were significantly associated with PCa aggressiveness. These alcohol-SNP interactions revealed contrasting effects of excessive alcohol intake on PCa aggressiveness according to the genotypes in the identified SNPs. We identified PCa patients with the rs13107662 (CAMK2D) AA genotype, the rs11925452 (ROBO1) AA genotype, and the rs9907521 (PRKCA) AG genotype were more vulnerable to excessive alcohol intake for developing aggressive PCa. Our findings support that the impact of excessive alcohol intake on PCa aggressiveness was varied by the selected genetic profiles.
Collapse
Affiliation(s)
- Hui-Yi Lin
- Biostatistics Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Xinnan Wang
- Biostatistics Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Tung-Sung Tseng
- Behavioral and Community Health Sciences Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Yu-Hsiang Kao
- Behavioral and Community Health Sciences Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Zhide Fang
- Biostatistics Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Patricia E Molina
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Comprehensive Alcohol Research Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Chia-Ho Cheng
- Biostatistics and Bioinformatics Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Anders E Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Rosalind A Eeles
- The Institute of Cancer Research, and The Royal Marsden NHS Foundation Trust, London, SM2 5NG, UK
| | - Kenneth R Muir
- Division of Population Health, Health Services Research, and Primary Care, University of Manchester, Oxford Road, Manchester, M139PT, UK
| | - Nora Pashayan
- Department of Applied Health Research, University College London, WC1E 7HB, London, UK
| | - Christopher A Haiman
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA 90015, USA
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), D-69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - The Practical Consortium
- The Prostate Cancer Association Group to Investigate Cancer Associated Alterations in the Genome Consortium (PRACTICAL, http://practical.icr.ac.uk/), London SM2 5NG, UK. Additional members from The PRACTICAL Consortium were provided in the Supplement
| | - Jong Y Park
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
17
|
Ibarra LE, Vilchez ML, Caverzán MD, Milla Sanabria LN. Understanding the glioblastoma tumor biology to optimize photodynamic therapy: From molecular to cellular events. J Neurosci Res 2020; 99:1024-1047. [PMID: 33370846 DOI: 10.1002/jnr.24776] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 11/29/2020] [Indexed: 12/19/2022]
Abstract
Photodynamic therapy (PDT) has recently gained attention as an alternative treatment of malignant gliomas. Glioblastoma (GBM) is the most prevalent within tumors of the central nervous system (CNS). Conventional treatments for this CNS tumor include surgery, radiation, and chemotherapy. Surgery is still being considered as the treatment of choice. Even so, the poor prognosis and/or recurrence of the disease after applying any of these treatments highlight the urgency of exploring new therapies and/or improving existing ones to achieve the definitive eradication of tumor masses and remaining cells. PDT is a therapeutic modality that involves the destruction of tumor cells by reactive oxygen species induced by light, which were previously treated with a photosensitizing agent. However, in recent years, its experimental application has expanded to other effects that could improve overall performance against GBM. In the current review, we revisit the main advances of PDT for GBM management and also, the recent mechanistic insights about cellular and molecular aspects related to tumoral resistance to PDT of GBM.
Collapse
Affiliation(s)
- Luis Exequiel Ibarra
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Río Cuarto (UNRC) y Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Río Cuarto, Argentina.,Departamento de Biología Molecular, Facultad de Ciencias Exactas Físico-Químicas y Naturales, UNRC, Río Cuarto, Argentina
| | - María Laura Vilchez
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Río Cuarto (UNRC) y Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Río Cuarto, Argentina.,Departamento de Biología Molecular, Facultad de Ciencias Exactas Físico-Químicas y Naturales, UNRC, Río Cuarto, Argentina
| | - Matías Daniel Caverzán
- Departamento de Biología Molecular, Facultad de Ciencias Exactas Físico-Químicas y Naturales, UNRC, Río Cuarto, Argentina
| | - Laura Natalia Milla Sanabria
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Río Cuarto (UNRC) y Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Río Cuarto, Argentina.,Departamento de Biología Molecular, Facultad de Ciencias Exactas Físico-Químicas y Naturales, UNRC, Río Cuarto, Argentina
| |
Collapse
|
18
|
Keleher MR, Erickson K, Kechris K, Yang IV, Dabelea D, Friedman JE, Boyle KE, Jansson T. Associations between the activity of placental nutrient-sensing pathways and neonatal and postnatal metabolic health: the ECHO Healthy Start cohort. Int J Obes (Lond) 2020; 44:2203-2212. [PMID: 32327723 PMCID: PMC8329931 DOI: 10.1038/s41366-020-0574-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 03/10/2020] [Accepted: 03/27/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Our hypothesis was that the activity of placental nutrient-sensing pathways is associated with adiposity and metabolic health in childhood. RESEARCH DESIGN AND METHODS Using placental villus samples from healthy mothers from the Healthy Start Study, we measured the abundance and phosphorylation of key intermediates in the mTOR, insulin, AMPK, and ER stress signaling pathways. Using multivariate multiple regression models, we tested the association between placental proteins and offspring adiposity (%fat mass) at birth (n = 109), 4-6 months (n = 104), and 4-6 years old (n = 64), adjusted for offspring sex and age. RESULTS Placental mTORC1 phosphorylation was positively associated with adiposity at birth (R2 = 0.13, P = 0.009) and 4-6 years (R2 = 0.15, P = 0.046). The mTORC2 target PKCα was positively associated with systolic blood pressure at 4-6 years (β = 2.90, P = 0.005). AMPK phosphorylation was positively associated with adiposity at birth (β = 2.32, P = 0.023), but the ratio of phosphorylated to total AMPK was negatively associated with skinfold thickness (β = -2.37, P = 0.022) and body weight (β = -2.92, P = 0.005) at 4-6 years. CONCLUSIONS This is the first report of associations between key placental protein activity measures and longitudinal child outcomes at various life stages. Our data indicate that AMPK and mTOR signaling are linked to cardiometabolic measures at birth and 4-6 years, providing novel insight into potential mechanisms underpinning how metabolic signaling in the placenta is associated with future risk of cardiovascular disease.
Collapse
Affiliation(s)
- Madeline Rose Keleher
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO, USA.
| | - Kathryn Erickson
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katerina Kechris
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO, USA
- Department of Biostatistics & Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - Ivana V Yang
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Dana Dabelea
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO, USA
- Department of Epidemiology, Colorado School of Public Health, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jacob E Friedman
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kristen E Boyle
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO, USA
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
19
|
A metabolic reaction-diffusion model for PKCα translocation via PIP2 hydrolysis in an endothelial cell. Biochem J 2020; 477:4071-4084. [PMID: 33026061 DOI: 10.1042/bcj20200484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/08/2020] [Accepted: 10/06/2020] [Indexed: 11/17/2022]
Abstract
Hydrolysis of the phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2) at the cell membrane induces the release of inositol 1,4,5-trisphosphate (IP3) into the cytoplasm and diffusion of diacylglycerol (DAG) through the membrane, respectively. Release of IP3 subsequently increases Ca2+ levels in the cytoplasm, which results in activation of protein kinase C α (PKCα) by Ca2+ and DAG, and finally the translocation of PKCα from the cytoplasm to the membrane. In this study, we developed a metabolic reaction-diffusion framework to simulate PKCα translocation via PIP2 hydrolysis in an endothelial cell. A three-dimensional cell model, divided into membrane and cytoplasm domains, was reconstructed from confocal microscopy images. The associated metabolic reactions were divided into their corresponding domain; PIP2 hydrolysis at the membrane domain resulted in DAG diffusion at the membrane domain and IP3 release into the cytoplasm domain. In the cytoplasm domain, Ca2+ was released from the endoplasmic reticulum, and IP3, Ca2+, and PKCα diffused through the cytoplasm. PKCα bound Ca2+ at, and diffused through, the cytoplasm, and was finally activated by binding with DAG at the membrane. Using our model, we analyzed IP3 and DAG dynamics, Ca2+ waves, and PKCα translocation in response to a microscopic stimulus. We found a qualitative agreement between our simulation results and our experimental results obtained by live-cell imaging. Interestingly, our results suggest that PKCα translocation is dominated by DAG dynamics. This three-dimensional reaction-diffusion mathematical framework could be used to investigate the link between PKCα activation in a cell and cell function.
Collapse
|
20
|
Zhao YY, Huang SX, Hao Z, Zhu HX, Xing ZL, Li MH. Fluid Shear Stress Induces Endothelial Cell Injury via Protein Kinase C Alpha-Mediated Repression of p120-Catenin and Vascular Endothelial Cadherin In Vitro. World Neurosurg 2020; 136:e469-e475. [PMID: 31953100 DOI: 10.1016/j.wneu.2020.01.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 01/04/2020] [Accepted: 01/06/2020] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The present study aimed to characterize the mechanism of fluid shear stress (FSS)-induced endothelial cell (EC) injury via protein kinase C alpha (PKCα)-mediated vascular endothelial cadherin (VE-cadherin) and p120-catenin (p120ctn) expression. METHODS We designed a T chamber system that produced stable FSS on ECs in vitro. Human umbilical vein endothelial cells (HUVECs) in which PKCα was knocked down and normal HUVECs were cultured on the coverslips. FSS was impinged on these 2 types of ECs for 0 hours and 6 hours. The morphology and density of HUVECs were evaluated, and expression levels of phosphorylated PKCα, p120-catenin (p120ctn), VE-cadherin, phosphorylated p120ctn at S879 (p-S879p120ctn), and nuclear factor kappa B (NF-κB) were analyzed by Western blot. RESULTS HUVECs exposed to FSS were characterized by a polygonal shape and decreased cell density. The phosphorylated PKCα level was increased under FSS at 6 hours (P < 0.05). In normal HUVECs during FSS, p120ctn and VE-cadherin were decreased, whereas p-S879p120ctn and NF-κB were increased, at 6 hours (P < 0.05). In HUVECs after PKCα knockdown, p120ctn and VE-cadherin were not significantly changed (P > 0.05), p-S879p120ctn was undetectable, but NF-κB was decreased (P < 0.05) at 6 hours. CONCLUSIONS The possible mechanism of FSS-induced EC injury may be as follows: 1) PKCα induces low expression of p120ctn, which leads to activation of NF-κB and degradation of VE-cadherin; 2) PKCα-mediated phosphorylation of p120ctn at S879 disrupts p120ctn binding to VE-cadherin.
Collapse
Affiliation(s)
- Ye-Yu Zhao
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shao-Xin Huang
- College of Basic Medicine, Jiujiang University, Jiujiang, China
| | - Zheng Hao
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hua-Xin Zhu
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ze-Long Xing
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Mei-Hua Li
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
21
|
Jiang W, Xiao T, Han W, Xiong J, He T, Liu Y, Huang Y, Yang K, Bi X, Xu X, Yu Y, Li Y, Gu J, Zhang J, Huang Y, Zhang B, Zhao J. Klotho inhibits PKCα/p66SHC-mediated podocyte injury in diabetic nephropathy. Mol Cell Endocrinol 2019; 494:110490. [PMID: 31207271 DOI: 10.1016/j.mce.2019.110490] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/12/2019] [Accepted: 06/12/2019] [Indexed: 10/26/2022]
Abstract
Diabetic nephropathy (DN) is a progressive disease, the main pathogeny of which is podocyte injury. As a calcium-dependent serine/threonine protein kinase involved in podocyte injury, protein kinase C isoform α (PKCα) was reported to regulate the phosphorylation of p66SHC. However, the role of PKCα/p66SHC in DN remains unknown. Klotho, an anti-aging protein with critical roles in protecting kidney, is expressed predominantly in the kidney and secreted in the blood. Nonetheless, the mechanism underlying amelioration of podocyte injury by Klotho in DN remains unclear. Our data showed that Klotho was decreased in STZ-treated mice and was further declined in diabetic KL ± mice. As expected, Klotho deficiency aggravated diabetes-induced proteinuria and podocyte injury, accompanied by the activation of PKCα and p66SHC. In contrast, overexpression of Klotho partially ameliorated PKCα/p66SHC-mediated podocyte injury and proteinuria. In addition, in vitro experiments showed that activation of PKCα and subsequently increased intracellular reactive oxygen species (ROS) was involved in podocytic apoptosis induced by high glucose (HG), which could be partially reversed by Klotho. Hence, we conclude that Klotho might inhibit PKCα/p66SHC-mediated podocyte injury in diabetic nephropathy.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Tangli Xiao
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Wenhao Han
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jiachuan Xiong
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Ting He
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yong Liu
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yinghui Huang
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Ke Yang
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xianjin Bi
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xinli Xu
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yanlin Yu
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yan Li
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jun Gu
- State Key Laboratory of Protein and Plant Gene Research, College of Life Science, Peking University, Beijing, China
| | - Jingbo Zhang
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yunjian Huang
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Bo Zhang
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| | - Jinghong Zhao
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
22
|
Geribaldi-Doldán N, Gómez-Oliva R, Domínguez-García S, Nunez-Abades P, Castro C. Protein Kinase C: Targets to Regenerate Brain Injuries? Front Cell Dev Biol 2019; 7:39. [PMID: 30949480 PMCID: PMC6435489 DOI: 10.3389/fcell.2019.00039] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/04/2019] [Indexed: 12/28/2022] Open
Abstract
Acute or chronic injury to the central nervous system (CNS), causes neuronal death and irreversible cognitive deficits or sensory-motor alteration. Despite the capacity of the adult CNS to generate new neurons from neural stem cells (NSC), neuronal replacement following an injury is a restricted process, which does not naturally result in functional regeneration. Therefore, potentiating endogenous neurogenesis is one of the strategies that are currently being under study to regenerate damaged brain tissue. The insignificant neurogenesis that occurs in CNS injuries is a consequence of the gliogenic/non-neurogenic environment that inflammatory signaling molecules create within the injured area. The modification of the extracellular signals to generate a neurogenic environment would facilitate neuronal replacement. However, in order to generate this environment, it is necessary to unearth which molecules promote or impair neurogenesis to introduce the first and/or eliminate the latter. Specific isozymes of the protein kinase C (PKC) family differentially contribute to generate a gliogenic or neurogenic environment in injuries by regulating the ADAM17 mediated release of growth factor receptor ligands. Recent reports describe several non-tumorigenic diterpenes isolated from plants of the Euphorbia genus, which specifically modulate the activity of PKC isozymes promoting neurogenesis. Diterpenes with 12-deoxyphorbol or lathyrane skeleton, increase NPC proliferation in neurogenic niches in the adult mouse brain in a PKCβ dependent manner exerting their effects on transit amplifying cells, whereas PKC inhibition in injuries promotes neurogenesis. Thus, compounds that balance PKC activity in injuries might be of use in the development of new drugs and therapeutic strategies to regenerate brain injuries.
Collapse
Affiliation(s)
- Noelia Geribaldi-Doldán
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomedica de Cádiz (INIBICA), Cádiz, Spain
| | - Ricardo Gómez-Oliva
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomedica de Cádiz (INIBICA), Cádiz, Spain
| | - Samuel Domínguez-García
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomedica de Cádiz (INIBICA), Cádiz, Spain
| | - Pedro Nunez-Abades
- Instituto de Investigación e Innovación Biomedica de Cádiz (INIBICA), Cádiz, Spain.,Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
| | - Carmen Castro
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomedica de Cádiz (INIBICA), Cádiz, Spain
| |
Collapse
|
23
|
Parks XX, Ronzier E, O-Uchi J, Lopes CM. Fluvastatin inhibits Rab5-mediated IKs internalization caused by chronic Ca 2+-dependent PKC activation. J Mol Cell Cardiol 2019; 129:314-325. [PMID: 30898664 DOI: 10.1016/j.yjmcc.2019.03.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 02/26/2019] [Accepted: 03/16/2019] [Indexed: 10/27/2022]
Abstract
Statins, in addition to their cholesterol lowering effects, can prevent isoprenylation of Rab GTPase proteins, a key protein family for the regulation of protein trafficking. Rab-GTPases have been shown to be involved in the control of membrane expression level of ion channels, including one of the major cardiac repolarizing channels, IKs. Decreased IKs function has been observed in a number of disease states and associated with increased propensity for arrhythmias, but the mechanism underlying IKs decrease remains elusive. Ca2+-dependent PKC isoforms (cPKC) are chronically activated in variety of human diseases and have been suggested to acutely regulate IKs function. We hypothesize that chronic cPKC stimulation leads to Rab-mediated decrease in IKs membrane expression, and that can be prevented by statins. In this study we show that chronic cPKC stimulation caused a dramatic Rab5 GTPase-dependent decrease in plasma membrane localization of the IKs pore forming subunit KCNQ1, reducing IKs function. Our data indicates fluvastatin inhibition of Rab5 restores channel localization and function after cPKC-mediated channel internalization. Our results indicate a novel statin anti-arrhythmic effect that would be expected to inhibit pathological electrical remodeling in a number of disease states associated with high cPKC activation. Because Rab-GTPases are important regulators of membrane trafficking they may underlie other statin pleiotropic effects.
Collapse
Affiliation(s)
- Xiaorong Xu Parks
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
| | - Elsa Ronzier
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
| | - Jin O-Uchi
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America; Lillehei Heart Institute, University of Minnesota, 2231 6th Street SE, Minneapolis, MN 55455, United States of America
| | - Coeli M Lopes
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America.
| |
Collapse
|
24
|
Liu CH, Hua N, Fu X, Pan YL, Li B, Li XD. Metformin regulates atrial SK2 and SK3 expression through inhibiting the PKC/ERK signaling pathway in type 2 diabetic rats. BMC Cardiovasc Disord 2018; 18:236. [PMID: 30545309 PMCID: PMC6293565 DOI: 10.1186/s12872-018-0950-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 11/09/2018] [Indexed: 01/01/2023] Open
Abstract
Background Our previous study showed that metformin regulates the mRNA and protein levels of type 2 small conductance calcium-activated potassium channel (SK2) and type 3 small conductance calcium-activated potassium channels (SK3) in atrial tissue as well as the ion current of atrial myocytes in rats with type 2 diabetes mellitus (T2DM), but the underlying signaling mechanism is unknown. This study aimed to investigate whether metformin regulates atrial SK2 and SK3 protein expression in T2DM rats though the protein kinase C (PKC)/extracellular signal-regulated kinase (ERK) signaling pathway. Methods A T2DM rat model was established using a high-fat and high-sugar diet combined with a low-dose intraperitoneal injection of streptozotocin (STZ). The rats were randomly divided into the following five groups: the control group, the untreated T2DM group, the metformin-treated only group, the phorbol 12-myristate 13-acetate (PMA; a PKC agonist administered by intraperitoneal injection) treatment group, and the recombinant human epidermal growth factor (rh-EGF; an ERK agonist administered by tail vein injection) treatment group. The activity of PKC in atrial tissues was assayed by a PKC kinase activity assay kit. The protein expression of SK2, SK3, and phosphorylated ERK (pERK) were determined by western blotting and immunohistochemistry. Results Compared with the Control group, atrial PKC activity and pERK and SK3 protein expression were increased, while SK2 protein expression was decreased in atrial tissues of T2DM rats. Eight weeks of metformin treatment inhibited the PKC activity and pERK and SK3 expression, and elevated SK2 expression compared with the T2DM group. Compared with the metformin-treated only group, the injection of rh-EGF increased pERK and SK3 expression, and decreased SK2 expression; the injection of PMA increased PKC activity and SK3 expression, and decreased SK2 expression. In addition, the injection with PMA significantly elevated the expression of pERK. Conclusions The PKC/ERK signaling pathway is involved in the downregulation of SK2 expression and the upregulation of SK3 expression in the atrium of T2DM rats. Long-term metformin treatment prevents the SK2 downregulation and the SK3 upregulation through inhibiting the PKC/ERK signaling pathway. Electronic supplementary material The online version of this article (10.1186/s12872-018-0950-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chang-He Liu
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Na Hua
- Department of Otolaryngology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Xi Fu
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Yi-Long Pan
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Bin Li
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Xiao-Dong Li
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
25
|
Markovsky E, de Stanchina E, Itzkowitz A, Haimovitz-Friedman A, Rotenberg SA. Phosphorylation state of Ser 165 in α-tubulin is a toggle switch that controls proliferating human breast tumors. Cell Signal 2018; 52:74-82. [PMID: 30176291 PMCID: PMC6765385 DOI: 10.1016/j.cellsig.2018.08.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/29/2018] [Accepted: 08/31/2018] [Indexed: 11/18/2022]
Abstract
Engineered overexpression of protein kinase Cα (PKCα) is known to phosphorylate Ser165 in α-tubulin resulting in stimulated microtubule dynamics and cell motility, and activation of an epithelial-mesenchymal transition (EMT) in non-transformed human breast cells. Here it is shown that endogenous phosphorylation of native α-tubulin in two metastatic breast cell lines, MDA-MB-231-LM2-4175 and MDA-MB-468 is detected at PKC phosphorylation sites. α-Tubulin mutants that simulated phosphorylated (S165D) or non-phosphorylated (S165 N) states were stably expressed in MDA-MB-231-LM2-4175 cells. The S165D-α-tubulin mutant engendered expression of the EMT biomarker N-cadherin, whereas S165 N-α-tubulin suppressed N-cadherin and induced E-cadherin expression, revealing a 'cadherin switch'. S165 N-α-tubulin engendered more rapid passage through the cell cycle, induced shorter spindle fibers and exhibited more rapid proliferation. In nude mice injected with MDA-MB-231-LM2-4175 cells, cells expressing S165 N-α-tubulin (but not the S165D mutant) produced hyper-proliferative lung tumors with increased tumor incidence and higher Ki67 expression. These results implicate the phosphorylation state of Ser165 in α-tubulin as a PKC-regulated molecular switch that causes breast cells to exhibit either EMT characteristics or hyper-proliferation. Evaluation of genomic databases of human tumors strengthens the clinical significance of these findings.
Collapse
Affiliation(s)
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility of Memorial Sloan-Kettering Cancer Institute, USA
| | | | | | - Susan A Rotenberg
- Department of Chemistry & Biochemistry, Queens College, USA; Graduate Center of The City University of New York, USA.
| |
Collapse
|
26
|
Sera T, Arai M, Cui Z, Onose K, Karimi A, Kudo S. Unloading of intercellular tension induces the directional translocation of PKCα. J Cell Physiol 2018; 234:9764-9777. [PMID: 30387146 DOI: 10.1002/jcp.27662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/02/2018] [Indexed: 12/13/2022]
Abstract
The migration of endothelial cells (ECs) is closely associated with a Ca2+ -dependent protein, protein kinase Cα (PKCα). The disruption of intercellular adhesion by single-cell wounding has been shown to induce the directional translocation of PKCα. We hypothesized that this translocation of PKCα is induced by mechanical stress, such as unloading of intercellular tension, or by intercellular communication, such as gap junction-mediated and paracrine signaling. In the current study, we found that the disruption of intercellular adhesion induced the directional translocation of PKCα even when gap junction-mediated and paracrine signaling were inhibited. Conversely, it did not occur when the mechanosensitive channel was inhibited. In addition, the strain field of substrate attributable to the disruption of intercellular adhesion tended to be larger at the areas corresponding with PKCα translocation. Recently, we found that a direct mechanical stimulus induced the accumulation of PKCα at the stimulus area, involving Ca 2+ influx from extracellular space. These results indicated that the unloading of intercellular tension induced directional translocation of PKCα, which required Ca 2+ influx from extracellular space. The results of this study indicate the involvement of PKCα in the Ca 2+ signaling pathway in response to mechanical stress in ECs.
Collapse
Affiliation(s)
- Toshihiro Sera
- Department of Mechanical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Masataka Arai
- Department of Mechanical Engineering, Graduate School of Engineering, Kyushu University, Fukuoka, Japan
| | - Zhonghua Cui
- Department of Mechanical Engineering, Graduate School of Engineering, Kyushu University, Fukuoka, Japan
| | - Koichi Onose
- Department of Mechanical Engineering, Graduate School of Engineering, Kyushu University, Fukuoka, Japan
| | - Alireza Karimi
- International Research Fellow, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Susumu Kudo
- Department of Mechanical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| |
Collapse
|
27
|
Bonvini A, Coqueiro AY, Tirapegui J, Calder PC, Rogero MM. Immunomodulatory role of branched-chain amino acids. Nutr Rev 2018; 76:840-856. [PMID: 30124936 DOI: 10.1093/nutrit/nuy037] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Branched-chain amino acids (BCAAs) have been associated with immunomodulation since the mid-1970s and 1980s and have been used in the nutritional therapy of critically ill patients. Evidence shows that BCAAs can directly contribute to immune cell function, aiding recovery of an impaired immune system, as well as improving the nutritional status in cancer and liver diseases. Branched-chain amino acids may also play a role in treatment of patients with sepsis or trauma, contributing to improved clinical outcomes and survival. Branched-chain amino acids, especially leucine, are activators of the mammalian target of rapamycin (mTOR), which, in turn, interacts with several signaling pathways involved in biological mechanisms of insulin action, protein synthesis, mitochondrial biogenesis, inflammation, and lipid metabolism. Although many in vitro and human and animal model studies have provided evidence for the biological activity of BCAAs, findings have been conflicting, and the mechanisms of action of these amino acids are still poorly understood. This review addresses several aspects related to BCAAs, including their transport, oxidation, and mechanisms of action, as well as their role in nutritional therapy and immunomodulation.
Collapse
Affiliation(s)
- Andrea Bonvini
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Audrey Y Coqueiro
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Julio Tirapegui
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Philip C Calder
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, United Kingdom
| | - Marcelo M Rogero
- Department of Nutrition, Faculty of Public Health, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
28
|
Malik N, Sansom OJ, Michie AM. The role of mTOR-mediated signals during haemopoiesis and lineage commitment. Biochem Soc Trans 2018; 46:1313-1324. [PMID: 30154096 PMCID: PMC6195642 DOI: 10.1042/bst20180141] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
Abstract
The serine/threonine protein kinase mechanistic target of rapamycin (mTOR) has been implicated in the regulation of an array of cellular functions including protein and lipid synthesis, proliferation, cell size and survival. Here, we describe the role of mTOR during haemopoiesis within the context of mTORC1 and mTORC2, the distinct complexes in which it functions. The use of conditional transgenic mouse models specifically targeting individual mTOR signalling components, together with selective inhibitors, have generated a significant body of research emphasising the critical roles played by mTOR, and individual mTOR complexes, in haemopoietic lineage commitment and development. This review will describe the profound role of mTOR in embryogenesis and haemopoiesis, underscoring the importance of mTORC1 at the early stages of haemopoietic cell development, through modulation of stem cell potentiation and self-renewal, and erythroid and B cell lineage commitment. Furthermore, the relatively discrete role of mTORC2 in haemopoiesis will be explored during T cell development and B cell maturation. Collectively, this review aims to highlight the functional diversity of mTOR signalling and underline the importance of this pathway in haemopoiesis.
Collapse
Affiliation(s)
- Natasha Malik
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, U.K
| | - Owen J Sansom
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, U.K
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, U.K
| | - Alison M Michie
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, U.K.
| |
Collapse
|
29
|
Rosenberg S, Simeonova I, Bielle F, Verreault M, Bance B, Le Roux I, Daniau M, Nadaradjane A, Gleize V, Paris S, Marie Y, Giry M, Polivka M, Figarella-Branger D, Aubriot-Lorton MH, Villa C, Vasiljevic A, Lechapt-Zalcman E, Kalamarides M, Sharif A, Mokhtari K, Pagnotta SM, Iavarone A, Lasorella A, Huillard E, Sanson M. A recurrent point mutation in PRKCA is a hallmark of chordoid gliomas. Nat Commun 2018; 9:2371. [PMID: 29915258 PMCID: PMC6006150 DOI: 10.1038/s41467-018-04622-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 05/14/2018] [Indexed: 12/31/2022] Open
Abstract
Chordoid glioma (ChG) is a characteristic, slow growing, and well-circumscribed diencephalic tumor, whose mutational landscape is unknown. Here we report the analysis of 16 ChG by whole-exome and RNA-sequencing. We found that 15 ChG harbor the same PRKCAD463H mutation. PRKCA encodes the Protein kinase C (PKC) isozyme alpha (PKCα) and is mutated in a wide range of human cancers. However the hot spot PRKCAD463H mutation was not described in other tumors. PRKCAD463H is strongly associated with the activation of protein translation initiation (EIF2) pathway. PKCαD463H mRNA levels are more abundant than wild-type PKCα transcripts, while PKCαD463H is less stable than the PCKαWT protein. Compared to PCKαWT, the PKCαD463H protein is depleted from the cell membrane. The PKCαD463H mutant enhances proliferation of astrocytes and tanycytes, the cells of origin of ChG. In conclusion, our study identifies the hallmark mutation for chordoid gliomas and provides mechanistic insights on ChG oncogenesis. Chordoid glioma is a slow growing diencephalic tumor whose mutational landscape is poorly characterized. Here, the authors perform whole-exome and RNA-sequencing and find that 15 of 16 chordoid glioma cases studied harbor the same PRKCA mutation which results in enhanced proliferation.
Collapse
Affiliation(s)
- Shai Rosenberg
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France.,Gaffin Center for Neuro-oncology, Sharett Institute for Oncology, Hadassah - Hebrew University Medical Center, 91120, Jerusalem, Israel
| | - Iva Simeonova
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France
| | - Franck Bielle
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France.,Laboratoire R Escourolle, AP-HP, Hôpital de la Pitié-Salpêtrière, F-75013, Paris, France
| | - Maite Verreault
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France
| | - Bertille Bance
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France
| | - Isabelle Le Roux
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France
| | - Mailys Daniau
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France
| | - Arun Nadaradjane
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France
| | - Vincent Gleize
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France
| | - Sophie Paris
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France
| | - Yannick Marie
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France.,Onconeurotek Tumor Bank, Institut du Cerveau et de la Moelle épinère-ICM, F-75013, Paris, France
| | - Marine Giry
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France
| | - Marc Polivka
- Department of Pathology, AP-HP, Hôpital Lariboisière, F-75010, Paris, France
| | - Dominique Figarella-Branger
- Pathology and Neuropathology Department, Assistance Publique-Hôpitaux de Marseille (AP-HM), CHU Timone, 13005, Marseille, France
| | | | - Chiara Villa
- Department of Pathological Cytology and Anatomy, Foch Hospital, Suresnes, F-92151, Paris, France
| | - Alexandre Vasiljevic
- Centre de Biologie et Pathologie Est, Groupement Hospitalier Est, Hospices Civils de Lyon, 69500, Bron, France
| | - Emmanuèle Lechapt-Zalcman
- Department of Pathology, CHU de Caen, Caen, France Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP Group, 14000, Caen, France
| | - Michel Kalamarides
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France.,Service de Neurochirurgie, AP-HP, Hôpital de la Pitié-Salpêtrière, F-75013, Paris, France
| | - Ariane Sharif
- INSERM U1172, "Development and Plasticity of the Neuroendocrine Brain", F-59045, Lille, France
| | - Karima Mokhtari
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France.,Laboratoire R Escourolle, AP-HP, Hôpital de la Pitié-Salpêtrière, F-75013, Paris, France
| | - Stefano Maria Pagnotta
- Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio, 82100, Benevento, Italy.,Institute for Cancer Genetics, Columbia University Medical Center, New York City, NY, 10032, USA
| | - Antonio Iavarone
- Institute for Cancer Genetics, Columbia University Medical Center, New York City, NY, 10032, USA.,Departments of Neurology and Pathology, Institute for Cancer Genetics, Irving Comprehensive Research Center, New York, NY, 10032, USA
| | - Anna Lasorella
- Institute for Cancer Genetics, Columbia University Medical Center, New York City, NY, 10032, USA.,Departments of Pediatrics and Pathology, Institute for Cancer Genetics, Irving Comprehensive Research Center, New York, NY, 10032, USA
| | - Emmanuelle Huillard
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France
| | - Marc Sanson
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, F-75013, Paris, France. .,Onconeurotek Tumor Bank, Institut du Cerveau et de la Moelle épinère-ICM, F-75013, Paris, France. .,AP-HP, Hôpital de la Pitié-Salpêtrière, Service de Neurologie 2, F-75013, Paris, France. .,Site de Recherche Intégrée sur le Cancer (SiRIC) "CURAMUS", F-75013, Paris, France.
| |
Collapse
|
30
|
Arai M, Sera T, Hasegawa T, Kudo S. Spatial and temporal translocation of PKCα in single endothelial cell in response to mechanical stimulus. Exp Cell Res 2018; 367:205-215. [DOI: 10.1016/j.yexcr.2018.03.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 03/28/2018] [Accepted: 03/29/2018] [Indexed: 12/14/2022]
|
31
|
Guo Y, Bao Y, Ma M, Zhang S, Zhang Y, Yuan M, Liu B, Yang Y, Cui W, Ansong E, Dong H, Macias V, Yang W. Clinical significance of the correlation between PLCE 1 and PRKCA in esophageal inflammation and esophageal carcinoma. Oncotarget 2018; 8:33285-33299. [PMID: 28402280 PMCID: PMC5464868 DOI: 10.18632/oncotarget.16635] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 03/11/2017] [Indexed: 12/16/2022] Open
Abstract
Esophagitis and Barrett's esophagus are linked to esophageal squamous cell carcinoma and adenocarcinoma, respectively. However, the underlying mechanisms are still unclear. This study analyzed the expression levels of and correlation between PLCE1 and PRKCA in human esophagitis, carcinogen NMBA-induced rat esophagus, PLCE1 genetic deficient mouse esophageal epithelial tissues and human esophageal cancer cell line, integrated with Online oncology data sets. We found that the expression levels of both PLCE1 and PRKCA were significantly elevated in human esophagitis, esophageal squamous cell carcinoma, Barrett's esophagus, esophageal adenocarcinoma and in NMBA-treated rat esophageal epithelia. However, PRKCA and cytokines were significantly downregulated in PLCE1-deficient mouse esophageal epithelia, and knockdown of PLCE1 in human esophageal cancer cells led to reduction of PRKCA and cytokines. Finally, high expression of both PLCE1 and PRKCA is significantly associated with poor outcomes of the patients with esophageal cancers. In conclusion, this study defined the initiation and progression of esophageal inflammation and malignant transformation, in which the positive correlation of PLCE1 and PRKCA exhibits critical clinical significance.
Collapse
Affiliation(s)
- Yongchen Guo
- Department of Pathology and Institute of Precision Medicine, Jining Medical University, Jining 272067, China
| | - Yonghua Bao
- Department of Pathology and Institute of Precision Medicine, Jining Medical University, Jining 272067, China
| | - Ming Ma
- Department of Thoracic Surgery, Affiliated Hospital, Jining Medical University, Jining 272067, China
| | - Shanshan Zhang
- Department of Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Yongmeng Zhang
- Department of Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Ming Yuan
- Department of Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Bing Liu
- Department of Pathology and Institute of Precision Medicine, Jining Medical University, Jining 272067, China
| | - Yiqiong Yang
- Department of Pathology and Institute of Precision Medicine, Jining Medical University, Jining 272067, China
| | - Wen Cui
- Department of Pathology and Institute of Precision Medicine, Jining Medical University, Jining 272067, China
| | - Emmanuel Ansong
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Huali Dong
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Virgilia Macias
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Wancai Yang
- Department of Pathology and Institute of Precision Medicine, Jining Medical University, Jining 272067, China.,Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
32
|
Guo A, Chen R, Wang Y, Huang CK, Chen B, Kutschke W, Hong J, Song LS. Transient activation of PKC results in long-lasting detrimental effects on systolic [Ca 2+] i in cardiomyocytes by altering actin cytoskeletal dynamics and T-tubule integrity. J Mol Cell Cardiol 2018; 115:104-114. [PMID: 29307535 DOI: 10.1016/j.yjmcc.2018.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/18/2017] [Accepted: 01/03/2018] [Indexed: 11/25/2022]
Abstract
AIMS Protein kinase C (PKC) isozymes contribute to the development of heart failure through dysregulation of Ca2+ handling properties and disruption of contractile function in cardiomyocytes. However, the mechanisms by which PKC activation leads to Ca2+ dysfunction are incompletely understood. METHODS AND RESULTS Shortly upon ventricular pressure overload in mice, we detected transient PKC activation that was associated with pulsed actin cytoskeletal rearrangement. In cultured cardiomyocytes, transient activation of PKC promoted long-term deleterious effects on the integrity of the transverse (T)- tubule system, resulting in a significant decrease in the amplitude and increase in the rising kinetics of Ca2+ transients. Treatment with a PKCα/β inhibitor restored the synchronization of Ca2+ transients and maintained T-tubule integrity in cultured cardiomyocytes. Supporting these data, PKCα/β inhibition protected against T-tubule remodeling and cardiac dysfunction in a mouse model of pressure overload-induced heart failure. Mechanistically, transient activation of PKC resulted in biphasic actin cytoskeletal rearrangement, consistent with in vivo observations in the pressure overloaded mouse model. Transient inhibition of actin polymerization or depolymerization resulted in severe T-tubule damage, recapitulating the T-tubule damage induced by PKC activation. Moreover, inhibition of stretch activated channels (SAC) protected against T-tubule remodeling and E-C coupling dysfunction induced by transient PKC activation and actin cytoskeletal rearrangement. CONCLUSIONS These data identify a key mechanistic link between transient PKC activation and long-term Ca2+ handling defects through PKC-induced actin cytoskeletal rearrangement and resultant T-tubule damage.
Collapse
Affiliation(s)
- Ang Guo
- Division of Cardiovascular Medicine, Department of Internal Medicine & Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Rong Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine & Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Yihui Wang
- Division of Cardiovascular Medicine, Department of Internal Medicine & Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Chun-Kai Huang
- Division of Cardiovascular Medicine, Department of Internal Medicine & Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Biyi Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine & Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - William Kutschke
- Division of Cardiovascular Medicine, Department of Internal Medicine & Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Jiang Hong
- Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine & Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Veterans Affairs Medical Center, Iowa City, IA 52242, USA.
| |
Collapse
|
33
|
Wang Y, Yin X, Zhao L, Li S, Duan J, Kuang R, Duan J. MicroRNA-200b inhibits pituitary tumor cell proliferation and invasion by targeting PKCα. Exp Ther Med 2017; 14:1706-1714. [PMID: 28810639 DOI: 10.3892/etm.2017.4681] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 01/13/2017] [Indexed: 12/27/2022] Open
Abstract
The present study aimed to investigate the expression of miR-200b and protein kinase Cα (PKCα) in pituitary tumors and to determine whether miR-200b may inhibit proliferation and invasion of pituitary tumor cells. The regulation of PKCα expression was targeted in order to find novel targets for the treatment of pituitary tumors. In total, 53 pituitary tumor tissue samples were collected; these included 28 cases of invasive pituitary tumors and 25 cases of non-invasive tumors, in addition to 5 normal pituitaries. The expression level of miR-200b in the pituitary tumor tissue was detected by quantitative polymerase chain reaction (qPCR) and the expression of PKCα protein was detected by immunohistochemistry. A PKCα 3'untranslated region (UTR) luciferase vector was constructed and a dual luciferase reporter gene assay was employed in order to examine the effect of miR-200b on the PKCα 3'UTR luciferase activity. AtT-20 cells were transfected with miR-200b mimics, PKCα siRNA and miR-200b mimics + PKCα, and the changes in cellular proliferation, invasion and apoptosis were observed via MTT, Transwell assay and flow cytometric analysis. Furthermore, PKCα mRNA expression was determined by qPCR, and Western blotting was performed to detect the expression of PKCα protein. miR-200b revealed downregulation in invasive pituitary tumor tissue, and the expression level was significantly down-regulated compared with normal and non-invasive pituitary tumor tissue (P<0.01). In addition, the positive rate of PKCα protein expression in invasive pituitary tumor tissues was significantly higher than in normal and non-invasive tissues (P<0.01). PKCα protein levels are inversely correlated with miR-200b levels in invasive pituitary tumor tissues (r=-0.436, P=0.021). The dual luciferase reporter gene assay revealed that miR-200b could specifically bind to the 3'UTR of PKCα and significantly inhibit the luciferase activity by 39% (P<0.01). Upregulation of miR-200b or downregulation of PKCα could suppress cell proliferation and invasion, and increase apoptosis of AtT-20 cells. It was revealed that PKCα siRNA could suppress both proliferation and invasion of AtT-20 cells and partially simulate the function of miR-200b. Expression of PKCα mRNA and protein decreased significantly in AtT-20 cells overexpressing miR-200b. Additionally, miR-200b was significantly down-regulated in invasive pituitary tumor tissue and inversely correlated with PKCα protein levels. In conclusion, miR-200b inhibited proliferation and invasiveness and promoted the apoptosis of pituitary tumor cells by targeting PKCα. The observations of the present study indicate that miR-200b and PKCα may serve as promising therapeutic targets for invasive pituitary tumors.
Collapse
Affiliation(s)
- Yuanchuan Wang
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Xiaohong Yin
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Long Zhao
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Shun Li
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Jie Duan
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Renzhao Kuang
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Junwei Duan
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
34
|
Interacting post-muscarinic receptor signaling pathways potentiate matrix metalloproteinase-1 expression and invasion of human colon cancer cells. Biochem J 2017; 474:647-665. [PMID: 28008134 DOI: 10.1042/bcj20160704] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 12/16/2016] [Accepted: 12/21/2016] [Indexed: 12/26/2022]
Abstract
M3 muscarinic receptor (M3R) expression is increased in colon cancer; M3R activation stimulates colon cancer cell invasion via cross-talk with epidermal growth factor receptors (EGFR), post-EGFR activation of mitogen-activated protein kinase (MAPK) extracellular signal-related kinase 1/2 (ERK1/2), and induction of matrix metalloproteinase-1 (MMP1) expression. MMP1 expression is strongly associated with tumor metastasis and adverse outcomes. Here, we asked whether other MAPKs regulate M3R agonist-induced MMP1 expression. In addition to activating ERK1/2, we found that treating colon cancer cells with acetylcholine (ACh) stimulated robust time- and dose-dependent phosphorylation of p38 MAPK. Unlike ERK1/2 activation, ACh-induced p38 phosphorylation was EGFR-independent and blocked by inhibiting protein kinase C-α (PKC-α). Inhibiting activation of PKC-α, EGFR, ERK1/2, or p38-α/β alone attenuated, but did not abolish ACh-induced MMP1 expression, a finding that predicted potentiating interactions between these pathways. Indeed, ACh-induced MMP1 expression was abolished by incubating cells with either an EGFR or MEK/ERK1/2 inhibitor combined with a p38-α/β inhibitor. Activating PKC-α and EGFR directly with the combination of phorbol 12-myristate 13-acetate (PMA) and EGF potentiated MMP1 gene and protein expression, and cell invasion. PMA- and ACh-induced MMP1 expression were strongly diminished by inhibiting Src and abolished by concurrently inhibiting both p38-α/β and Src, indicating that Src mediates the cross-talk between PKC-α and EGFR signaling. Using siRNA knockdown, we identified p38-α as the relevant p38 isoform. Collectively, these studies uncover novel functional interactions between post-muscarinic receptor signaling pathways that augment MMP1 expression and drive colon cancer cell invasion; targeting these potentiating interactions has therapeutic potential.
Collapse
|
35
|
Pany S, You Y, Das J. Curcumin Inhibits Protein Kinase Cα Activity by Binding to Its C1 Domain. Biochemistry 2016; 55:6327-6336. [PMID: 27776404 DOI: 10.1021/acs.biochem.6b00932] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Curcumin is a polyphenolic nutraceutical that acts on multiple biological targets, including protein kinase C (PKC). PKC is a family of serine/threonine kinases central to intracellular signal transduction. We have recently shown that curcumin selectively inhibits PKCα, but not PKCε, in CHO-K1 cells [Pany, S. (2016) Biochemistry 55, 2135-2143]. To understand which domain(s) of PKCα is responsible for curcumin binding and inhibitory activity, we made several domain-swapped mutants in which the C1 (combination of C1A and C1B) and C2 domains are swapped between PKCα and PKCε. Phorbol ester-induced membrane translocation studies using confocal microscopy and immunoblotting revealed that curcumin inhibited phorbol ester-induced membrane translocation of PKCε mutants, in which the εC1 domain was replaced with αC1, but not the PKCα mutant in which αC1 was replaced with the εC1 domain, suggesting that αC1 is a determinant for curcumin's inhibitory effect. In addition, curcumin inhibited membrane translocation of PKCε mutants, in which the εC1A and εC1B domains were replaced with the αC1A and αC1B domains, respectively, indicating the role of both αC1A and αC1B domains in curcumin's inhibitory effects. Phorbol 13-acetate inhibited the binding of curcumin to αC1A and αC1B with IC50 values of 6.27 and 4.47 μM, respectively. Molecular docking and molecular dynamics studies also supported the higher affinity of curcumin for αC1B than for αC1A. The C2 domain-swapped mutants were inactive in phorbol ester-induced membrane translocation. These results indicate that curcumin binds to the C1 domain of PKCα and highlight the importance of this domain in achieving PKC isoform selectivity.
Collapse
Affiliation(s)
- Satyabrata Pany
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston , Houston, Texas 77204, United States
| | - Youngki You
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston , Houston, Texas 77204, United States
| | - Joydip Das
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston , Houston, Texas 77204, United States
| |
Collapse
|
36
|
Maurya AK, Vinayak M. PI-103 attenuates PI3K-AKT signaling and induces apoptosis in murineT-cell lymphoma. Leuk Lymphoma 2016; 58:1153-1161. [PMID: 27658642 DOI: 10.1080/10428194.2016.1225207] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Aberrant activation of PI3K-AKT signaling in many pathological conditions including cancer has attracted much of interest for drug targeting. Various isoforms are known from three classes of PI3K. Targeting selective isoform is advantageous to overcome the global deleterious effects of drug. PI-103 is a specific inhibitor of p110α of class I PI3K. The present study is aimed to analyze anti-carcinogenic activity of PI-103 in Dalton's lymphoma ascite (DLA) cells. Result shows regression in cell proliferation and increased apoptosis in terms of increased Annexin V binding, nuclear fragmentation and active caspase 3 level. It is correlated with attenuation of PI3K-AKT signaling by PI-103 via downregulation of the level of p110α, phospho-p85α, phospho- AKT, and PKCα in DLA cells as well as in H2O2 induced DLA cells. Additionally, ROS accumulation is declined in H2O2 induced DLA cells. Overall result suggests that PI-103 attenuates PI3K-AKT signaling via induction of apoptosis in murine T-cell lymphoma.
Collapse
Affiliation(s)
- Akhilendra Kumar Maurya
- a Biochemistry & Molecular Biology Laboratory, Department of Zoology, Institute of Science , Banaras Hindu University , Varanasi , India
| | - Manjula Vinayak
- a Biochemistry & Molecular Biology Laboratory, Department of Zoology, Institute of Science , Banaras Hindu University , Varanasi , India
| |
Collapse
|
37
|
Shimshoni JA, Winkler I, Golan E, Nutt D. Neurochemical binding profiles of novel indole and benzofuran MDMA analogues. Naunyn Schmiedebergs Arch Pharmacol 2016; 390:15-24. [PMID: 27650729 DOI: 10.1007/s00210-016-1297-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 09/02/2016] [Indexed: 12/11/2022]
Abstract
3,4-Methylenedioxy-N-methylamphetamine (MDMA) has been shown to be effective in the treatment of post-traumatic stress disorder (PTSD) in numerous clinical trials. In the present study, we have characterized the neurochemical binding profiles of three MDMA-benzofuran analogues (1-(benzofuran-5-yl)-propan-2-amine, 5-APB; 1-(benzofuran-6-yl)-N-methylpropan-2-amine, 6-MAPB; 1-(benzofuran-5-yl)-N-methylpropan-2-amine, 5-MAPB) and one MDMA-indole analogue (1-(1H-indol-5-yl)-2-methylamino-propan-1-ol, 5-IT). These compounds were screened as potential second-generation anti-PTSD drugs, against a battery of human and non-human receptors, transporters, and enzymes, and their potencies as 5-HT2 receptor agonist and monoamine uptake inhibitors determined. All MDMA analogues displayed high binding affinities for 5-HT2a,b,c and NEα2 receptors, as well as significant 5-HT, DA, and NE uptake inhibition. 5-APB revealed significant agonist activity at the 5-HT2a,b,c receptors, while 6-MAPB, 5-MAPB, and 5-IT exhibited significant agonist activity at the 5-HT2c receptor. There was a lack of correlation between the results of functional uptake and the monoamine transporter binding assay. MDMA analogues emerged as potent and selective monoamine oxidase A inhibitors. Based on 6-MAPB favorable pharmacological profile, it was further subjected to IC50 determination for monoamine transporters. Overall, all MDMA analogues displayed higher monoamine receptor/transporter binding affinities and agonist activity at the 5-HT2a,c receptors as compared to MDMA.
Collapse
Affiliation(s)
- Jakob A Shimshoni
- Department of Toxicology, Kimron Veterinary Institute, Bet Dagan, Israel.
| | | | | | - David Nutt
- Neuropsychopharmacology Unit, Imperial College London, London, UK
| |
Collapse
|
38
|
Wang Y, Qi W, Song Y. Antibody-free detection of protein phosphorylation using intrinsic peroxidase-like activity of platinum/carbon dot hybrid nanoparticles. Chem Commun (Camb) 2016; 52:7994-7. [PMID: 27264272 DOI: 10.1039/c6cc02771g] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Platinum and carbon dot hybrid nanomaterials are prepared for visualized detection of phosphoproteins without the need for antibodies or enzymes. This new strategy can be used for colorimetric detection of phosphoproteins induced by protein kinase as well as protein phosphorylation sites on cell membranes.
Collapse
Affiliation(s)
- Yuzhen Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210093, China.
| | | | | |
Collapse
|
39
|
Protein Kinase C-α is a Critical Protein for Antisense Oligonucleotide-mediated Silencing in Mammalian Cells. Mol Ther 2016; 24:1117-1125. [PMID: 26961407 DOI: 10.1038/mt.2016.54] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 02/18/2016] [Indexed: 01/08/2023] Open
Abstract
We have identified the existence of a productive, PKC-α-dependent endocytotic silencing pathway that leads gymnotically-delivered locked nucleic acid (LNA)-gapmer phosphorothioate antisense oligonucleotides (ASOs) into late endosomes. By blocking the maturation of early endosomes to late endosomes, silencing the expression of PKC-α results in the potent reduction of ASO silencing ability in the cell. We have also demonstrated that silencing of gene expression in the cytoplasm is vitiated when PKC-α expression is reduced. Restoring PKC-α expression via a reconstitution experiment reinstates the ability of ASOs to silence. These results advance our understanding of intracellular ASO trafficking and activity following gymnotic delivery, and further demonstrate the existence of two distinct silencing pathways in mammalian cells, one in the cytoplasmic and the other in the nuclear compartment.
Collapse
|
40
|
Suppression of A549 cell proliferation and metastasis by calycosin via inhibition of the PKC‑α/ERK1/2 pathway: An in vitro investigation. Mol Med Rep 2015; 12:7992-8002. [PMID: 26498639 PMCID: PMC4758323 DOI: 10.3892/mmr.2015.4449] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 08/24/2015] [Indexed: 12/28/2022] Open
Abstract
The migration and invasion of lung cancer cells into the extracellular matrix contributes to the high mortality rates of lung cancer. The protein kinase C (PKC) and downstream signaling pathways are important in the invasion and migration of lung cancer cells. Calycosin (Cal), an effector chemical from Astragalus has been reported to affect the recurrence and metastasis of cancer cells via the regulation of the protein expression of matrix metalloproteinases (MMPs). The inhibition of Cal on the migration and invasion of A549 cells was investigated in the present study. Cell viability and apoptosis assays were performed using MTT and flow cytometric analyses. A wound healing assay and Transwell invasion assay were performed to evaluate the effect of Cal on A549 cell migration and invasion. Invasion-associated proteins, including MMP-2, MMP-9, E-cadherin (E-cad), integrin β1, PKC-α and extracellular signal-regulated kinase 1/2 (ERK1/2) were detected using western blotting. In addition, PKC-α inhibitor, AEB071, and ERK1/2 inhibitor, PD98059, were used to determine the association between the suppression of PKC-α/ERK1/2 and invasion, MMP-2, MMP-9, E-cad and integrin β1. Cal was observed to suppress cell proliferation and induce apoptosis. There were significant differences between the phorbol-12-myristate-13-acetate (TPA)-induced A549 cells treated with Cal and the untreated cells in the rates of migration and invasion. The levels of MMP-2, MMP-9, E-cad and integrin β1 in the TPA-induced A549 cells changed markedly, compared with the untreated cells. In addition, the suppression of Cal was affected by the PKC inhibitor, AEB071, an ERK1/2 inhibitor, PD98059. The results of the present study indicated that Cal inhibited the proliferation, adhesion, migration and invasion of the TPA-induced A549 cells. The Cal-induced repression of PKC-α/ERK1/2, increased the expression of E-Cad and inhibited the expression levels of MMP-2, MMP-9 and integrin β1, which possibly demonstrates the mechanism underlying the biological anticancer effects of Cal.
Collapse
|
41
|
Rostam MA, Piva TJ, Rezaei HB, Kamato D, Little PJ, Zheng W, Osman N. Peptidyl-prolyl isomerases: functionality and potential therapeutic targets in cardiovascular disease. Clin Exp Pharmacol Physiol 2015; 42:117-24. [PMID: 25377120 DOI: 10.1111/1440-1681.12335] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/26/2014] [Accepted: 10/30/2014] [Indexed: 02/06/2023]
Abstract
Peptidyl-prolyl cis/trans isomerases (PPIases) are a conserved group of enzymes that catalyse the conversion between cis and trans conformations of proline imidic peptide bonds. These enzymes play critical roles in regulatory mechanisms of cellular function and pathophysiology of disease. There are three different classes of PPIases and increasing interest in the development of specific PPIase inhibitors. Cyclosporine A, FK506, rapamycin and juglone are known PPIase inhibitors. Herein, we review recent advances in elucidating the role and regulation of the PPIase family in vascular disease. We focus on peptidyl-prolyl cis/trans isomerase NIMA-interacting 1 (Pin1), an important member of the PPIase family that plays a role in cell cycle progression, gene expression, cell signalling and cell proliferation. In addition, Pin1 may be involved in atherosclerosis. The unique role of Pin1 as a molecular switch that impacts on multiple downstream pathways necessitates the evaluation of a highly specific Pin1 inhibitor to aid in potential therapeutic drug discovery.
Collapse
Affiliation(s)
- Muhamad A Rostam
- Discipline of Pharmacy, RMIT University, Melbourne, Vic., Australia; Diabetes Complications Group, Metabolism, Exercise and Disease Program, Health Innovations Research Institute, RMIT University, Melbourne, Vic., Australia; International Islamic University Malaysia, Kuala Lumpur, Malaysia
| | | | | | | | | | | | | |
Collapse
|
42
|
Yang J, Han H, Zhao Y, Qin H. Specific miRNA and its target in neutrophils after traumatic injury. Acta Biochim Biophys Sin (Shanghai) 2015; 47:749-54. [PMID: 26232986 DOI: 10.1093/abbs/gmv072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/24/2015] [Indexed: 01/09/2023] Open
Abstract
Traumatic injury is a leading cause of mortality and morbidity. MicroRNAs (miRNAs) regulate the cellular responses when traumatic injury occurs. Previously, we reported that miR-3945, miR-125a-5p, miR-363-3p, and miR-150-5p were significantly altered in neutrophils of patients who suffered traumatic injury. In the present study, by comparing neutrophils of patients suffering from major trauma with neutrophils of patients with a inflammatory disease, we found that the variation trend of miR-150-5p was relatively different in the process of these two diseases. Gene Ontology and pathway analysis of miR-150-5p revealed that it may activate the mitogen-activated protein kinase and Toll-like receptor signaling pathways and cell adhesion molecules when the traumatic injury occurs. In addition, protein kinase C alpha (PRKCA) was also identified as a direct target of miR-150-5p by establishing a miRNA-mRNA network, and this target was validated via dual-luciferase reporter and western blot analysis. Our results suggested that the expression of miR-150-5p was down-regulated in neutrophils after a major traumatic injury. miR-150-5p and its identified target PRKCA play important roles in the development of traumatic process.
Collapse
Affiliation(s)
- Jun Yang
- Department of Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Huazhong Han
- Department of Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yijun Zhao
- Department of Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Huanlong Qin
- Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| |
Collapse
|
43
|
Anticarcinogenic action of quercetin by downregulation of phosphatidylinositol 3-kinase (PI3K) and protein kinase C (PKC) via induction of p53 in hepatocellular carcinoma (HepG2) cell line. Mol Biol Rep 2015; 42:1419-29. [DOI: 10.1007/s11033-015-3921-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/20/2015] [Indexed: 02/06/2023]
|
44
|
Chu LH, Annex BH, Popel AS. Computational drug repositioning for peripheral arterial disease: prediction of anti-inflammatory and pro-angiogenic therapeutics. Front Pharmacol 2015; 6:179. [PMID: 26379552 PMCID: PMC4548203 DOI: 10.3389/fphar.2015.00179] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/10/2015] [Indexed: 12/17/2022] Open
Abstract
Peripheral arterial disease (PAD) results from atherosclerosis that leads to blocked arteries and reduced blood flow, most commonly in the arteries of the legs. PAD clinical trials to induce angiogenesis to improve blood flow conducted in the last decade have not succeeded. We have recently constructed PADPIN, protein-protein interaction network (PIN) of PAD, and here we combine it with the drug-target relations to identify potential drug targets for PAD. Specifically, the proteins in the PADPIN were classified as belonging to the angiome, immunome, and arteriome, characterizing the processes of angiogenesis, immune response/inflammation, and arteriogenesis, respectively. Using the network-based approach we predict the candidate drugs for repositioning that have potential applications to PAD. By compiling the drug information in two drug databases DrugBank and PharmGKB, we predict FDA-approved drugs whose targets are the proteins annotated as anti-angiogenic and pro-inflammatory, respectively. Examples of pro-angiogenic drugs are carvedilol and urokinase. Examples of anti-inflammatory drugs are ACE inhibitors and maraviroc. This is the first computational drug repositioning study for PAD.
Collapse
Affiliation(s)
- Liang-Hui Chu
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University Baltimore, MD, USA
| | - Brian H Annex
- Division of Cardiovascular Medicine, Department of Medicine and Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine Charlottesville, VA, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University Baltimore, MD, USA
| |
Collapse
|
45
|
Maurya AK, Vinayak M. Modulation of PKC signaling and induction of apoptosis through suppression of reactive oxygen species and tumor necrosis factor receptor 1 (TNFR1): key role of quercetin in cancer prevention. Tumour Biol 2015; 36:8913-24. [DOI: 10.1007/s13277-015-3634-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/01/2015] [Indexed: 02/06/2023] Open
|
46
|
Bhavanasi D, Badolia R, Manne BK, Janapati S, Dangelmaier CT, Mazharian A, Jin J, Kim S, Zhang X, Chen X, Senis YA, Kunapuli SP. Cross talk between serine/threonine and tyrosine kinases regulates ADP-induced thromboxane generation in platelets. Thromb Haemost 2015; 114:558-68. [PMID: 25947062 DOI: 10.1160/th14-09-0775] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 03/26/2015] [Indexed: 11/05/2022]
Abstract
ADP-induced thromboxane generation depends on Src family kinases (SFKs) and is enhanced with pan-protein kinase C (PKC) inhibitors, but it is not clear how these two events are linked. The aim of the current study is to investigate the role of Y311 phosphorylated PKCδ in regulating ADP-induced platelet activation. In the current study, we employed various inhibitors and murine platelets from mice deficient in specific molecules to evaluate the role of PKCδ in ADP-induced platelet responses. We show that, upon stimulation of platelets with 2MeSADP, Y311 on PKCδ is phosphorylated in a P2Y1/Gq and Lyn-dependent manner. By using PKCδ and Lyn knockout murine platelets, we also show that tyrosine phosphorylated PKCδ plays a functional role in mediating 2MeSADP-induced thromboxane generation. 2MeSADP-induced PKCδ Y311 phosphorylation and thromboxane generation were potentiated in human platelets pre-treated with either a pan-PKC inhibitor, GF109203X or a PKC α/β inhibitor and in PKC α or β knockout murine platelets compared to controls. Furthermore, we show that PKC α/β inhibition potentiates the activity of SFK, which further hyper-phosphorylates PKCδ and potentiates thromboxane generation. These results show for the first time that tyrosine phosphorylated PKCδ regulates ADP-induced thromboxane generation independent of its catalytic activity and that classical PKC isoforms α/β regulate the tyrosine phosphorylation on PKCδ and subsequent thromboxane generation through tyrosine kinase, Lyn, in platelets.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Satya P Kunapuli
- Satya P. Kunapuli PhD, Department of Physiology and Sol Sherry Thrombosis Center,, Temple University School of Medicine,, 3420 North Broad street, MRB 414, Philadelphia PA, 19140, USA, Tel.: +1 215 707 4615, Fax: +1 215 707 6944, E-mail:
| |
Collapse
|
47
|
Vaškovičová K, Szabadosová E, Čermák V, Gandalovičová A, Kasalová L, Rösel D, Brábek J. PKCα promotes the mesenchymal to amoeboid transition and increases cancer cell invasiveness. BMC Cancer 2015; 15:326. [PMID: 25924946 PMCID: PMC4423130 DOI: 10.1186/s12885-015-1347-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 04/22/2015] [Indexed: 12/13/2022] Open
Abstract
Background The local invasion of tumor cells into the surrounding tissue is the first and most critical step of the metastatic cascade. Cells can invade either collectively, or individually. Individual cancer cell invasion can occur in the mesenchymal or amoeboid mode, which are mutually interchangeable. This plasticity of individual cancer cell invasiveness may represent an escape mechanism for invading cancer cells from anti-metastatic treatment. Methods To identify new signaling proteins involved in the plasticity of cancer cell invasiveness, we performed proteomic analysis of the amoeboid to mesenchymal transition with A375m2 melanoma cells in a 3D Matrigel matrix. Results In this screen we identified PKCα as an important protein for the maintenance of amoeboid morphology. We found that the activation of PKCα resulted in the mesenchymal-amoeboid transition of mesenchymal K2 and MDA-MB-231 cell lines. Consistently, PKCα inhibition led to the amoeboid-mesenchymal transition of amoeboid A375m2 cells. Next, we showed that PKCα inhibition resulted in a considerable decrease in the invading abilities of all analyzed cancer cell lines. Conclusions Our results suggest that PKCα is an important protein for maintenance of the amoeboid morphology of cancer cells, and that downregulation of PKCα results in the amoeboid to mesenchymal transition. Our data also suggest that PKCα is important for both mesenchymal and amoeboid invasiveness, making it an attractive target for anti-metastatic therapies. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1347-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katarína Vaškovičová
- Department of Cell Biology, Laboratory of Cancer Cell Invasion, Charles University in Prague, Prague, Czech Republic. .,Current affiliation: Microscopy Unit, Institute of Experimental Medicine, The Czech Academy of Sciences, Prague, Czech Republic.
| | - Emilia Szabadosová
- Department of Cell Biology, Laboratory of Cancer Cell Invasion, Charles University in Prague, Prague, Czech Republic.
| | - Vladimír Čermák
- Department of Cell Biology, Laboratory of Cancer Cell Invasion, Charles University in Prague, Prague, Czech Republic.
| | - Aneta Gandalovičová
- Department of Cell Biology, Laboratory of Cancer Cell Invasion, Charles University in Prague, Prague, Czech Republic.
| | - Lenka Kasalová
- Department of Cell Biology, Laboratory of Cancer Cell Invasion, Charles University in Prague, Prague, Czech Republic.
| | - Daniel Rösel
- Department of Cell Biology, Laboratory of Cancer Cell Invasion, Charles University in Prague, Prague, Czech Republic.
| | - Jan Brábek
- Department of Cell Biology, Laboratory of Cancer Cell Invasion, Charles University in Prague, Prague, Czech Republic.
| |
Collapse
|
48
|
Protein phosphorylation detection using dual-mode field-effect devices and nanoplasmonic sensors. Sci Rep 2015; 5:8687. [PMID: 25732235 PMCID: PMC4346972 DOI: 10.1038/srep08687] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 01/27/2015] [Indexed: 01/25/2023] Open
Abstract
Phosphorylation by kinases is an important post-translational modification of proteins. It is a critical control for the regulation of vital cellular activities, and its dysregulation is implicated in several diseases. A common drug discovery approach involves, therefore, time-consuming screenings of large libraries of candidate compounds to identify novel inhibitors of protein kinases. In this work, we propose a novel method that combines localized surface plasmon resonance (LSPR) and electrolyte insulator semiconductor (EIS)-based proton detection for the rapid identification of novel protein kinase inhibitors. In particular, the selective detection of thiophosphorylated proteins by LSPR is achieved by changing their resonance properties via a pre-binding with gold nanoparticles. In parallel, the EIS field-effect structure allows the real-time electrochemical monitoring of the protein phosphorylation by detecting the release of protons associated with the kinases activity. This innovative combination of both field-effect and nanoplasmonic sensing makes the detection of protein phosphorylation more reliable and effective. As a result, the screening of protein kinase inhibitors becomes more rapid, sensitive, robust and cost-effective.
Collapse
|
49
|
González-Arenas A, Peña-Ortiz MÁ, Hansberg-Pastor V, Marquina-Sánchez B, Baranda-Ávila N, Nava-Castro K, Cabrera-Wrooman A, González-Jorge J, Camacho-Arroyo I. PKCα and PKCδ activation regulates transcriptional activity and degradation of progesterone receptor in human astrocytoma cells. Endocrinology 2015; 156:1010-22. [PMID: 25514083 DOI: 10.1210/en.2014-1137] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Progesterone regulates cancer cell proliferation and invasion through its receptors (PR-A and PR-B), whose phosphorylation modifies their transcriptional activity and induce their degradation. We identified by in silico analysis a putative residue (Ser400) in PR that might be phosphorylated by protein kinase C (PKC), a family of enzymes involved in the proliferation and infiltration of astrocytomas, the most frequent and aggressive brain tumors. A grade III human astrocytoma-derived cell line was used to study the role of PKC in PR phosphorylation, transcriptional activity, and degradation. Treatment with PKC activator [tetradecanoyl phorbol acetate (TPA)] increased PR phosphorylation in Ser400 after 5 minutes, which in turn induced PR transcriptional activity and its subsequent degradation by the 26S proteasome 3-5 hours after treatment. Silencing or inhibition of PKCα and PKCδ blocked PR phosphorylation and degradation induced by TPA. Both PR isoforms were associated with PKCα and reached the maximum association after 5 minutes of TPA addition. These data correlated with immunnofluorescence assays in which nuclear colocalization of PKCα with PR increased after TPA treatment. We observed a 2-fold increase in cell proliferation after PKC activation with TPA that was reduced with the PR antagonist, RU486. The PR S400A mutant revealed that this residue is essential for PKC-mediated PR phosphorylation and degradation. Our results show a key participation of PKCα and PKCδ in PR regulation and function.
Collapse
Affiliation(s)
- Aliesha González-Arenas
- Departamento de Biología (A.G.-A., M.A.P.-O., V.H.-P., B.M.-S., K.N.-C., A.C.-W., J.G.-J., I.C.-A.), Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán 04510, Distrito Federal, México; and División de Investigación Básica (N.B.-A.), Instituto Nacional de Cancerología, México City 14080, Distrito Federal, México
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Fukahori H, Chida N, Maeda M, Tasaki M, Kawashima T, Matsuoka H, Suzuki K, Ishikawa T, Tanaka A, Higashi Y. Effect of AS2521780, a novel PKCθ selective inhibitor, on T cell-mediated immunity. Eur J Pharmacol 2014; 745:217-22. [DOI: 10.1016/j.ejphar.2014.10.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 10/23/2014] [Accepted: 10/28/2014] [Indexed: 01/29/2023]
|