1
|
Guo X, Lu Y, Zhang X, Wang X, Wang H, Zhang Z. A 4-phenylbutyric acid-conjugated platinum diimine complex: Photocytotoxicity, cell cycle arrest and apoptosis in vitro. J Inorg Biochem 2025; 270:112930. [PMID: 40262391 DOI: 10.1016/j.jinorgbio.2025.112930] [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: 08/08/2024] [Revised: 04/09/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025]
Abstract
4-phenylbutyric acid (PBA) is a class I and II histone deacetylase inhibitor, which can decrease cell proliferation, enhance cell differentiation, and induce apoptosis and cell cycle arrest in various cancer cells. PBA-modified photoactive platinum diimine complex has been prepared and characterized to augment its photodynamic therapy efficacy. Its ability to generate singlet oxygen, behavior in the presence of esterase, photocytotoxicity, cell cycle distribution, and apoptosis-inducing ability in MCF-7 cells have also been studied. The results suggested that the PBA-modified platinum diimine complex could act as a good singlet oxygen producer, release PBA in the presence of esterase, induce potent photocytotoxicity in tumor cells with a IC50 value of 3.67 ± 0.67 μM, arrest cell cycle at S phase, and induce cell death via apoptosis with the percentage of total apoptotic cells being 34.65 %. The results also revealed that PBA might be able to modulate the mode of cell cycle arrest induced by the photosensitizer in tumor cells.
Collapse
Affiliation(s)
- Xuemei Guo
- Institute of Molecular Science, Chemical Biology and Molecular Engineering Laboratory of Education Ministry, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Yuxin Lu
- Institute of Molecular Science, Chemical Biology and Molecular Engineering Laboratory of Education Ministry, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Xu Zhang
- Institute of Molecular Science, Chemical Biology and Molecular Engineering Laboratory of Education Ministry, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Xin Wang
- Institute of Molecular Science, Chemical Biology and Molecular Engineering Laboratory of Education Ministry, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Hongfei Wang
- Institute of Molecular Science, Chemical Biology and Molecular Engineering Laboratory of Education Ministry, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Zhigang Zhang
- Institute of Molecular Science, Chemical Biology and Molecular Engineering Laboratory of Education Ministry, Shanxi University, Taiyuan, Shanxi 030006, China.
| |
Collapse
|
2
|
Kalsum S, Xu R, Akber M, Huang S, Lerm M, Chen Y, Lourda M, Zhou Y, Brighenti S. Dual GSK-3β/HDAC Inhibitors Enhance the Efficacy of Macrophages to Control Mycobacterium tuberculosis Infection. Biomolecules 2025; 15:550. [PMID: 40305296 PMCID: PMC12024928 DOI: 10.3390/biom15040550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/29/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Multitarget drug discovery, including host-directed therapy, is particularly promising for tuberculosis (TB) due to the resilience of Mycobacterium tuberculosis (Mtb) as well as the complexity of the host's immune response. In this proof-of-concept study, we used high-content imaging to test a novel panel of dual glycogen synthase kinase 3 beta (GSK-3β) and histone deacetylase (HDAC) 1 and 6 inhibitor candidates for their efficacy in reducing the growth of green fluorescent protein (GFP)-expressing mycobacteria in human primary macrophages. We demonstrate that all ten test compounds, also including the GSK-3β inhibitor SB415286, exhibit an antimycobacterial effect of 20-60% at low micromolar doses and are non-toxic to host cells. Mtb growth showed a positive correlation with the respective 50% inhibitory concentration (IC50) values of GSK-3β, HDAC1, and HDAC6 in each compound, indicating that compounds with a potent IC50 value for HDAC1, in particular, corresponded to higher antimycobacterial activity. Furthermore, the results from multiparametric flow cytometry and a customized multiplex RNA array demonstrated that SB415286 and selected compounds, C02 and C06, could modulate immune polarization and inflammation in Mtb-infected macrophages involving an enhanced expression of CCL2, IL-10 and S100A9, but a decrease in inflammatory mediators including COX-2, TNF-α, and NFκB. These data suggest that GSK-3β inhibition alone can decrease the intracellular growth of mycobacteria and regulate macrophage inflammation, while dual GSK-3β/HDAC inhibitors enhance this efficacy. Accordingly, the tailored design of dual GSK-3β/HDAC inhibitors could represent an innovative approach to host-directed therapy in TB.
Collapse
Affiliation(s)
- Sadaf Kalsum
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, ANA Futura, 141 52 Huddinge, Sweden; (S.K.); (R.X.); (M.A.)
- Division of Medical Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden;
| | - Ruilan Xu
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, ANA Futura, 141 52 Huddinge, Sweden; (S.K.); (R.X.); (M.A.)
| | - Mira Akber
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, ANA Futura, 141 52 Huddinge, Sweden; (S.K.); (R.X.); (M.A.)
| | - Shengjie Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China; (S.H.); (Y.C.)
| | - Maria Lerm
- Division of Medical Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden;
| | - Yuqing Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China; (S.H.); (Y.C.)
| | - Magda Lourda
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, 141 52 Huddinge, Sweden;
| | - Yang Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China; (S.H.); (Y.C.)
| | - Susanna Brighenti
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, ANA Futura, 141 52 Huddinge, Sweden; (S.K.); (R.X.); (M.A.)
| |
Collapse
|
3
|
Shahani A, Slika H, Elbeltagy A, Lee A, Peters C, Dotson T, Raj D, Tyler B. The epigenetic mechanisms involved in the treatment resistance of glioblastoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:12. [PMID: 40201311 PMCID: PMC11977385 DOI: 10.20517/cdr.2024.157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/21/2024] [Accepted: 12/03/2024] [Indexed: 04/10/2025]
Abstract
Glioblastoma (GBM) is an aggressive malignant brain tumor with almost inevitable recurrence despite multimodal management with surgical resection and radio-chemotherapy. While several genetic, proteomic, cellular, and anatomic factors interplay to drive recurrence and promote treatment resistance, the epigenetic component remains among the most versatile and heterogeneous of these factors. Herein, the epigenetic landscape of GBM refers to a myriad of modifications and processes that can alter gene expression without altering the genetic code of cancer cells. These processes encompass DNA methylation, histone modification, chromatin remodeling, and non-coding RNA molecules, all of which have been found to be implicated in augmenting the tumor's aggressive behavior and driving its resistance to therapeutics. This review aims to delve into the underlying interactions that mediate this role for each of these epigenetic components. Further, it discusses the two-way relationship between epigenetic modifications and tumor heterogeneity and plasticity, which are crucial to effectively treat GBM. Finally, we build on the previous characterization of epigenetic modifications and interactions to explore specific targets that have been investigated for the development of promising therapeutic agents.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Betty Tyler
- Hunterian Neurosurgical Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
4
|
Duraj T, Kalamian M, Zuccoli G, Maroon JC, D'Agostino DP, Scheck AC, Poff A, Winter SF, Hu J, Klement RJ, Hickson A, Lee DC, Cooper I, Kofler B, Schwartz KA, Phillips MCL, Champ CE, Zupec-Kania B, Tan-Shalaby J, Serfaty FM, Omene E, Arismendi-Morillo G, Kiebish M, Cheng R, El-Sakka AM, Pflueger A, Mathews EH, Worden D, Shi H, Cincione RI, Spinosa JP, Slocum AK, Iyikesici MS, Yanagisawa A, Pilkington GJ, Chaffee A, Abdel-Hadi W, Elsamman AK, Klein P, Hagihara K, Clemens Z, Yu GW, Evangeliou AE, Nathan JK, Smith K, Fortin D, Dietrich J, Mukherjee P, Seyfried TN. Clinical research framework proposal for ketogenic metabolic therapy in glioblastoma. BMC Med 2024; 22:578. [PMID: 39639257 PMCID: PMC11622503 DOI: 10.1186/s12916-024-03775-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor in adults, with a universally lethal prognosis despite maximal standard therapies. Here, we present a consensus treatment protocol based on the metabolic requirements of GBM cells for the two major fermentable fuels: glucose and glutamine. Glucose is a source of carbon and ATP synthesis for tumor growth through glycolysis, while glutamine provides nitrogen, carbon, and ATP synthesis through glutaminolysis. As no tumor can grow without anabolic substrates or energy, the simultaneous targeting of glycolysis and glutaminolysis is expected to reduce the proliferation of most if not all GBM cells. Ketogenic metabolic therapy (KMT) leverages diet-drug combinations that inhibit glycolysis, glutaminolysis, and growth signaling while shifting energy metabolism to therapeutic ketosis. The glucose-ketone index (GKI) is a standardized biomarker for assessing biological compliance, ideally via real-time monitoring. KMT aims to increase substrate competition and normalize the tumor microenvironment through GKI-adjusted ketogenic diets, calorie restriction, and fasting, while also targeting glycolytic and glutaminolytic flux using specific metabolic inhibitors. Non-fermentable fuels, such as ketone bodies, fatty acids, or lactate, are comparatively less efficient in supporting the long-term bioenergetic and biosynthetic demands of cancer cell proliferation. The proposed strategy may be implemented as a synergistic metabolic priming baseline in GBM as well as other tumors driven by glycolysis and glutaminolysis, regardless of their residual mitochondrial function. Suggested best practices are provided to guide future KMT research in metabolic oncology, offering a shared, evidence-driven framework for observational and interventional studies.
Collapse
Affiliation(s)
- Tomás Duraj
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA.
| | | | - Giulio Zuccoli
- Neuroradiology, Private Practice, Philadelphia, PA, 19103, USA
| | - Joseph C Maroon
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Dominic P D'Agostino
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | - Adrienne C Scheck
- Department of Child Health, University of Arizona College of Medicine, Phoenix, Phoenix, AZ, 85004, USA
| | - Angela Poff
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | - Sebastian F Winter
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, 02114, USA
| | - Jethro Hu
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Rainer J Klement
- Department of Radiotherapy and Radiation Oncology, Leopoldina Hospital Schweinfurt, 97422, Schweinfurt, Germany
| | | | - Derek C Lee
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA
| | - Isabella Cooper
- Ageing Biology and Age-Related Diseases Group, School of Life Sciences, University of Westminster, London, W1W 6UW, UK
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Müllner Hauptstr. 48, 5020, Salzburg, Austria
| | - Kenneth A Schwartz
- Department of Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Matthew C L Phillips
- Department of Neurology, Waikato Hospital, Hamilton, 3204, New Zealand
- Department of Medicine, University of Auckland, Auckland, 1142, New Zealand
| | - Colin E Champ
- Exercise Oncology & Resiliency Center and Department of Radiation Oncology, Allegheny Health Network, Pittsburgh, PA, 15212, USA
| | | | - Jocelyn Tan-Shalaby
- School of Medicine, University of Pittsburgh, Veteran Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA
| | - Fabiano M Serfaty
- Department of Clinical Medicine, State University of Rio de Janeiro (UERJ), Rio de Janeiro, RJ, 20550-170, Brazil
- Serfaty Clínicas, Rio de Janeiro, RJ, 22440-040, Brazil
| | - Egiroh Omene
- Department of Oncology, Cross Cancer Institute, Edmonton, AB, T6G 1Z2, Canada
| | - Gabriel Arismendi-Morillo
- Department of Medicine, Faculty of Health Sciences, University of Deusto, 48007, Bilbao (Bizkaia), Spain
- Facultad de Medicina, Instituto de Investigaciones Biológicas, Universidad del Zulia, Maracaibo, 4005, Venezuela
| | | | - Richard Cheng
- Cheng Integrative Health Center, Columbia, SC, 29212, USA
| | - Ahmed M El-Sakka
- Metabolic Terrain Institute of Health, East Congress Street, Tucson, AZ, 85701, USA
| | - Axel Pflueger
- Pflueger Medical Nephrologyand , Internal Medicine Services P.L.L.C, 6 Nelson Road, Monsey, NY, 10952, USA
| | - Edward H Mathews
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, 0002, South Africa
| | | | - Hanping Shi
- Department of Gastrointestinal Surgery and Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Raffaele Ivan Cincione
- Department of Clinical and Experimental Medicine, University of Foggia, 71122, Foggia, Puglia, Italy
| | - Jean Pierre Spinosa
- Integrative Oncology, Breast and Gynecologic Oncology Surgery, Private Practice, Rue Des Terreaux 2, 1002, Lausanne, Switzerland
| | | | - Mehmet Salih Iyikesici
- Department of Medical Oncology, Altınbaş University Bahçelievler Medical Park Hospital, Istanbul, 34180, Turkey
| | - Atsuo Yanagisawa
- The Japanese College of Intravenous Therapy, Tokyo, 150-0013, Japan
| | | | - Anthony Chaffee
- Department of Neurosurgery, Sir Charles Gairdner Hospital, Perth, 6009, Australia
| | - Wafaa Abdel-Hadi
- Clinical Oncology Department, Cairo University, Giza, 12613, Egypt
| | - Amr K Elsamman
- Neurosurgery Department, Cairo University, Giza, 12613, Egypt
| | - Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, 6410 Rockledge Drive, Suite 610, Bethesda, MD, 20817, USA
| | - Keisuke Hagihara
- Department of Advanced Hybrid Medicine, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Zsófia Clemens
- International Center for Medical Nutritional Intervention, Budapest, 1137, Hungary
| | - George W Yu
- George W, Yu Foundation For Nutrition & Health and Aegis Medical & Research Associates, Annapolis, MD, 21401, USA
| | - Athanasios E Evangeliou
- Department of Pediatrics, Medical School, Aristotle University of Thessaloniki, Papageorgiou Hospital, Efkarpia, 56403, Thessaloniki, Greece
| | - Janak K Nathan
- Dr. DY Patil Medical College, Hospital and Research Centre, Pune, Maharashtra, 411018, India
| | - Kris Smith
- Barrow Neurological Institute, Dignity Health St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA
| | - David Fortin
- Université de Sherbrooke, Sherbrooke, QC, J1K 2R1, Canada
| | - Jorg Dietrich
- Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, 02114, USA
| | | | | |
Collapse
|
5
|
Begum S, Irvin SD, Cox CK, Huang Z, Wilson JJ, Monroe JD, Gibert Y. Anti-ovarian cancer migration and toxicity characteristics of a platinum(IV) pro-drug with axial HDAC inhibitor ligands in zebrafish models. Invest New Drugs 2024; 42:644-654. [PMID: 39433643 PMCID: PMC11625067 DOI: 10.1007/s10637-024-01479-3] [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: 06/24/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024]
Abstract
Ovarian cancer is the fifth leading cause of cancer related death in the United States. Cisplatin is a platinum-based anti-cancer drug used against ovarian cancer that enters malignant cells and then damages DNA causing cell death. Typically, ovarian cancer cells become resistant to cisplatin making it necessary to increase subsequent dosage, which usually leads to side-effects including irreversible damage to kidney and auditory system tissue. Ovarian cancer resistance is often associated with upregulation of histone deacetylase (HDAC) enzymes that cause DNA to adopt a closed configuration which reduces the ability of cisplatin to target and damage DNA. Compound B, a platinum(IV) complex with two axial phenylbutyrate (PBA) HDAC inhibitor ligands attached to a cisplatin core, can simultaneously inhibit HDAC activity and damage DNA causing decreased cancer cell viability in cisplatin-sensitive (A2780) and -resistant (A2780cis) ovarian cancer cell lines. However, compound B was not previously evaluated in vivo. As simultaneously inhibiting HDAC-mediated resistance with cisplatin treatment could potentiate the platinum drug's effect, we first confirmed the anti-cancer effect of compound B in the A2780 and A2780cis cell lines using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide spectrophotometric assay. Then, we used zebrafish embryo and transgenic animal models to comparatively analyze the effect of cisplatin, compound B, and controls on general organismal, auditory, and renal system toxicity, and cancer metastasis. We found that lower dosages of compound B (0.3 or 0.6 µM) than of cisplatin (2.0 µM) could cause similar or decreased levels of general, auditory, and renal tissue toxicity, and at 0.6 µM, compound B reduces cancer metastasis more than 2.0 µM cisplatin.
Collapse
Affiliation(s)
- Salma Begum
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 State Street, Jackson, MS, 39216, USA
| | - Scheldon D Irvin
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 State Street, Jackson, MS, 39216, USA
| | - Carol K Cox
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 State Street, Jackson, MS, 39216, USA
| | - Zhouyang Huang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853- 1301, USA
| | - Justin J Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853- 1301, USA
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA, 93106-9510, USA
| | - Jerry D Monroe
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 State Street, Jackson, MS, 39216, USA
| | - Yann Gibert
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
6
|
Kusaczuk M, Tyszka N, Krętowski R, Cechowska-Pasko M. The Proteasome Inhibitor Marizomib Evokes Endoplasmic Reticulum Stress and Promotes Apoptosis in Human Glioblastoma Cells. Pharmaceuticals (Basel) 2024; 17:1089. [PMID: 39204194 PMCID: PMC11357632 DOI: 10.3390/ph17081089] [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: 07/25/2024] [Revised: 08/09/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Proteasomes play an important role in the physiology of cancer cells, and inhibition of their activity may be used as a promising therapeutic strategy against glioblastoma (GBM). Although certain proteasome inhibitors (PIs) have been approved for the treatment of other malignancies, they have limited effectiveness against GBM due to low brain bioavailability. Marizomib (MZB) is an irreversible, second-generation proteasome inhibitor, which unlike other PIs can penetrate through the blood-brain barrier, making it a promising therapeutic tool in brain malignancies. The antitumor activity of MZB was investigated in LN229 and U118 cells. The MTT test and the ATP-based assay were performed to evaluate cytotoxicity. Flow cytometry analysis was used to determine the apoptotic death of GBM cells. Luminescent assays were used to assess levels of reactive oxygen species (ROS) and the activity of caspase 3/7. RT-qPCR and Western blot analyses were used to determine gene and protein expressions. Marizomib decreased the viability and caused apoptotic death of GBM cells. The proapoptotic effect was accompanied by activation of caspase 3 and overexpression of cl-PARP, Noxa, Cyt C, and DR5. Moreover, treatment with MZB triggered endoplasmic reticulum (ER) stress, as shown by increased expressions of GRP78, IRE1α, p-EIF2α, p-SAPK/JNK, CHOP, ATF6α, and ATF4. On the contrary, overproduction of ROS or increased expressions of ERO1α, LC3 II, Beclin 1, and ATG5 were not detected, suggesting that neither oxidative stress nor autophagy were involved in the process of MZB-induced cell death. Thus, marizomib represents a potentially promising compound for facilitating further progress in brain cancer therapy.
Collapse
Affiliation(s)
- Magdalena Kusaczuk
- Department of Pharmaceutical Biochemistry, Medical University of Bialystok, Mickiewicza 2A, 15-222 Bialystok, Poland; (N.T.); (R.K.)
| | | | | | - Marzanna Cechowska-Pasko
- Department of Pharmaceutical Biochemistry, Medical University of Bialystok, Mickiewicza 2A, 15-222 Bialystok, Poland; (N.T.); (R.K.)
| |
Collapse
|
7
|
Phua YL, D'Annibale OM, Karunanidhi A, Mohsen AW, Kirmse B, Dobrowolski SF, Vockley J. A multiomics approach reveals evidence for phenylbutyrate as a potential treatment for combined D,L-2- hydroxyglutaric aciduria. Mol Genet Metab 2024; 142:108495. [PMID: 38772223 DOI: 10.1016/j.ymgme.2024.108495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/30/2024] [Accepted: 05/13/2024] [Indexed: 05/23/2024]
Abstract
PURPOSE To identify therapies for combined D, L-2-hydroxyglutaric aciduria (C-2HGA), a rare genetic disorder caused by recessive variants in the SLC25A1 gene. METHODS Patients C-2HGA were identified and diagnosed by whole exome sequencing and biochemical genetic testing. Patient derived fibroblasts were then treated with phenylbutyrate and the functional effects assessed by metabolomics and RNA-sequencing. RESULTS In this study, we demonstrated that C-2HGA patient derived fibroblasts exhibited impaired cellular bioenergetics. Moreover, Fibroblasts form one patient exhibited worsened cellular bioenergetics when supplemented with citrate. We hypothesized that treating patient cells with phenylbutyrate (PB), an FDA approved pharmaceutical drug that conjugates glutamine for renal excretion, would reduce mitochondrial 2-ketoglutarate, thereby leading to improved cellular bioenergetics. Metabolomic and RNA-seq analyses of PB-treated fibroblasts demonstrated a significant decrease in intracellular 2-ketoglutarate, 2-hydroxyglutarate, and in levels of mRNA coding for citrate synthase and isocitrate dehydrogenase. Consistent with the known action of PB, an increased level of phenylacetylglutamine in patient cells was consistent with the drug acting as 2-ketoglutarate sink. CONCLUSION Our pre-clinical studies suggest that citrate supplementation has the possibility exacerbating energy metabolism in this condition. However, improvement in cellular bioenergetics suggests phenylbutyrate might have interventional utility for this rare disease.
Collapse
MESH Headings
- Humans
- Phenylbutyrates/pharmacology
- Phenylbutyrates/therapeutic use
- Fibroblasts/metabolism
- Fibroblasts/drug effects
- Glutarates/metabolism
- Ketoglutaric Acids/metabolism
- Energy Metabolism/drug effects
- Energy Metabolism/genetics
- Mitochondria/drug effects
- Mitochondria/metabolism
- Mitochondria/genetics
- Metabolomics
- Exome Sequencing
- Citrate (si)-Synthase/metabolism
- Citrate (si)-Synthase/genetics
- Brain Diseases, Metabolic, Inborn/drug therapy
- Brain Diseases, Metabolic, Inborn/genetics
- Brain Diseases, Metabolic, Inborn/metabolism
- Isocitrate Dehydrogenase/genetics
- Isocitrate Dehydrogenase/metabolism
- Brain Diseases, Metabolic/drug therapy
- Brain Diseases, Metabolic/genetics
- Brain Diseases, Metabolic/metabolism
- Brain Diseases, Metabolic/pathology
- Multiomics
- Mitochondrial Proteins
- Organic Anion Transporters
Collapse
Affiliation(s)
- Yu Leng Phua
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Pathology, Clinical Biochemical Genetics Laboratory, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Olivia M D'Annibale
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Anuradha Karunanidhi
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Al-Walid Mohsen
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Brian Kirmse
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Steven F Dobrowolski
- Department of Pathology, Clinical Biochemical Genetics Laboratory, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Jerry Vockley
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA.
| |
Collapse
|
8
|
Gladwell LR, Ahiarah C, Rasheed S, Rahman SM, Choudhury M. Traditional Therapeutics and Potential Epidrugs for CVD: Why Not Both? Life (Basel) 2023; 14:23. [PMID: 38255639 PMCID: PMC10820772 DOI: 10.3390/life14010023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. In addition to the high mortality rate, people suffering from CVD often endure difficulties with physical activities and productivity that significantly affect their quality of life. The high prevalence of debilitating risk factors such as obesity, type 2 diabetes mellitus, smoking, hypertension, and hyperlipidemia only predicts a bleak future. Current traditional CVD interventions offer temporary respite; however, they compound the severe economic strain of health-related expenditures. Furthermore, these therapeutics can be prescribed indefinitely. Recent advances in the field of epigenetics have generated new treatment options by confronting CVD at an epigenetic level. This involves modulating gene expression by altering the organization of our genome rather than altering the DNA sequence itself. Epigenetic changes are heritable, reversible, and influenced by environmental factors such as medications. As CVD is physiologically and pathologically diverse in nature, epigenetic interventions can offer a ray of hope to replace or be combined with traditional therapeutics to provide the prospect of addressing more than just the symptoms of CVD. This review discusses various risk factors contributing to CVD, perspectives of current traditional medications in practice, and a focus on potential epigenetic therapeutics to be used as alternatives.
Collapse
Affiliation(s)
- Lauren Rae Gladwell
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| | - Chidinma Ahiarah
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| | - Shireen Rasheed
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| | - Shaikh Mizanoor Rahman
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al-Mouz, Nizwa 616, Oman
| | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| |
Collapse
|
9
|
Ling R, Wang J, Fang Y, Yu Y, Su Y, Sun W, Li X, Tang X. HDAC-an important target for improving tumor radiotherapy resistance. Front Oncol 2023; 13:1193637. [PMID: 37503317 PMCID: PMC10368992 DOI: 10.3389/fonc.2023.1193637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/28/2023] [Indexed: 07/29/2023] Open
Abstract
Radiotherapy is an important means of tumor treatment, but radiotherapy resistance has been a difficult problem in the comprehensive treatment of clinical tumors. The mechanisms of radiotherapy resistance include the repair of sublethal damage and potentially lethal damage of tumor cells, cell repopulation, cell cycle redistribution, and reoxygenation. These processes are closely related to the regulation of epigenetic modifications. Histone deacetylases (HDACs), as important regulators of the epigenetic structure of cancer, are widely involved in the formation of tumor radiotherapy resistance by participating in DNA damage repair, cell cycle regulation, cell apoptosis, and other mechanisms. Although the important role of HDACs and their related inhibitors in tumor therapy has been reviewed, the relationship between HDACs and radiotherapy has not been systematically studied. This article systematically expounds for the first time the specific mechanism by which HDACs promote tumor radiotherapy resistance in vivo and in vitro and the clinical application prospects of HDAC inhibitors, aiming to provide a reference for HDAC-related drug development and guide the future research direction of HDAC inhibitors that improve tumor radiotherapy resistance.
Collapse
Affiliation(s)
- Rui Ling
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jingzhi Wang
- Department of Radiotherapy Oncology, Affiliated Yancheng First Hospital of Nanjing University Medical School, First People’s Hospital of Yancheng, Yancheng, China
| | - Yuan Fang
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yunpeng Yu
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuting Su
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wen Sun
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaoqin Li
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiang Tang
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
10
|
Elverson K, Warwicker J, Freeman S, Manson F. Tadalafil Rescues the p.M325T Mutant of Best1 Chloride Channel. Molecules 2023; 28:molecules28083317. [PMID: 37110551 PMCID: PMC10142963 DOI: 10.3390/molecules28083317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/24/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
Bestrophin 1 (Best1) is a chloride channel that localises to the plasma membrane of retinal pigment epithelium (RPE) cells. Mutations in the BEST1 gene are associated with a group of untreatable inherited retinal dystrophies (IRDs) called bestrophinopathies, caused by protein instability and loss-of-function of the Best1 protein. 4PBA and 2-NOAA have been shown to rescue the function, expression, and localisation of Best1 mutants; however, it is of interest to find more potent analogues as the concentration of the drugs required is too high (2.5 mM) to be given therapeutically. A virtual docking model of the COPII Sec24a site, where 4PBA has been shown to bind, was generated and a library of 1416 FDA-approved compounds was screened at the site. The top binding compounds were tested in vitro in whole-cell patch-clamp experiments of HEK293T cells expressing mutant Best1. The application of 25 μM tadalafil resulted in full rescue of Cl- conductance, comparable to wild type Best1 levels, for p.M325T mutant Best1 but not for p.R141H or p.L234V mutants.
Collapse
Affiliation(s)
- Kathleen Elverson
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Jim Warwicker
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, Manchester Institute of Biotechnology, The University of Manchester, Manchester M1 7DN, UK
| | - Sally Freeman
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Forbes Manson
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
11
|
Synthesis, Characterization and Biological Investigation of the Platinum(IV) Tolfenamato Prodrug–Resolving Cisplatin-Resistance in Ovarian Carcinoma Cell Lines. Int J Mol Sci 2023; 24:ijms24065718. [PMID: 36982792 PMCID: PMC10056020 DOI: 10.3390/ijms24065718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
The research on the anticancer potential of platinum(IV) complexes represents one strategy to circumvent the deficits of approved platinum(II) drugs. Regarding the role of inflammation during carcinogenesis, the effects of non-steroidal anti-inflammatory drug (NSAID) ligands on the cytotoxicity of platinum(IV) complexes is of special interest. The synthesis of cisplatin- and oxaliplatin-based platinum(IV) complexes with four different NSAID ligands is presented in this work. Nine platinum(IV) complexes were synthesized and characterized by use of nuclear magnetic resonance (NMR) spectroscopy (1H, 13C, 195Pt, 19F), high-resolution mass spectrometry, and elemental analysis. The cytotoxic activity of eight compounds was evaluated for two isogenic pairs of cisplatin-sensitive and -resistant ovarian carcinoma cell lines. Platinum(IV) fenamato complexes with a cisplatin core showed especially high in vitro cytotoxicity against the tested cell lines. The most promising complex, 7, was further analyzed for its stability in different buffer solutions and behavior in cell cycle and cell death experiments. Compound 7 induces a strong cytostatic effect and cell line-dependent early apoptotic or late necrotic cell death processes. Gene expression analysis suggests that compound 7 acts through a stress-response pathway integrating p21, CHOP, and ATF3.
Collapse
|
12
|
Phua YL, D’Annibale OM, Karunanidhi A, Mohsen AW, Kirmse B, Dobrowolski SF, Vockley J. A multiomics approach to understanding pathology of Combined D,L-2- Hydroxyglutaric Aciduria and phenylbutyrate as potential treatment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.02.526527. [PMID: 36778323 PMCID: PMC9915603 DOI: 10.1101/2023.02.02.526527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Combined D, L-2-Hydroxyglutaric Aciduria (D,L-2HGA) is a rare genetic disorder caused by recessive mutations in the SLC25A1 gene that encodes the mitochondrial citrate carrier protein (CIC). SLC25A1 deficiency leads to a secondary increase in mitochondrial 2-ketoglutarate that, in turn, is reduced to neurotoxic 2-hydroxyglutarate. Clinical symptoms of Combined D,L-2HGA include neonatal encephalopathy, respiratory insufficiency and often with death in infancy. No current therapies exist, although replenishing cytosolic stores by citrate supplementation to replenish cytosolic stores has been proposed. In this study, we demonstrated that patient derived fibroblasts exhibited impaired cellular bioenergetics that were worsened with citrate supplementation. We hypothesized treating patient cells with phenylbutyrate, an FDA approved pharmaceutical drug, would reduce mitochondrial 2-ketoglutarate, leading to improved cellular bioenergetics including oxygen consumption and fatty acid oxidation. Metabolomic and RNA-seq analyses demonstrated a significant decrease in intracellular 2-ketoglutarate, 2-hydroxyglutarate, and in levels of mRNA coding for citrate synthase and isocitrate dehydrogenase. Consistent with the known action of phenylbutyrate, detected levels of phenylacetylglutamine was consistent with the drug acting as 2-ketoglutarate sink in patient cells. Our pre-clinical studies suggest citrate supplementation is unlikely to be an effective treatment of the disorder. However, cellular bioenergetics suggests phenylbutyrate may have interventional utility for this rare disease.
Collapse
Affiliation(s)
- Yu Leng Phua
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Pathology, Clinical Biochemical Genetics Laboratory, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Olivia M D’Annibale
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Anuradha Karunanidhi
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Al-Walid Mohsen
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Brian Kirmse
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Steven F Dobrowolski
- Department of Pathology, Clinical Biochemical Genetics Laboratory, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Jerry Vockley
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| |
Collapse
|
13
|
Everix L, Seane EN, Ebenhan T, Goethals I, Bolcaen J. Introducing HDAC-Targeting Radiopharmaceuticals for Glioblastoma Imaging and Therapy. Pharmaceuticals (Basel) 2023; 16:227. [PMID: 37259375 PMCID: PMC9967489 DOI: 10.3390/ph16020227] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 09/29/2023] Open
Abstract
Despite recent advances in multimodality therapy for glioblastoma (GB) incorporating surgery, radiotherapy, chemotherapy and targeted therapy, the overall prognosis remains poor. One of the interesting targets for GB therapy is the histone deacetylase family (HDAC). Due to their pleiotropic effects on, e.g., DNA repair, cell proliferation, differentiation, apoptosis and cell cycle, HDAC inhibitors have gained a lot of attention in the last decade as anti-cancer agents. Despite their known underlying mechanism, their therapeutic activity is not well-defined. In this review, an extensive overview is given of the current status of HDAC inhibitors for GB therapy, followed by an overview of current HDAC-targeting radiopharmaceuticals. Imaging HDAC expression or activity could provide key insights regarding the role of HDAC enzymes in gliomagenesis, thus identifying patients likely to benefit from HDACi-targeted therapy.
Collapse
Affiliation(s)
- Liesbeth Everix
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, 2610 Antwerpen, Belgium
| | - Elsie Neo Seane
- Department of Medical Imaging and Therapeutic Sciences, Cape Peninsula University of Technology, Cape Town 7530, South Africa
| | - Thomas Ebenhan
- Pre-Clinical Imaging Facility (PCIF), (NuMeRI) NPC, Pretoria 0001, South Africa
- Department of Science and Technology/Preclinical Drug Development Platform (PCDDP), North West University, Potchefstroom 2520, South Africa
- Nuclear Medicine, University of Pretoria, Pretoria 0001, South Africa
| | - Ingeborg Goethals
- Department of Nuclear Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Julie Bolcaen
- Radiation Biophysics Division, SSC laboratory, iThemba LABS, Cape Town 7131, South Africa
| |
Collapse
|
14
|
Targeting emerging cancer hallmarks by transition metal complexes: Epigenetic reprogramming and epitherapies. Part II. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
15
|
Koganti R, Yadavalli T, Sutar Y, Mallick S, Date A, Shukla D. Topical phenylbutyrate antagonizes NF-κB signaling and resolves corneal inflammation. iScience 2022; 25:105682. [PMID: 36536680 PMCID: PMC9758524 DOI: 10.1016/j.isci.2022.105682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/18/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
Chronic inflammation of the immune privileged cornea originating from viral or nonviral conditions results in significant vision loss. Topical corticosteroids are the common treatments for corneal inflammation, but the drugs cause serious and potentially blinding side effects in the long term. Therefore, new standalone and/or synergistic anti-inflammatory therapies with lower side effects are desperately needed. Here, we show that the aromatic fatty acid phenylbutyrate (PBA) acts as a potent inhibitor of inflammation in preclinical ocular-inflammation models. PBA prevents the transcription as well as translation of pro-inflammatory cytokines by LPS and poly(I:C) via persistent inhibition of NF-κB signaling. PBA quickens the resolution of ocular inflammation in mice by decreasing corneal thickness and immune cell infiltration. More importantly, PBA can synergize with the dexamethasone to antagonize NF-κB signaling at lower drug concentrations. Our results demonstrate that PBA therapy exerts previously unreported anti-inflammatory effects in the eye and facilitates corneal healing during persistent inflammation.
Collapse
Affiliation(s)
- Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, 1855 W. Taylor Street, MC 648, Chicago, IL 60612, USA
| | - Tejabhiram Yadavalli
- Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, 1855 W. Taylor Street, MC 648, Chicago, IL 60612, USA
| | - Yogesh Sutar
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii Hilo, Hilo, HI 96720, USA
- R. Ken Coit College of Pharmacy, The University of Arizona, Tuscon, AZ 85721, USA
| | - Sudipta Mallick
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii Hilo, Hilo, HI 96720, USA
| | - Abhijit Date
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii Hilo, Hilo, HI 96720, USA
- R. Ken Coit College of Pharmacy, The University of Arizona, Tuscon, AZ 85721, USA
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, 1855 W. Taylor Street, MC 648, Chicago, IL 60612, USA
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
16
|
The role of branched chain amino acids metabolic disorders in tumorigenesis and progression. Biomed Pharmacother 2022; 153:113390. [DOI: 10.1016/j.biopha.2022.113390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 11/20/2022] Open
|
17
|
Krętowski R, Cechowska-Pasko M. The Reduced Graphene Oxide (rGO) Induces Apoptosis, Autophagy and Cell Cycle Arrest in Breast Cancer Cells. Int J Mol Sci 2022; 23:9285. [PMID: 36012549 PMCID: PMC9409172 DOI: 10.3390/ijms23169285] [Citation(s) in RCA: 5] [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: 07/15/2022] [Revised: 08/02/2022] [Accepted: 08/12/2022] [Indexed: 11/18/2022] Open
Abstract
Reduced graphene oxide (rGO) has already been reported as a potential cytostatic agent in various cancers. However, the mechanisms underlying rGO's cytotoxicity are still insufficiently understood. Thus, the aim of the study was to investigate the molecular and cellular effects of rGO in breast cancer. Given this, two cell lines, MDA-MB-231 and ZR-75-1, were analyzed using MTT test, flow cytometry and Western blot assay. Incubation with rGO resulted in a multitude of effects, including the stimulation of autophagy, cell cycle arrest and, finally, the apoptotic death of cancer cells. Notably, rGO had minimal effect on normal human fibroblasts. Apoptosis in cancer cells was accompanied by decreased mitochondrial membrane potential, the deregulated expression of mitochondrial proteins and the activation of caspase 9 and caspase 3, suggesting that rGO predominantly induced apoptosis via intrinsic pathway. The analysis of LC3 protein expression revealed that rGO also caused autophagy in breast cancer cells. Moreover, rGO treatment resulted in cell cycle arrest, which was accompanied by deregulated p21 expression. Altogether, rGO seems to have multidirectional cytostatic and cytotoxic effects in breast cancer cells, making it a promising agent worthy of further investigation.
Collapse
Affiliation(s)
- Rafał Krętowski
- Department of Pharmaceutical Biochemistry, Medical University of Białystok, Mickiewicza 2A, 15-222 Białystok, Poland
| | | |
Collapse
|
18
|
Tao X, Zhu Y, Diaz-Perez Z, Yu SH, Foley JR, Stewart TM, Casero RA, Steet R, Zhai RG. Phenylbutyrate modulates polyamine acetylase and ameliorates Snyder-Robinson syndrome in a Drosophila model and patient cells. JCI Insight 2022; 7:e158457. [PMID: 35801587 PMCID: PMC9310527 DOI: 10.1172/jci.insight.158457] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/20/2022] [Indexed: 11/26/2022] Open
Abstract
Polyamine dysregulation plays key roles in a broad range of human diseases from cancer to neurodegeneration. Snyder-Robinson syndrome (SRS) is the first known genetic disorder of the polyamine pathway, caused by X-linked recessive loss-of-function mutations in spermine synthase. In the Drosophila SRS model, altered spermidine/spermine balance has been associated with increased generation of ROS and aldehydes, consistent with elevated spermidine catabolism. These toxic byproducts cause mitochondrial and lysosomal dysfunction, which are also observed in cells from SRS patients. No efficient therapy is available. We explored the biochemical mechanism and discovered acetyl-CoA reduction and altered protein acetylation as potentially novel pathomechanisms of SRS. We repurposed the FDA-approved drug phenylbutyrate (PBA) to treat SRS using an in vivo Drosophila model and patient fibroblast cell models. PBA treatment significantly restored the function of mitochondria and autolysosomes and extended life span in vivo in the Drosophila SRS model. Treating fibroblasts of patients with SRS with PBA ameliorated autolysosome dysfunction. We further explored the mechanism of drug action and found that PBA downregulates the first and rate-limiting spermidine catabolic enzyme spermidine/spermine N1-acetyltransferase 1 (SAT1), reduces the production of toxic metabolites, and inhibits the reduction of the substrate acetyl-CoA. Taken together, we revealed PBA as a potential modulator of SAT1 and acetyl-CoA levels and propose PBA as a therapy for SRS and potentially other polyamine dysregulation-related diseases.
Collapse
Affiliation(s)
- Xianzun Tao
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Yi Zhu
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Zoraida Diaz-Perez
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Seok-Ho Yu
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Jackson R. Foley
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Tracy Murray Stewart
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Robert A. Casero
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Richard Steet
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - R. Grace Zhai
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
19
|
Karissa P, Simpson T, Dawson SP, Low TY, Tay SH, Nordin FDA, Zain SM, Lee PY, Pung YF. Comparison Between Dichloroacetate and Phenylbutyrate Treatment for Pyruvate Dehydrogenase Deficiency. Br J Biomed Sci 2022; 79:10382. [PMID: 35996497 PMCID: PMC9302545 DOI: 10.3389/bjbs.2022.10382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022]
Abstract
Pyruvate dehydrogenase (PDH) deficiency is caused by a number of pathogenic variants and the most common are found in the PDHA1 gene. The PDHA1 gene encodes one of the subunits of the PDH enzyme found in a carbohydrate metabolism pathway involved in energy production. Pathogenic variants of PDHA1 gene usually impact the α-subunit of PDH causing energy reduction. It potentially leads to increased mortality in sufferers. Potential treatments for this disease include dichloroacetate and phenylbutyrate, previously used for other diseases such as cancer and maple syrup urine disease. However, not much is known about their efficacy in treating PDH deficiency. Effective treatment for PDH deficiency is crucial as carbohydrate is needed in a healthy diet and rice is the staple food for a large portion of the Asian population. This review analysed the efficacy of dichloroacetate and phenylbutyrate as potential treatments for PDH deficiency caused by PDHA1 pathogenic variants. Based on the findings of this review, dichloroacetate will have an effect on most PDHA1 pathogenic variant and can act as a temporary treatment to reduce the lactic acidosis, a common symptom of PDH deficiency. Phenylbutyrate can only be used on patients with certain pathogenic variants (p.P221L, p.R234G, p.G249R, p.R349C, p.R349H) on the PDH protein. It is hoped that the review would provide an insight into these treatments and improve the quality of lives for patients with PDH deficiency.
Collapse
Affiliation(s)
- Patricia Karissa
- Division of Biomedical Science, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Malaysia
| | - Timothy Simpson
- Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Simon P Dawson
- Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Teck Yew Low
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Sook Hui Tay
- Division of Biomedical Science, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Malaysia
| | | | - Shamsul Mohd Zain
- Department of Pharmacology, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - Pey Yee Lee
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Yuh-Fen Pung
- Division of Biomedical Science, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Malaysia
| |
Collapse
|
20
|
Iwase A, Takebayashi A, Aoi Y, Favero DS, Watanabe S, Seo M, Kasahara H, Sugimoto K. 4-Phenylbutyric acid promotes plant regeneration as an auxin by being converted to phenylacetic acid via an IBR3-independent pathway. PLANT BIOTECHNOLOGY (TOKYO, JAPAN) 2022; 39:51-58. [PMID: 35601015 PMCID: PMC9080989 DOI: 10.5511/plantbiotechnology.21.1224b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/24/2021] [Indexed: 05/07/2023]
Abstract
4-Phenylbutyric acid (4PBA) is utilized as a drug to treat urea cycle disorders and is also being studied as a potential anticancer drug that acts via its histone deacetylase (HDAC) inhibitor activity. During a search to find small molecules that affect plant regeneration in Arabidopsis, we found that 4PBA treatment promotes this process by mimicking the effect of exogenous auxin. Specifically, plant tissue culture experiments revealed that a medium containing 4PBA enhances callus formation and subsequent shoot regeneration. Analyses with auxin-responsive or cytokinin-responsive marker lines demonstrated that 4PBA specifically enhances AUXIN RESPONSE FACTOR (ARF)-dependent auxin responses. Our western blot analyses showed that 4PBA treatment does not enhance histone acetylation in Arabidopsis, in contrast to butyric acid and trichostatin A, other chemicals often used as HDAC inhibitors, suggesting this mechanism of action does not explain the observed effect of 4PBA on regeneration. Finally, mass spectroscopic analysis and genetic approaches uncovered that 4PBA in Arabidopsis plants is converted to phenylacetic acid (PAA), a known natural auxin, in a manner independent of peroxisomal IBR3-related β-oxidation. This study demonstrates that 4PBA application promotes regeneration in explants via its auxin activity and has potential applications to not only plant tissue culture engineering but also research on the plant β-oxidation pathway.
Collapse
Affiliation(s)
- Akira Iwase
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa 230-0045, Japan
- JST, PRESTO, Kawaguchi, Saitama 332-0012, Japan
- E-mail: Tel: +81-45-503-9570 Fax: +81-45-503-9591
| | - Arika Takebayashi
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa 230-0045, Japan
| | - Yuki Aoi
- Department of Biological Production Science, United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, Fuchu, Tokyo 183-8509, Japan
| | - David S Favero
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa 230-0045, Japan
| | - Shunsuke Watanabe
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa 230-0045, Japan
| | - Mitsunori Seo
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa 230-0045, Japan
| | - Hiroyuki Kasahara
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa 230-0045, Japan
- Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Tokyo 183-8509, Japan
| | - Keiko Sugimoto
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa 230-0045, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- E-mail: Tel: +81-45-503-9570 Fax: +81-45-503-9591
| |
Collapse
|
21
|
A Preliminary Study of the Effect of Quercetin on Cytotoxicity, Apoptosis, and Stress Responses in Glioblastoma Cell Lines. Int J Mol Sci 2022; 23:ijms23031345. [PMID: 35163269 PMCID: PMC8836052 DOI: 10.3390/ijms23031345] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
A growing body of evidence indicates that dietary polyphenols show protective effects against various cancers. However, little is known yet about their activity in brain tumors. Here we investigated the interaction of dietary flavonoid quercetin (QCT) with the human glioblastoma A172 and LBC3 cell lines. We demonstrated that QCT evoked cytotoxic effect in both tested cell lines. Microscopic observations, Annexin V-FITC/PI staining, and elevated expression and activity of caspase 3/7 showed that QCT caused predominantly apoptotic death of A172 cells. Further analyses confirmed enhanced ROS generation, deregulated expression of SOD1 and SOD2, depletion of ATP levels, and an overexpression of CHOP, suggesting the activation of oxidative stress and ER stress upon QCT exposure. Finally, elevated expression and activity of caspase 9, indicative of a mitochondrial pathway of apoptosis, was detected. Conversely, in LBC3 cells the pro-apoptotic effect was observed only after 24 h incubation with QCT, and a shift towards necrotic cell death was observed after 48 h of treatment. Altogether, our data indicate that exposure to QCT evoked cell death via activation of intrinsic pathway of apoptosis in A172 cells. These findings suggest that QCT is worth further investigation as a potential pharmacological agent in therapy of brain tumors.
Collapse
|
22
|
Kim HS, Park YH, Lee HS, Kwon MJ, Song JH, Chang IB. Propranolol Inhibits the Proliferation of Human Glioblastoma Cell Lines through Notch1 and Hes1 Signaling System. J Korean Neurosurg Soc 2021; 64:716-725. [PMID: 34376038 PMCID: PMC8435645 DOI: 10.3340/jkns.2021.0068] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/14/2021] [Indexed: 01/18/2023] Open
Abstract
Objective The anti-tumor effect of the beta-adrenergic receptor antagonist propranolol in breast cancer is well known; however, its activity in glioblastoma is not well-evaluated. The Notch-Hes pathway is known to regulate cell differentiation, proliferation, and apoptosis. We investigated the effect of propranolol to human glioblastoma cell lines, and the role of Notch and Hes signaling in this process.
Methods We performed immunohistochemical staining on 31 surgically resected primary human glioblastoma tissues. We also used glioblastoma cell lines of U87-MG, LN229, and neuroblastoma cell line of SH-SY5Y in this study. The effect of propranolol and isoproterenol on cell proliferation was evaluated using the MTT assay (absorbance 570 nm). The impact of propranolol on gene expression (Notch and Hes) was evaluated using real-time polymerase chain reaction (RT-PCR, whereas protein levels of Notch1 and Hes1 were measured using Western blotting (WB), simultaneously. Small interfering RNA (siRNA) was used to suppress the Notch gene to investigate its role in the proliferation of glioblastoma.
Results Propranolol and isoproterenol caused a dose-dependent decrease in cell proliferation (MTT assay). RT-PCR showed an increase in Notch1 and Hes1 expression by propranolol, whereas WB demonstrated increase in Notch1 protein, but a decrease in Hes1 by propranolol. The proliferation of U87-MG and LN229 was not significantly suppressed after transfection with Notch siRNA.
Conclusion These results demonstrated that propranolol suppressed the proliferation of glioblastoma cell lines and neuroblastoma cell line, and Hes1 was more closely involved than Notch1 was in glioblastoma proliferation.
Collapse
Affiliation(s)
- Hyun Sik Kim
- Department of Neurosurgery, Hallym University Sacred Heart Hospital, Anyang, Korea
| | - Young Han Park
- Department of Obstetrics and Gynecology, Hallym University Sacred Heart Hospital, Anyang, Korea
| | - Heui Seung Lee
- Department of Neurosurgery, Hallym University Sacred Heart Hospital, Anyang, Korea
| | - Mi Jung Kwon
- Department of Pathology, Hallym University Sacred Heart Hospital, Anyang, Korea
| | - Joon Ho Song
- Department of Neurosurgery, Hallym University Sacred Heart Hospital, Anyang, Korea
| | - In Bok Chang
- Department of Neurosurgery, Hallym University Sacred Heart Hospital, Anyang, Korea
| |
Collapse
|
23
|
Pereira S, Moore J, Li JX, Yu WQ, Ghanim H, Vlavcheski F, Joseph YD, Dandona P, Volchuk A, Cummins CL, Tsiani E, Giacca A. 4-Phenylbutyric acid improves free fatty acid-induced hepatic insulin resistance in vivo. Endocr Connect 2021; 10:861-872. [PMID: 34319253 PMCID: PMC8346193 DOI: 10.1530/ec-21-0248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/09/2021] [Indexed: 11/08/2022]
Abstract
Plasma free fatty acids (FFAs) are elevated in obesity and can induce insulin resistance via endoplasmic reticulum (ER) stress. However, it is unknown whether hepatic insulin resistance caused by the elevation of plasma FFAs is alleviated by chemical chaperones. Rats received one of the following i.v. treatments for 48 h: saline, intralipid plus heparin (IH), IH plus the chemical chaperone 4-phenylbutyric acid (PBA), or PBA alone and a hyperinsulinemic-euglycemic clamp was performed during the last 2 h. PBA co-infusion normalized IH-induced peripheral insulin resistance, similar to our previous findings with an antioxidant and an IκBα kinase β (IKKβ) inhibitor. Different from our previous results with the antioxidant and IKKβ inhibitor, PBA also improved IH-induced hepatic insulin resistance in parallel with activation of Akt. Unexpectedly, IH did not induce markers of ER stress in the liver, but PBA prevented IH-induced elevation of phosphorylated eukaryotic initiation factor-2α protein in adipose tissue. PBA tended to decrease circulating fetuin-A and significantly increased circulating fibroblast growth factor 21 (FGF21) without affecting markers of activation of hepatic protein kinase C-δ or p38 mitogen-activated protein kinase that we have previously involved in hepatic insulin resistance in this model. In conclusion: (i) PBA prevented hepatic insulin resistance caused by prolonged plasma FFA elevation without affecting hepatic ER stress markers; (ii) the PBA effect is likely due to increased FGF21 and/or decreased fetuin-A, which directly signal to upregulate Akt activation.
Collapse
Affiliation(s)
- Sandra Pereira
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Jessy Moore
- Department of Health Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Jia-Xu Li
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Wen Qin Yu
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Husam Ghanim
- Division of Endocrinology, Diabetes, and Metabolism, State University of New York at Buffalo, Kaleida Health, Buffalo, New York, USA
| | - Filip Vlavcheski
- Department of Health Sciences, Brock University, St. Catharines, Ontario, Canada
| | | | - Paresh Dandona
- Division of Endocrinology, Diabetes, and Metabolism, State University of New York at Buffalo, Kaleida Health, Buffalo, New York, USA
| | - Allen Volchuk
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Evangelia Tsiani
- Department of Health Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Adria Giacca
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
- Correspondence should be addressed to A Giacca:
| |
Collapse
|
24
|
Efficient Heat Shock Response Affects Hyperthermia-Induced Radiosensitization in a Tumor Spheroid Control Probability Assay. Cancers (Basel) 2021; 13:cancers13133168. [PMID: 34201993 PMCID: PMC8269038 DOI: 10.3390/cancers13133168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/27/2022] Open
Abstract
Hyperthermia (HT) combined with irradiation is a well-known concept to improve the curative potential of radiotherapy. Technological progress has opened new avenues for thermoradiotherapy, even for recurrent head and neck squamous cell carcinomas (HNSCC). Preclinical evaluation of the curative radiosensitizing potential of various HT regimens remains ethically, economically, and technically challenging. One key objective of our study was to refine an advanced 3-D assay setup for HT + RT research and treatment testing. For the first time, HT-induced radiosensitization was systematically examined in two differently radioresponsive HNSCC spheroid models using the unique in vitro "curative" analytical endpoint of spheroid control probability. We further investigated the cellular stress response mechanisms underlying the HT-related radiosensitization process with the aim to unravel the impact of HT-induced proteotoxic stress on the overall radioresponse. HT disrupted the proteome's thermal stability, causing severe proteotoxic stress. It strongly enhanced radiation efficacy and affected paramount survival and stress response signaling networks. Transcriptomics, q-PCR, and western blotting data revealed that HT + RT co-treatment critically triggers the heat shock response (HSR). Pre-treatment with chemical chaperones intensified the radiosensitizing effect, thereby suppressing HT-induced Hsp27 expression. Our data suggest that HT-induced radiosensitization is adversely affected by the proteotoxic stress response. Hence, we propose the inhibition of particular heat shock proteins as a targeting strategy to improve the outcome of combinatorial HT + RT.
Collapse
|
25
|
Duangchan T, Tawonsawatruk T, Angsanuntsukh C, Trachoo O, Hongeng S, Kitiyanant N, Supokawej A. Amelioration of osteogenesis in iPSC-derived mesenchymal stem cells from osteogenesis imperfecta patients by endoplasmic reticulum stress inhibitor. Life Sci 2021; 278:119628. [PMID: 34015290 DOI: 10.1016/j.lfs.2021.119628] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 12/20/2022]
Abstract
AIM Osteogenesis imperfecta (OI) is a hereditary connective tissue disorder primarily caused by mutations in COL1A1 or COL1A2, which encode type I collagen. These mutations affect the quantity and/or quality of collagen composition in bones, leading to bone fragility. Currently, there is still a lack of treatment that addresses disease-causing factors due to an insufficient understanding of the pathological mechanisms involved. MAIN METHODS Induced pluripotent stem cells (iPSCs) were generated from OI patients with glycine substitution mutations in COL1A1 and COL1A2 and developed into mesenchymal stem cells (iPS-MSCs). OI-derived iPS-MSCs underwent in vitro osteogenic induction to study cell growth, osteogenic differentiation capacity, mRNA expression of osteogenic and unfolded protein response (UPR) markers and apoptosis. The effects of 4-phenylbutyric acid (4-PBA) were examined after treatment of OI iPS-MSCs during osteogenesis. KEY FINDINGS OI-derived iPS-MSCs exhibited decreased cell growth and impaired osteogenic differentiation and collagen expression. Expression of UPR genes was increased, which led to an increase in apoptotic cell death. 4-PBA treatment decreased apoptotic cells and reduced expression of UPR genes, including HSPA5, XBP1, ATF4, DDIT3, and ATF6. Osteogenic phenotypes, including RUNX2, SPP1, BGLAP, and IBPS expression, as well as calcium mineralization, were also improved. SIGNIFICANCE MSCs differentiated from disease-specific iPSCs have utility as a disease model for identifying disease-specific treatments. In addition, the ER stress-associated UPR could be a pathogenic mechanism associated with OI. Treatment with 4-PBA alleviated OI pathogenesis by attenuating UPR markers and apoptotic cell death.
Collapse
Affiliation(s)
- Thitinat Duangchan
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phutthamonthon, Nakhon Pathom 73170, Thailand
| | - Tulyapruek Tawonsawatruk
- Department of Orthopedics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Chanika Angsanuntsukh
- Department of Orthopedics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Objoon Trachoo
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Narisorn Kitiyanant
- Stem Cell Research Group, Institute of Molecular Biosciences, Mahidol University, Phutthamonthon, Nakhon Pathom 73170, Thailand
| | - Aungkura Supokawej
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phutthamonthon, Nakhon Pathom 73170, Thailand.
| |
Collapse
|
26
|
Bondarev AD, Attwood MM, Jonsson J, Chubarev VN, Tarasov VV, Schiöth HB. Recent developments of HDAC inhibitors: Emerging indications and novel molecules. Br J Clin Pharmacol 2021; 87:4577-4597. [PMID: 33971031 DOI: 10.1111/bcp.14889] [Citation(s) in RCA: 235] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 04/23/2021] [Accepted: 05/01/2021] [Indexed: 02/06/2023] Open
Abstract
The histone deacetylase (HDAC) enzymes, a class of epigenetic regulators, are historically well established as attractive therapeutic targets. During investigation of trends within clinical trials, we have identified a high number of clinical trials involving HDAC inhibitors, prompting us to further evaluate the current status of this class of therapeutic agents. In total, we have identified 32 agents with HDAC-inhibiting properties, of which 29 were found to interact with the HDAC enzymes as their primary therapeutic target. In this review, we provide an overview of the clinical drug development highlighting the recent advances and provide analysis of specific trials and, where applicable, chemical structures. We found haematologic neoplasms continue to represent the majority of clinical indications for this class of drugs; however, it is clear that there is an ongoing trend towards diversification. Therapies for non-oncology indications including HIV infection, muscular dystrophies, inflammatory diseases as well as neurodegenerative diseases such as Alzheimer's disease, frontotemporal dementia and Friedreich's ataxia are achieving promising clinical progress. Combinatory regimens are proving to be useful to improve responsiveness among FDA-approved agents; however, it often results in increased treatment-related toxicities. This analysis suggests that the indication field is broadening through a high number of clinical trials while several fields of preclinical development are also promising.
Collapse
Affiliation(s)
- Andrey D Bondarev
- Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, Russia.,Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Misty M Attwood
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Jörgen Jonsson
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Vladimir N Chubarev
- Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vadim V Tarasov
- Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, Russia.,Institute of Translational Medicine and Biotechnology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Helgi B Schiöth
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden.,Institute of Translational Medicine and Biotechnology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
27
|
Gyawali A, Kang YS. Transport Alteration of 4-Phenyl Butyric Acid Mediated by a Sodium- and Proton-Coupled Monocarboxylic Acid Transporter System in ALS Model Cell Lines (NSC-34) Under Inflammatory States. J Pharm Sci 2020; 110:1374-1384. [PMID: 33098824 DOI: 10.1016/j.xphs.2020.10.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/16/2020] [Accepted: 10/18/2020] [Indexed: 12/20/2022]
Abstract
4-Phenyl butyric acid (PBA) has histone deacetylase inhibitory and neuroprotective effects. We aimed to examine the transport alteration activity of PBA in control (WT) and disease (MT) model cell lines of an amyotrophic lateral sclerosis (ALS) model. The transport characteristics of PBA were examined uptake rates and mRNA expression levels in NSC-34 cell lines. PBA uptake was pH, sodium, and concentration dependent. The Km and Vmax values for PBA uptake in the MT were more than two-fold higher than those in the WT. The presence of monocarboxylic acids (MA) and inhibitors of MA transporter (MCT) inhibited the uptake of PBA. PBA showed competitive inhibition in the presence of MAs in both cell lines. SiRNA transfection studies showed that PBA can be transported to NSC-34 cell lines through sodium-coupled MCT1. TNF-α and H2O2 increased, but LPS and glutamate reduced the uptake rate after the pretreatment of the MT cell lines. SMCT1 mRNA expression levels, in the presence of oxidative stress inducing agents, showed consistent results with the uptake results. These results demonstrate that PBA can be transported to the ALS model NSC-34 cell lines by sodium- and proton-coupled MCTs, and MA plays a vital role in the prevention of neurodegenerative diseases.
Collapse
Affiliation(s)
- Asmita Gyawali
- College of Pharmacy and Drug Information Research Institute, Sookmyung Women's University, Seoul, Republic of Korea
| | - Young-Sook Kang
- College of Pharmacy and Drug Information Research Institute, Sookmyung Women's University, Seoul, Republic of Korea.
| |
Collapse
|
28
|
Duan C, Evison A, Taylor L, Onur S, Morten K, Townley H. The common diabetes drug metformin can diminish the action of citral against Rhabdomyosarcoma cells in vitro. Phytother Res 2020; 35:1378-1388. [PMID: 33280183 DOI: 10.1002/ptr.6898] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/17/2020] [Accepted: 09/20/2020] [Indexed: 01/12/2023]
Abstract
Rhabdomyosarcoma (RMS) is a rare type of soft tissue sarcoma most commonly found in pediatric patients. Despite progress, new and improved drug regimens are needed to increase survival rates. Citral, a natural product plant oil can induce cell death in cancer cells. Another compound, metformin, isolated originally from French lilac and used by diabetics, has been shown to reduce the incidence of cancer in these patients. Application of citral to RMS cells showed increase in cell death, and RD and RH30 cells showed half maximal inhibitory concentration (IC50 ) values as low as 36.28 μM and 62.37 μM, respectively. It was also shown that the citral initiated cell apoptosis through an increase in reactive oxygen species (ROS) and free calcium. In comparison, metformin only showed moderate cell death in RMS cell lines at a very high concentration (1,000 μM). Combinatorial experiments, however, indicated that citral and metformin worked antagonistically when used together. In particular, the ability of metformin to quench the ROS induced by citral could lead to the suppression of activity. These results clearly indicate that while clinical use of citral is a promising anti-tumor therapy, caution should be exercised in patients using metformin for diabetes.
Collapse
Affiliation(s)
- Chengchen Duan
- Nuffield Department of Women's and Reproductive Health, Oxford University, John Radcliffe Hospital, Oxford, UK
| | - Anna Evison
- Nuffield Department of Women's and Reproductive Health, Oxford University, John Radcliffe Hospital, Oxford, UK
| | - Lucy Taylor
- Nuffield Department of Women's and Reproductive Health, Oxford University, John Radcliffe Hospital, Oxford, UK
| | - Simone Onur
- Department of Molecular Prevention, Kiel University, Kiel, Germany
| | - Karl Morten
- Nuffield Department of Women's and Reproductive Health, Oxford University, John Radcliffe Hospital, Oxford, UK
| | - Helen Townley
- Nuffield Department of Women's and Reproductive Health, Oxford University, John Radcliffe Hospital, Oxford, UK.,Department of Engineering Science, Oxford University, Oxford, UK
| |
Collapse
|
29
|
Monitoring of the Surface Charge Density Changes of Human Glioblastoma Cell Membranes upon Cinnamic and Ferulic Acids Treatment. Int J Mol Sci 2020; 21:ijms21186972. [PMID: 32971943 PMCID: PMC7555054 DOI: 10.3390/ijms21186972] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
Cinnamic acid (CA) and ferulic acid (FA) are naturally occurring phenolic acids claimed to exert beneficial effects against disorders related to oxidative stress, including cancer. One such malignancy that still remains a therapeutic challenge mainly due to its heterogeneity and inaccessibility to therapeutic agents is Glioblastoma multiforme (GBM). Here, the influence of CA and FA on the surface charge density of human GBM cell line LN-229 was studied using the electrophoretic light scattering technique. Also, the cytotoxicity of both phenolic acids was determined by metabolic activity-assessing tetrazolium test (MTT) analysis after exposure to CA and FA for 24 h and 48 h. Results showed that both compounds reduced cell viability of LN-229 cells, with more pronounced effect evoked by CA as reflected in IC50 values. Further analyses demonstrated that, after treatment with both phenolic acids, the negative charge of membranes decreased at high pH values and the positive charge of the membranes increased at low pH values compared to the data obtained for untreated cells. Afterward, a four-equilibrium model was applied to estimate the total surface concentrations of both acidic and basic functional groups and their association constants with solution ions in order to calculate theoretical values of membrane surface charge densities. Then, the theoretical data were compared to the experimental data in order to verify the mathematical model. As such, our results indicate that application of electrochemical methods to determine specific drug-membrane interactions might be crucial for predicting their pharmacological activity and bioavailability.
Collapse
|
30
|
He B, Moreau R. R-α-Lipoic Acid and 4-Phenylbutyric Acid Have Distinct Hypolipidemic Mechanisms in Hepatic Cells. Biomedicines 2020; 8:biomedicines8080289. [PMID: 32824248 PMCID: PMC7460023 DOI: 10.3390/biomedicines8080289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 11/16/2022] Open
Abstract
The constitutive activation of the mechanistic target of rapamycin complex 1 (mTORC1) leads to the overproduction of apoB-containing triacylglycerol-rich lipoproteins in HepG2 cells. R-α-lipoic acid (LA) and 4-phenylbutyric acid (PBA) have hypolipidemic function but their mechanisms of action are not well understood. Here, we reported that LA and PBA regulate hepatocellular lipid metabolism via distinct mechanisms. The use of SQ22536, an inhibitor of adenylyl cyclase, revealed cAMP’s involvement in the upregulation of CPT1A expression by LA but not by PBA. LA decreased the secretion of proprotein convertase subtilisin/kexin type 9 (PCSK9) in the culture media of hepatic cells and increased the abundance of LDL receptor (LDLR) in cellular extracts in part through transcriptional upregulation. Although PBA induced LDLR gene expression, it did not translate into more LDLR proteins. PBA regulated cellular lipid homeostasis through the induction of CPT1A and INSIG2 expression via an epigenetic mechanism involving the acetylation of histone H3, histone H4, and CBP-p300 at the CPT1A and INSIG2 promoters.
Collapse
|
31
|
Dunmore BJ, Yang X, Crosby A, Moore S, Long L, Huang C, Southwood M, Austin ED, Rana A, Upton PD, Morrell NW. 4PBA Restores Signaling of a Cysteine-substituted Mutant BMPR2 Receptor Found in Patients with Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol 2020; 63:160-171. [PMID: 32255665 DOI: 10.1165/rcmb.2019-0321oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Mutations in the gene encoding BMPR2 (bone morphogenetic protein type 2 receptor) are the major cause of heritable pulmonary arterial hypertension (PAH). Point mutations in the BMPR2 ligand-binding domain involving cysteine residues (such as C118W) are causative of PAH and predicted to cause protein misfolding. Using heterologous overexpression systems, we showed previously that these mutations lead to retention of BMPR2 in the endoplasmic reticulum but are partially rescued by chemical chaperones. Here, we sought to determine whether the chemical chaperone 4-phenylbutyrate (4PBA) restores BMPR2 signaling in primary cells and in a knockin mouse harboring a C118W mutation. First, we confirmed dysfunctional BMP signaling in dermal fibroblasts isolated from a family with PAH segregating the BMPR2 C118W mutation. After BMP4 treatment, the induction of downstream signaling targets (Smad1/5, ID1 [inhibitor of DNA binding 1], and ID2) was significantly reduced in C118W mutant cells. Treatment with 4PBA significantly rescued Smad1/5, ID1, and ID2 expression. Pulmonary artery smooth muscle cells isolated from the lungs of heterozygous mice harboring the Bmpr2 C118W mutation exhibited significantly increased proliferation. In the presence of 4PBA, hyperproliferation was dramatically reduced. Furthermore, in vivo, 4PBA treatment of Bmpr2 C118W mice partially rescued Bmpr2 expression, restored downstream signaling, and improved vascular remodeling. These findings demonstrate in primary cells and in a knockin mouse that the repurposed small-molecule chemical chaperone 4PBA might be a promising precision medicine approach to treat PAH in patients with specific subtypes of BMPR2 mutation involving cysteine substitutions in the ligand-binding domain.
Collapse
Affiliation(s)
- Benjamin J Dunmore
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, United Kingdom
| | - XuDong Yang
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, United Kingdom
| | - Alexi Crosby
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, United Kingdom
| | - Stephen Moore
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, United Kingdom
| | - Lu Long
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, United Kingdom
| | - Christopher Huang
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, United Kingdom
| | - Mark Southwood
- Pathology Research, Royal Papworth Hospital NHS Foundation Trust, Cambridge, United Kingdom; and
| | - Eric D Austin
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Amer Rana
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, United Kingdom
| | - Paul D Upton
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, United Kingdom
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, United Kingdom
| |
Collapse
|
32
|
A Small Molecule, 4-Phenylbutyric Acid, Suppresses HCV Replication via Epigenetically Induced Hepatic Hepcidin. Int J Mol Sci 2020; 21:ijms21155516. [PMID: 32752233 PMCID: PMC7432483 DOI: 10.3390/ijms21155516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 07/30/2020] [Accepted: 07/30/2020] [Indexed: 01/02/2023] Open
Abstract
Hepatic hepcidin is a well-known major iron regulator and has been reported to be closely related to hepatitis C virus (HCV) replication. However, pharmacological targeting of the hepcidin in HCV replication has not been reported. A short-chain fatty acid, 4-Phenyl butyrate (4-PBA), is an acid chemical chaperone that acts as a histone deacetylase inhibitor (HDACi) to promote chromosomal histone acetylation. Here, we investigated the therapeutic effect of 4-PBA on hepcidin expression and HCV replication. We used HCV genotype 1b Huh 7.5-Con1 replicon cells and engraftment of NOD/SCID mice as in vitro and in vivo models to test the effect of 4-PBA. It was found that 4-PBA inhibited HCV replication in Huh7.5-Con1 replicon cells in a concentration- and time-dependent manner through the induction of hepcidin expression by epigenetic modification and subsequent upregulation of interferon-α signaling. HCV formed a membranous web composed of double-membrane vesicles and was utilized for RNA replication. Moreover, 4-PBA also disrupted the integrity of the membranous web and interfered with the molecular interactions critical for the assembly of the HCV replication complex. These findings suggest that 4-PBA is a key epigenetic inducer of anti-HCV hepatic hepcidin and might at least in part play a role in targeting host factors related to HCV infection as an attractive complement to current HCV therapies.
Collapse
|
33
|
Chen R, Zhang M, Zhou Y, Guo W, Yi M, Zhang Z, Ding Y, Wang Y. The application of histone deacetylases inhibitors in glioblastoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:138. [PMID: 32682428 PMCID: PMC7368699 DOI: 10.1186/s13046-020-01643-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/13/2020] [Indexed: 12/14/2022]
Abstract
The epigenetic abnormality is generally accepted as the key to cancer initiation. Epigenetics that ensure the somatic inheritance of differentiated state is defined as a crucial factor influencing malignant phenotype without altering genotype. Histone modification is one such alteration playing an essential role in tumor formation, progression, and resistance to treatment. Notably, changes in histone acetylation have been strongly linked to gene expression, cell cycle, and carcinogenesis. The balance of two types of enzyme, histone acetyltransferases (HATs) and histone deacetylases (HDACs), determines the stage of histone acetylation and then the architecture of chromatin. Changes in chromatin structure result in transcriptional dysregulation of genes that are involved in cell-cycle progression, differentiation, apoptosis, and so on. Recently, HDAC inhibitors (HDACis) are identified as novel agents to keep this balance, leading to numerous researches on it for more effective strategies against cancers, including glioblastoma (GBM). This review elaborated influences on gene expression and tumorigenesis by acetylation and the antitumor mechanism of HDACis. Besdes, we outlined the preclinical and clinical advancement of HDACis in GBM as monotherapies and combination therapies.
Collapse
Affiliation(s)
- Rui Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Mengxian Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Yangmei Zhou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenjing Guo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ming Yi
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ziyan Zhang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Yanpeng Ding
- Department of Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430030, China
| | - Yali Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
34
|
Romeo MA, Gilardini Montani MS, Benedetti R, Garufi A, D’Orazi G, Cirone M. PBA Preferentially Impairs Cell Survival of Glioblastomas Carrying mutp53 by Reducing Its Expression Level, Stabilizing wtp53, Downregulating the Mevalonate Kinase and Dysregulating UPR. Biomolecules 2020; 10:biom10040586. [PMID: 32290231 PMCID: PMC7226434 DOI: 10.3390/biom10040586] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/01/2020] [Accepted: 04/08/2020] [Indexed: 12/27/2022] Open
Abstract
Phenylbutyrate (PBA) is a derivative of Butyric Acid (BA), which has the characteristics of being a histone deacetylase (HDAC) inhibitor and acting as a chemical chaperone. It has the potential to counteract a variety of different diseases, from neurodegeneration to cancer. In this study, we investigated the cytotoxic effect of PBA against glioblastoma cells carrying wt or mutant (mut) p53 and found that it exerted a higher cytotoxic effect against the latter in comparison with the former. This could be due to the downregulation of mutp53, to whose pro-survival effects cancer cells become addicted. In correlation with mutp53 reduction and wtp53 activation, PBA downregulated the expression level of mevalonate kinase (MVK), a key kinase of the mevalonate pathway strongly involved in cancer cell survival. Here we differentiated the chaperoning function of PBA from the others anti-cancer potentiality by comparing its effects to those exerted by NaB, another HDACi that derives from BA but, lacking the phenyl group, cannot act as a chemical chaperone. Interestingly, we observed that PBA induced a stronger cytotoxic effect compared to NaB against U373 cells as it skewed the Unfolded Protein Response (UPR) towards cell death induction, upregulating CHOP and downregulating BIP, and was more efficient in downregulating MVK. The findings of this study suggest that PBA represents a promising molecule against glioblastomas, especially those carrying mutp53, and its use, approved by FDA for urea cycle disorders, should be extended to the glioblastoma anticancer therapy.
Collapse
Affiliation(s)
- Maria Anele Romeo
- Department of Experimental Medicine, “Sapienza” University of Rome, Italy, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy; (M.A.R.); (M.S.G.M.); (R.B.)
| | - Maria Saveria Gilardini Montani
- Department of Experimental Medicine, “Sapienza” University of Rome, Italy, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy; (M.A.R.); (M.S.G.M.); (R.B.)
| | - Rossella Benedetti
- Department of Experimental Medicine, “Sapienza” University of Rome, Italy, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy; (M.A.R.); (M.S.G.M.); (R.B.)
| | - Alessia Garufi
- Department of Research, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.G.); (G.D.)
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio”, 66013 Chieti, Italy
| | - Gabriella D’Orazi
- Department of Research, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.G.); (G.D.)
| | - Mara Cirone
- Department of Experimental Medicine, “Sapienza” University of Rome, Italy, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy; (M.A.R.); (M.S.G.M.); (R.B.)
- Correspondence: ; Tel.: +39-06-4997-3319; Fax: +39-06-4456-229
| |
Collapse
|
35
|
Active DNA unwinding and transport by a membrane-adapted helicase nanopore. Nat Commun 2019; 10:5083. [PMID: 31704937 PMCID: PMC6841704 DOI: 10.1038/s41467-019-13047-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 10/14/2019] [Indexed: 02/05/2023] Open
Abstract
Nanoscale transport through nanopores and live-cell membranes plays a vital role in both key biological processes as well as biosensing and DNA sequencing. Active translocation of DNA through these nanopores usually needs enzyme assistance. Here we present a nanopore derived from truncated helicase E1 of bovine papillomavirus (BPV) with a lumen diameter of c.a. 1.3 nm. Cryogenic electron microscopy (cryo-EM) imaging and single channel recording confirm its insertion into planar lipid bilayer (BLM). The helicase nanopore in BLM allows the passive single-stranded DNA (ssDNA) transport and retains the helicase activity in vitro. Furthermore, we incorporate this helicase nanopore into the live cell membrane of HEK293T cells, and monitor the ssDNA delivery into the cell real-time at single molecule level. This type of nanopore is expected to provide an interesting tool to study the biophysics of biomotors in vitro, with potential applications in biosensing, drug delivery and real-time single cell analysis. Active translocation of DNA through nanopores usually needs enzyme assistance. Here authors present a nanopore derived from helicase E1 of bovine papillomavirus (BPV) which acts as a conductive pore embedded in lipid membrane to allow the translocation of ssDNA and unwinding of dsDNA.
Collapse
|
36
|
Naumowicz M, Kusaczuk M, Kruszewski MA, Gál M, Krętowski R, Cechowska-Pasko M, Kotyńska J. The modulating effect of lipid bilayer/p-coumaric acid interactions on electrical properties of model lipid membranes and human glioblastoma cells. Bioorg Chem 2019; 92:103242. [DOI: 10.1016/j.bioorg.2019.103242] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023]
|
37
|
The Modulating Effect of p-Coumaric Acid on The Surface Charge Density of Human Glioblastoma Cell Membranes. Int J Mol Sci 2019; 20:ijms20215286. [PMID: 31653017 PMCID: PMC6862159 DOI: 10.3390/ijms20215286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 01/17/2023] Open
Abstract
p-Coumaric acid (p-CoA), a phenolic acid belonging to the hydroxycinnamic acids family, is a compound with tentative anticancer potential. Microelectrophoretic mobility measurements conducted at various pH values of electrolyte solution were applied to study p-CoA effects on electrical properties of human glioblastoma cell membranes. The obtained results demonstrated that after the p-CoA treatment, the surface charge density of cancer cells changed in alkaline pH solutions, while no noticeable changes were observed in cell membranes incubated with p-CoA compared to control at acidic pH solutions. A four-equilibrium model was used to describe the phenomena occurring on the cell membrane surface. The total surface concentrations of both acidic and basic functional groups and their association constants with solution ions were calculated and used to define theoretical curves of membrane surface charge density versus pH. The resulting theoretical curves and the experimental data were compared to verify the reliability and validity of the adopted model. The deviation of both kinds of data obtained at a higher pH may be caused by disregarding interactions between the functional groups of cancer cells. Processes occurring in the cell membranes after their incubation with p-CoA can lead to disorders of existing equilibria, which result in changes in values of the parameters describing these equilibria.
Collapse
|
38
|
Yang H, Jin X, Dan H, Chen Q. Histone modifications in oral squamous cell carcinoma and oral potentially malignant disorders. Oral Dis 2019; 26:719-732. [PMID: 31056829 DOI: 10.1111/odi.13115] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 04/17/2019] [Accepted: 04/29/2019] [Indexed: 02/05/2023]
Affiliation(s)
- Huamei Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Medicine of Carcinogenesis and Management West China Hospital of Stomatology, Sichuan University Chengdu China
| | - Xin Jin
- College of Stomatology Chongqing Medical University Chongqing China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences Chongqing China
| | - Hongxia Dan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Medicine of Carcinogenesis and Management West China Hospital of Stomatology, Sichuan University Chengdu China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Medicine of Carcinogenesis and Management West China Hospital of Stomatology, Sichuan University Chengdu China
| |
Collapse
|
39
|
Mostoufi A, Baghgoli R, Fereidoonnezhad M. Synthesis, cytotoxicity, apoptosis and molecular docking studies of novel phenylbutyrate derivatives as potential anticancer agents. Comput Biol Chem 2019; 80:128-137. [DOI: 10.1016/j.compbiolchem.2019.03.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/25/2019] [Accepted: 03/21/2019] [Indexed: 02/06/2023]
|
40
|
Li Y, Wang F, Chen X, Wang J, Zhao Y, Li Y, He B. Zinc-dependent Deacetylase (HDAC) Inhibitors with Different Zinc Binding Groups. Curr Top Med Chem 2019; 19:223-241. [PMID: 30674261 DOI: 10.2174/1568026619666190122144949] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/13/2018] [Accepted: 11/15/2018] [Indexed: 12/24/2022]
Abstract
The state of histone acetylation plays a very crucial role in carcinogenesis and its development by chromatin remodeling and thus altering transcription of oncogenes and tumor suppressor genes. Such epigenetic regulation was controlled by zinc-dependent histone deacetylases (HDACs), one of the major regulators. Due to the therapeutic potential of HDACs as one of the promising drug targets in cancer, HDAC inhibitors have been intensively investigated over the last few decades. Notably, there are five HDAC inhibitors already approved to the market. Vorinostat (SAHA), Belinostat (PXD-101) and Romidepsin (FK228) have been approved by Food and Drug Administration (FDA) in USA for treating cutaneous T-cell lymphoma (CTCL) or peripheral T cell lymphoma (PTCL) while Panbinostat (LBH-589) has also been approved by the FDA for the treatment of multiple myeloma. Recently, Chidamide was approved by China Food and Drug Administration (CFDA) for the treatment of PTCL. The structural feature of almost all HDAC inhibitors consists of Cap group, linker, and zinc-binding group (ZBG). The binding of ZBG groups to zinc ion plays a decisive role in the inhibition of HDAC. Therefore, we will summarize the developed HDAC inhibitors according to different ZBG groups and discuss their binding mode with zinc ion.
Collapse
Affiliation(s)
- Yan Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, China.,School of Basic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Fang Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, China.,School of Pharmacy, Guizhou Medical University, Guiyang 550004, China
| | - Xiaoxue Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, China.,School of Pharmacy, Guizhou Medical University, Guiyang 550004, China
| | - Jie Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, China.,School of Pharmacy, Guizhou Medical University, Guiyang 550004, China
| | - Yonglong Zhao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, China.,School of Pharmacy, Guizhou Medical University, Guiyang 550004, China
| | - Yongjun Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, China.,School of Pharmacy, Guizhou Medical University, Guiyang 550004, China.,Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China
| | - Bin He
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, China.,School of Pharmacy, Guizhou Medical University, Guiyang 550004, China
| |
Collapse
|
41
|
Wawruszak A, Kalafut J, Okon E, Czapinski J, Halasa M, Przybyszewska A, Miziak P, Okla K, Rivero-Muller A, Stepulak A. Histone Deacetylase Inhibitors and Phenotypical Transformation of Cancer Cells. Cancers (Basel) 2019; 11:cancers11020148. [PMID: 30691229 PMCID: PMC6406474 DOI: 10.3390/cancers11020148] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 01/18/2019] [Accepted: 01/22/2019] [Indexed: 12/12/2022] Open
Abstract
Histone deacetylase inhibitors (HDIs) are a group of potent epigenetic drugs which have been investigated for their therapeutic potential in various clinical disorders, including hematological malignancies and solid tumors. Currently, several HDIs are already in clinical use and many more are on clinical trials. HDIs have shown efficacy to inhibit initiation and progression of cancer cells. Nevertheless, both pro-invasive and anti-invasive activities of HDIs have been reported, questioning their impact in carcinogenesis. The aim of this review is to compile and discuss the most recent findings on the effect of HDIs on the epithelial-mesenchymal transition (EMT) process in human cancers. We have summarized the impact of HDIs on epithelial (E-cadherin, β-catenin) and mesenchymal (N-cadherin, vimentin) markers, EMT activators (TWIST, SNAIL, SLUG, SMAD, ZEB), as well as morphology, migration and invasion potential of cancer cells. We further discuss the use of HDIs as monotherapy or in combination with existing or novel anti-neoplastic drugs in relation to changes in EMT.
Collapse
Affiliation(s)
- Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
| | - Joanna Kalafut
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
| | - Estera Okon
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
| | - Jakub Czapinski
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Trojdena 2a St., 02-091 Warsaw, Poland.
| | - Marta Halasa
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
| | - Alicja Przybyszewska
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
| | - Paulina Miziak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
| | - Karolina Okla
- The First Department of Gynecologic Oncology and Gynecology, Medical University of Lublin, Staszica 16 St., 20-081 Lublin, Poland.
- Tumor Immunology Laboratory, Medical University of Lublin, Staszica 16 St., 20-081 Lublin, Poland.
| | - Adolfo Rivero-Muller
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
- Faculty of Science and Engineering, Cell Biology, Abo Akademi University, Tykistokatu 6A, 20520 Turku, Finland.
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
| |
Collapse
|
42
|
Liao PL, Huang SH, Hung CH, Huang WK, Tsai CH, Kang JJ, Wang HP, Cheng YW. Efficacy of Azatyrosine-Phenylbutyric Hydroxamides, a Histone Deacetylase Inhibitor, on Chemotherapy-Induced Gastrointestinal Mucositis. Int J Mol Sci 2019; 20:ijms20020249. [PMID: 30634582 PMCID: PMC6359543 DOI: 10.3390/ijms20020249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/27/2018] [Accepted: 01/06/2019] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal mucositis is a serious side effect of chemotherapy. Currently, no effective treatment exists for chemotherapy-induced mucositis, prompting the need to develop an anti-mucositis agent for use in clinics. The present study investigated whether azatyrosine-PBHA (AzP), a histone deacetylase inhibitor, has a therapeutic effect on intestinal mucosa. The results indicated that AzP did not affect the proliferation and viability of cancer cells, outcomes that are achieved by suberoylanilide hydroxamic acid (SAHA). However, AzP could decrease production of the inflammatory mediators interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), and tumor-necrosis factor-α (TNF-α). In vivo histopathological assessment showed that AzP reduced cisplatin-induced injury to the jejunum villi and triggered weight loss in the C57BL/6 mice. Immunohistochemistry (IHC) results demonstrated that mice treated with AzP also recovered from cisplatin-induced injury to the intestinal mucosa. Mechanistic in vitro study using DAVID/KEGG enrichment analysis of microarray data and confirmation by a Western blot indicated the influence of AzP on the MEK/ERK and AKT-dependent pathway. In conclusion, the study demonstrated that AzP might regulate the MEK/ERK MAPK signaling pathway to attenuate MCP-1, TNF-α, and IL-6 production and provide opportunities for the development of new anti-inflammatory drugs targeting mucositis.
Collapse
Affiliation(s)
- Po-Lin Liao
- Institute of Food Safety and Health Risk Assessment, School of Pharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan.
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan.
| | - Shih-Hsuan Huang
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan.
| | - Chien-Hung Hung
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Wei-Kuang Huang
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan.
| | - Chi-Hao Tsai
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan.
| | - Jaw-Jou Kang
- Institute of Food Safety and Health Risk Assessment, School of Pharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Hui-Po Wang
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yu-Wen Cheng
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
43
|
Xu YW, Zheng SB, Chen BS, Wen Y, Zhu SW. [Effect of sodium phenylbutyrate on the sensitivity of PC3/DTX-resistant prostate cancer cells to docetaxel]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 37:130-134. [PMID: 28109113 PMCID: PMC6765754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Indexed: 07/30/2024]
Abstract
OBJECTIVE To investigate the effect of sodium phenylbutyrate (SPB) in modulating docetaxel resistance in human prostate cancer cells in vitro. METHODS A PC3/docetaxel-resistant human prostate cancer cell line PC3/DTX was induced and examined for proliferation, viability, and cell inhibition rate in the presence of SPB. The concentration of concentration of docetaxel required to kill 50% of PC3/DTX cells incubated with 0, 1, 2, and 4 mmol/L SPB was determined using MTT assay. Cell apoptosis rate was analyzed with flow cytometry and the cellular expressions of p21, cyclin D1 and survivin proteins were detected using Western blotting. RESULTS Treatment of PC3/DTX cells with 0, 1, 2, and 4 mmol/L of SPB for 48 h resulted in cell viabilities of (99.85∓2.69)%, (84.68∓3.87)%, (68.65∓4.54)% and (43.54∓5.69)%, and cell inhibition rates of (10.69∓3.65)%, (25.78∓4.58)%, (54.68∓3.98)% and (69.84∓6.54)%, respectively (P<0.05). The concentration of docetaxel required to kill 50% of PC3/DTX cells cultured in the presence of with 0, 1, 2, and 4 mmol/L SPB was 135.98∓2.69, 109.65∓3.87, 87.65∓3.84 and 64.62∓2.98 nmol/L, respectively (P<0.05), and the cell apoptosis rates were (7.2∓0.8)%, (10.2∓0.9)%, (19.8∓2.1)% and (27.4∓2.5)%, respectively. SPB treatment promoted the protein expression of p21 and suppressed the expressions of cyclin D1 and survivin in PC3/DTX cells. CONCLUSION SPB can affect the expressions of p21, cyclin D1, and survivin in PC3/DTX cells and increase the sensitivity to the drug-resistant cells to docetaxel.
Collapse
Affiliation(s)
- Ya-Wen Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.E-mail:
| | | | | | | | | |
Collapse
|
44
|
Xu YW, Zheng SB, Chen BS, Wen Y, Zhu SW. [Effect of sodium phenylbutyrate on the sensitivity of PC3/DTX-resistant prostate cancer cells to docetaxel]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 37:130-134. [PMID: 28109113 PMCID: PMC6765754 DOI: 10.3969/j.issn.1673-4254.2017.01.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Indexed: 06/06/2023]
Abstract
OBJECTIVE To investigate the effect of sodium phenylbutyrate (SPB) in modulating docetaxel resistance in human prostate cancer cells in vitro. METHODS A PC3/docetaxel-resistant human prostate cancer cell line PC3/DTX was induced and examined for proliferation, viability, and cell inhibition rate in the presence of SPB. The concentration of concentration of docetaxel required to kill 50% of PC3/DTX cells incubated with 0, 1, 2, and 4 mmol/L SPB was determined using MTT assay. Cell apoptosis rate was analyzed with flow cytometry and the cellular expressions of p21, cyclin D1 and survivin proteins were detected using Western blotting. RESULTS Treatment of PC3/DTX cells with 0, 1, 2, and 4 mmol/L of SPB for 48 h resulted in cell viabilities of (99.85∓2.69)%, (84.68∓3.87)%, (68.65∓4.54)% and (43.54∓5.69)%, and cell inhibition rates of (10.69∓3.65)%, (25.78∓4.58)%, (54.68∓3.98)% and (69.84∓6.54)%, respectively (P<0.05). The concentration of docetaxel required to kill 50% of PC3/DTX cells cultured in the presence of with 0, 1, 2, and 4 mmol/L SPB was 135.98∓2.69, 109.65∓3.87, 87.65∓3.84 and 64.62∓2.98 nmol/L, respectively (P<0.05), and the cell apoptosis rates were (7.2∓0.8)%, (10.2∓0.9)%, (19.8∓2.1)% and (27.4∓2.5)%, respectively. SPB treatment promoted the protein expression of p21 and suppressed the expressions of cyclin D1 and survivin in PC3/DTX cells. CONCLUSION SPB can affect the expressions of p21, cyclin D1, and survivin in PC3/DTX cells and increase the sensitivity to the drug-resistant cells to docetaxel.
Collapse
Affiliation(s)
- Ya-Wen Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.E-mail:
| | | | | | | | | |
Collapse
|
45
|
Kusaczuk M, Krętowski R, Naumowicz M, Stypułkowska A, Cechowska-Pasko M. Silica nanoparticle-induced oxidative stress and mitochondrial damage is followed by activation of intrinsic apoptosis pathway in glioblastoma cells. Int J Nanomedicine 2018; 13:2279-2294. [PMID: 29695906 PMCID: PMC5905826 DOI: 10.2147/ijn.s158393] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Introduction Recently, the focus of oncological research has been on the optimization of therapeutic strategies targeted at malignant diseases. Nanomedicine utilizing silicon dioxide nanoparticles (SiNPs) is one such strategy and is rapidly developing as a promising tool for cancer diagnosis, imaging, and treatment. Nevertheless, little is known about the mechanisms of action of SiNPs in brain tumors. Materials and methods Here, we explored the effects of 5-15 nm SiNPs in the human glioblastoma cell line LN229. In this respect, MTT assays, microscopic observations, flow cytometry analyses, and luminescent assays were performed. Moreover, RT-qPCR and Western blot analyses were done to determine gene and protein expressions. Results We demonstrated that SiNPs triggered evident cytotoxicity, with microscopic observations of the nuclei, annexin V-fluorescein isothiocyanate/propidium iodide staining, and elevated caspase 3/7 activity, suggesting that SiNPs predominantly induced apoptotic death in LN229 cells. We further showed the occurrence of oxidative stress induced by enhanced reactive oxygen-species generation. This effect was followed by deregulated expression of genes encoding the antioxidant enzymes SOD1, SOD2, and CAT, and impaired mitochondria function. SiNP- induced mitochondrial dysfunction was characterized by membrane-potential collapse, ATP depletion, elevated expression of BAX, PUMA, and NOXA with simultaneous downregulation of BCL2/BCL2L1, and activation of caspase 9. Moreover, RT-qPCR and Western blot analyses demonstrated increased levels of the endoplasmic reticulum stress markers GRP78, GRP94, and DDIT3, as well as strongly increased expressions of the IL1B and COX2 genes, suggesting activation of endoplasmic reticulum stress and a proinflammatory response. Conclusions Altogether, our data indicate that in LN229 cells, SiNPs evoke cell death via activation of the intrinsic apoptosis pathway and suggest that other aspects of cellular function may also be affected. As such, SiNPs represent a potentially promising agent for facilitating further progress in brain cancer therapy. However, further exploration of SiNP long-term toxicity and molecular effects is necessary prior to their widespread application.
Collapse
Affiliation(s)
- Magdalena Kusaczuk
- Department of Pharmaceutical Biochemistry, Medical University of Białystok
| | - Rafał Krętowski
- Department of Pharmaceutical Biochemistry, Medical University of Białystok
| | - Monika Naumowicz
- Institute of Chemistry, University of Białystok, Białystok, Poland
| | - Anna Stypułkowska
- Department of Pharmaceutical Biochemistry, Medical University of Białystok
| | | |
Collapse
|
46
|
Phenyl butyrate inhibits pyruvate dehydrogenase kinase 1 and contributes to its anti-cancer effect. Eur J Pharm Sci 2017; 110:93-100. [DOI: 10.1016/j.ejps.2017.04.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 12/15/2022]
|
47
|
The Effects of Silica Nanoparticles on Apoptosis and Autophagy of Glioblastoma Cell Lines. NANOMATERIALS 2017; 7:nano7080230. [PMID: 28825685 PMCID: PMC5575712 DOI: 10.3390/nano7080230] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 08/10/2017] [Accepted: 08/11/2017] [Indexed: 12/16/2022]
Abstract
Silica nanoparticles (SiNPs) are one of the most commonly used nanomaterials in various medical applications. However, possible mechanisms of the toxicity caused by SiNPs remain unclear. The study presented here provides novel information on molecular and cellular effects of SiNPs in glioblastoma LBC3 and LN-18 cells. It has been demonstrated that SiNPs of 7 nm, 5–15 nm and 10–20 nm induce time- and dose-dependent cytotoxicity in LBC3 and LN-18 cell lines. In contrast to glioblastoma cells, we observed only weak reduction in viability of normal skin fibroblasts treated with SiNPs. Furthermore, in LBC3 cells treated with 5–15 nm SiNPs we noticed induction of apoptosis and necrosis, while in LN-18 cells only necrosis. The 5–15 nm SiNPs were also found to cause oxidative stress, a loss in mitochondrial membrane potential, and changes in the ultrastructure of the mitochondria in LBC3 cells. Quantitative real-time PCR results showed that in LBC3 cells the mRNA levels of pro-apoptotic genes Bim, Bax, Puma, and Noxa were significantly upregulated. An increase in activity of caspase-9 in these cells was also observed. Moreover, the activation of SiNP-induced autophagy was demonstrated in LBC3 cells as shown by an increase in LC3-II/LC3-I ratio, the upregulation of Atg5 gene and an increase in AVOs-positive cells. In conclusion, this research provides novel information concerning molecular mechanisms of apoptosis and autophagy in LBC3 cells.
Collapse
|
48
|
Chemical chaperone, TUDCA unlike PBA, mitigates protein aggregation efficiently and resists ER and non-ER stress induced HepG2 cell death. Sci Rep 2017. [PMID: 28630443 PMCID: PMC5476595 DOI: 10.1038/s41598-017-03940-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stress induced BSA (bovine serum albumin) protein aggregation is effectively mitigated in vitro by TUDCA (tauroursodeoxycholic acid) than by PBA (4- phenylbutyric acid), chemical chaperones approved by FDA for the treatment of biliary cirrhosis and urea cycle disorders respectively. TUDCA, unlike PBA, enhances trypsin mediated digestion of BSA. TUDCA activates PERK, an ER-resident kinase that phosphorylates the alpha-subunit of eukaryotic initiation factor2 (eIF2α) and promotes the expression of activated transcription factor 4 (ATF4) in HepG2 cells. In contrast, PBA induced eIF2α phosphorylation is not mediated by PERK activation and results in low ATF4 expression. Neither chaperones promote expression of BiP, an ER chaperone, and CHOP (C/EBP homologous protein), downstream target of eIF2α-ATF4 pathway. Both chaperones mitigate tunicamycin induced PERK-eIF2α-ATF4-CHOP arm of UPR and expression of BiP. TUDCA, unlike PBA does not decrease cell viability and it also mitigates tunicamycin, UV-irradiation and PBA induced PARP (poly ADP-ribose polymerase) cleavage and cell death. These findings therefore suggest that TUDCA’s antiapoptotic activity to protect HepG2 cells and PBA’s activity that limits tumor cell progression may be important while considering their therapeutic potential.
Collapse
|
49
|
Xue P, Zeng F, Duan Q, Xiao J, Liu L, Yuan P, Fan L, Sun H, Malyarenko OS, Lu H, Xiu R, Liu S, Shao C, Zhang J, Yan W, Wang Z, Zheng J, Zhu F. BCKDK of BCAA Catabolism Cross-talking With the MAPK Pathway Promotes Tumorigenesis of Colorectal Cancer. EBioMedicine 2017; 20:50-60. [PMID: 28501528 PMCID: PMC5478211 DOI: 10.1016/j.ebiom.2017.05.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 05/01/2017] [Accepted: 05/01/2017] [Indexed: 01/13/2023] Open
Abstract
Branched-chain amino acids catabolism plays an important role in human cancers. Colorectal cancer is the third most commonly diagnosed cancer in males and the second in females, and the new global incidence is over 1.2 million cases. The branched-chain α-keto acid dehydrogenase kinase (BCKDK) is a rate-limiting enzyme in branched-chain amino acids catabolism, which plays an important role in many serious human diseases. Here we investigated that abnormal branched-chain amino acids catabolism in colorectal cancer is a result of the disease process, with no role in disease initiation; BCKDK is widely expressed in colorectal cancer patients, and those patients that express higher levels of BCKDK have shorter survival times than those with lower levels; BCKDK promotes cell transformation or colorectal cancer ex vivo or in vivo. Mechanistically, BCKDK promotes colorectal cancer by enhancing the MAPK signaling pathway through direct MEK phosphorylation, rather than by branched-chain amino acids catabolism. And the process above could be inhibited by a BCKDK inhibitor, phenyl butyrate.
Collapse
Affiliation(s)
- Peipei Xue
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Fanfan Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Qiuhong Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Juanjuan Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Lin Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Ping Yuan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Linni Fan
- Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Huimin Sun
- Department of Urology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Olesya S Malyarenko
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China; G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of Russian Academy of Sciences, Laboratory of Enzyme Chemistry, Vladivostok, Russia
| | - Hui Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Ruijuan Xiu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Shaoqing Liu
- Department of State Key Laboratory of Cancer Biology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Chen Shao
- Department of Urology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Jianmin Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Wei Yan
- Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| | - Zhe Wang
- Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| | - Jianyong Zheng
- Department of State Key Laboratory of Cancer Biology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| | - Feng Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China.
| |
Collapse
|
50
|
Khan S, Komarya SK, Jena G. Phenylbutyrate and β-cell function: contribution of histone deacetylases and ER stress inhibition. Epigenomics 2017; 9:711-720. [PMID: 28470097 DOI: 10.2217/epi-2016-0160] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Incidences of diabetes are increasing globally due to involvement of genetic and epigenetic factors. Phenylbutyrate (PBA) is a US FDA approved drug for treatment of urea cycle disorder in children. PBA reduces endoplasmic reticulum (ER) stress and is proven as a potent histone deacetylases (HDACs) inhibitor. Chronic ER stress results in unfolding protein response, which triggers apoptosis. Abnormal ER homoeostasis is responsible for defective processing of several genes/proteins and contributes to β-cell death/failure. Accumulated evidences indicated that HDACs modulate key biochemical pathways and HDAC inhibitors improve β-cell function and insulin resistance by modulating multiple targets. This review highlights the role of PBA on β-cell functions, insulin resistance for possible treatment of diabetes through inhibition of ER stress and HDACs.
Collapse
Affiliation(s)
- Sabbir Khan
- Facility for Risk Assessment & Intervention Studies, Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Sector-67, SAS Nagar, Punjab-160062, India
| | - Sandeep K Komarya
- Facility for Risk Assessment & Intervention Studies, Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Sector-67, SAS Nagar, Punjab-160062, India
| | - Gopabandhu Jena
- Facility for Risk Assessment & Intervention Studies, Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Sector-67, SAS Nagar, Punjab-160062, India
| |
Collapse
|