1
|
Li X, Saiyin H, Chen X, Yu Q, Ma L, Liang W. Ketamine impairs growth cone and synaptogenesis in human GABAergic projection neurons via GSK-3β and HDAC6 signaling. Mol Psychiatry 2024; 29:1647-1659. [PMID: 36414713 PMCID: PMC11371642 DOI: 10.1038/s41380-022-01864-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/23/2022]
Abstract
The growth cone guides the axon or dendrite of striatal GABAergic projection neurons that protrude into the midbrain and cortex and form complex neuronal circuits and synaptic networks in a developing brain, aberrant projections and synaptic connections in the striatum related to multiple brain disorders. Previously, we showed that ketamine, an anesthetic, reduced dendritic growth, dendritic branches, and spine density in human striatal GABAergic neurons. However, whether ketamine affects the growth cone, the synaptic connection of growing striatal GABAergic neurons has not been tested. Using human GABAergic projection neurons derived from human inducible pluripotent stem cells (hiPSCs) and embryonic stem cells (ES) in vitro, we tested ketamine effects on the growth cones and synapses in developing GABAergic neurons by assessing the morphometry and the glycogen synthase kinase-3 (GSK-3) and histone deacetylase 6 (HDAC6) pathway. Ketamine exposure impairs growth cone formation, synaptogenesis, dendritic development, and maturation via ketamine-mediated activation of GSK-3 pathways and inhibiting HDAC6, an essential stabilizing protein for dendritic morphogenesis and synapse maturation. Our findings identified a novel ketamine neurotoxic pathway that depends on GSK-3β and HDAC6 signaling, suggesting that microtubule acetylation is a potential target for reducing ketamine's toxic effect on GABAergic projection neuronal development.
Collapse
Affiliation(s)
- Xuan Li
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, China
- Department of Anesthesiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Hexige Saiyin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Xinyu Chen
- Department of Anatomy and Histology & Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qiong Yu
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Lixiang Ma
- Department of Anatomy and Histology & Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Weimin Liang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Barrett RRG, Nash C, Diennet M, Cotnoir-White D, Doyle C, Mader S, Thomson AA, Gleason JL. Dual-function antiandrogen/HDACi hybrids based on enzalutamide and entinostat. Bioorg Med Chem Lett 2021; 55:128441. [PMID: 34767912 DOI: 10.1016/j.bmcl.2021.128441] [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: 08/01/2021] [Revised: 10/06/2021] [Accepted: 10/31/2021] [Indexed: 11/02/2022]
Abstract
The combination of androgen receptor antagonists with histone deacetylase inhibitors (HDACi) has been shown to be more effective than antiandrogens alone in halting growth of prostate cancer cell lines. Here we have designed, synthesized and assessed a series of antiandrogen/HDACi hybrids by combining structural features of enzalutamide with either SAHA or entinostat. The hybrids are demonstrated to maintain bifunctionality using a fluorometric HDAC assay and a bioluminescence resonance energy transfer (BRET) antiandrogen assay. Antiproliferative assays showed that hybrids bearing o-aminoanilide-based HDACi motifs outperformed hydroxamic acid based HDACi's. The hybrids demonstrated selectivity for epithelial cell lines vs. stromal cell lines, suggesting a potentially useful therapeutic window.
Collapse
Affiliation(s)
- Ryan R G Barrett
- Department of Chemistry, McGill University, 801 Sherbrooke W., Montreal, QC H3A 0B8, Canada
| | - Claire Nash
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Marine Diennet
- Institute for Research in Immunology and Cancer, Pavillon Marcelle Coutu, Université de Montréal, 2950 chemin de Polytechnique, Montreal, QC H3T1J4, Canada
| | - David Cotnoir-White
- Institute for Research in Immunology and Cancer, Pavillon Marcelle Coutu, Université de Montréal, 2950 chemin de Polytechnique, Montreal, QC H3T1J4, Canada
| | - Christopher Doyle
- Department of Chemistry, McGill University, 801 Sherbrooke W., Montreal, QC H3A 0B8, Canada
| | - Sylvie Mader
- Institute for Research in Immunology and Cancer, Pavillon Marcelle Coutu, Université de Montréal, 2950 chemin de Polytechnique, Montreal, QC H3T1J4, Canada; Department of Biochemistry and Molecular Medicine, Pavillon Roger Gaudry, Université de Montréal, 2900 bd Edouard Montpetit, Montreal, QC H3T1J4, Canada
| | - Axel A Thomson
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - James L Gleason
- Department of Chemistry, McGill University, 801 Sherbrooke W., Montreal, QC H3A 0B8, Canada.
| |
Collapse
|
3
|
Liu P, Xiao J, Wang Y, Song X, Huang L, Ren Z, Kitazato K, Wang Y. Posttranslational modification and beyond: interplay between histone deacetylase 6 and heat-shock protein 90. Mol Med 2021; 27:110. [PMID: 34530730 PMCID: PMC8444394 DOI: 10.1186/s10020-021-00375-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/07/2021] [Indexed: 12/17/2022] Open
Abstract
Posttranslational modification (PTM) and regulation of protein stability are crucial to various biological processes. Histone deacetylase 6 (HDAC6), a unique histone deacetylase with two functional catalytic domains (DD1 and DD2) and a ZnF-UBP domain (ubiquitin binding domain, BUZ), regulates a number of biological processes, including gene expression, cell motility, immune response, and the degradation of misfolded proteins. In addition to the deacetylation of histones, other nonhistone proteins have been identified as substrates for HDAC6. Hsp90, a molecular chaperone that is a critical modulator of cell signaling, is one of the lysine deacetylase substrates of HDAC6. Intriguingly, as one of the best-characterized regulators of Hsp90 acetylation, HDAC6 is the client protein of Hsp90. In addition to regulating Hsp90 at the post-translational modification level, HDAC6 also regulates Hsp90 at the gene transcription level. HDAC6 mainly regulates the Hsp90-HSF1 complex through the ZnF-UBP domain, thereby promoting the HSF1 entry into the nucleus and activating gene transcription. The mutual interaction between HDAC6 and Hsp90 plays an important role in the regulation of protein stability, cell migration, apoptosis and other functions. Plenty of of studies have indicated that blocking HDAC6/Hsp90 has a vital regulatory role in multifarious diseases, mainly in cancers. Therefore, developing inhibitors or drugs against HDAC6/Hsp90 becomes a promising development direction. Herein, we review the current knowledge on molecular regulatory mechanisms based on the interaction of HDAC6 and Hsp90 and inhibition of HDAC6 and/or Hsp90 in oncogenesis and progression, antiviral and immune-related diseases and other vital biological processes.
Collapse
Affiliation(s)
- Ping Liu
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Ji Xiao
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Yiliang Wang
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Xiaowei Song
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Lianzhou Huang
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China.,College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhe Ren
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Kaio Kitazato
- Department of Clinical Research Pharmacy, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.
| | - Yifei Wang
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China.
| |
Collapse
|
4
|
Evangelista L, Giuliani L, Pagliei V, Varrassi M, Bruno F, Palumbo P, Arrigoni F, Splendiani A, Di Cesare E, Masciocchi C, Barile A. When to perform vertebroplasty? A retrospective analysis from a single center and a review of the literature. ACTA BIO-MEDICA : ATENEI PARMENSIS 2021; 92:e2021402. [PMID: 34505841 PMCID: PMC8477064 DOI: 10.23750/abm.v92is5.11955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/27/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIM To establish an optimal timing for vertebroplasty in order to obtain a clinically important pain reduction and improving quality of live in patients with osteoporotic or traumatic vertebral fractures. METHODS This study includes 22 vertebroplasty procedures performed from October 2018 to July 2020 in 21 patients with traumatic or osteoporotic vertebral fractures (19 female, two men; age between 53 and 89 years). All treatments were executed under fluoroscopic guidance using 11 or 13 G needle through transpedicular or costovertebral unilateral approach. Each patient underwent conscious sedation, continuously monitored by an anesthesiologist. Preoperative MRI images, obtained by 3T or 1.5T MRI scanner, always showed bone marrow edema. The VAS scale and Roland Morris disability questionnaire (RMdq) were administered to patients before and after the treatment to evaluate pain and life quality. RESULTS 7 patients were treated in the first month after the injury, one was treated twice; 8 patients in the second month, 6 in the third. We observed a reduction of: 5.5 points in the vas scale, 10.3 in the RMdq in the first month; 5.6 points vas, 11.6 points RMdq in the second month; 4 points vas and 9.75 points RMdq in the third month. CONCLUSIONS This study demostrated that, in our preliminary experience, vertebroplasty has the best outcome if performed at 2 months from injury.
Collapse
Affiliation(s)
- Laura Evangelista
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Luca Giuliani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Valeria Pagliei
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Marco Varrassi
- Neuroradiology and Interventional Radiology, San Salvatore Hospital, L'Aquila, Italy.
| | - Federico Bruno
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy and Italian Society of Medical and Interventional Radiology (SIRM) - SIRM Foundation, via della Signora 2, Milan, Italy.
| | - Pierpaolo Palumbo
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy and Italian Society of Medical and Interventional Radiology (SIRM) - SIRM Foundation, via della Signora 2, Milan, Italy.
| | - Francesco Arrigoni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Alessandra Splendiani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Ernesto Di Cesare
- Department of Clinical Medicine, Public Health, Life and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Carlo Masciocchi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Antonio Barile
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
5
|
Palumbo P, Daffinà J, Bruno F, Arrigoni F, Splendiani A, Di Cesare E, Barile A, Masciocchi C. Basics in Magnetic Resonance guided Focused Ultrasound: technical basis and clinical application. A brief overview. ACTA BIO-MEDICA : ATENEI PARMENSIS 2021; 92:e2021403. [PMID: 34505842 PMCID: PMC8477067 DOI: 10.23750/abm.v92is5.11881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/28/2021] [Indexed: 11/30/2022]
Abstract
First applications of high focused ultrasound as intracranial ablative therapy were firstly described in early 50’. Since then, the technological innovations have shown an increasingly safe and effective face of this technique. And in the last few years, Magnetic Resonance (MR) guided Focused Ultrasound (gFUS) has become a valid minimally invasive technique in the treatment of several diseases, from bone tumors to symptomatic uterine fibroids or essential tremors. MR guidance, through the tomographic view, offers the advantage of an accurate target detection and treatment planning. Moreover, real-time monitoring sequences allow to avoid non-target ablation. An adequate knowledge of FUS is essential to understand its clinical effectiveness. Therefore, this brief review aims to debate the physical characteristics of US and the main fields of clinical application.
Collapse
Affiliation(s)
- Pierpaolo Palumbo
- Department of Diagnostic Imaging, area of Cardiovascular and Interventional Imaging, Abruzzo Health Unit 1, Italy and Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy.
| | - Julia Daffinà
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Federico Bruno
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy and Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy.
| | - Francesco Arrigoni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Alessandra Splendiani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Ernesto Di Cesare
- Department of Clinical Medicine, Public Health, Life and Environmental Science, University of L'Aquila, L'Aquila, Italy.
| | - Antonio Barile
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Carlo Masciocchi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
6
|
Sgalambro F, Giordano AV, Carducci S, Varrassi M, Perri M, Arrigoni F, Palumbo P, Bruno F, Bardi L, Mangoni di S Stefano ML, Danti G, Gentili F, Mazzei MA, Di Cesare E, Splendiani A, Masciocchi C, Barile A. The role of interventional radiology in hepatic and renal hemorrhage embolization: single center experience and literature review. ACTA BIO-MEDICA : ATENEI PARMENSIS 2021; 92:e2021405. [PMID: 34505844 PMCID: PMC8477065 DOI: 10.23750/abm.v92is5.11876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/28/2021] [Indexed: 12/03/2022]
Abstract
Background and aim: Intraabdominal hemorrhage secondary to liver and kidney injury is a major cause of morbidity and mortality. Endovascular arterial embolization is an established interventional radiology technique used to treat active bleeding, and its role in managing abdominal hemorrhages is growing, given the increasing trend for conservative treatment. Our study aims to retrospectively evaluate the technical and clinical results and the possible complications of arterial embolization procedures performed in emergency, in post-traumatic, iatrogenic, and pathological hepatic and renal bleedings. Methods: We performed a ten-year, single-center retrospective survey (from January 2010 to December 2019) of all patients treated in emergency by intra-arterial embolization of liver and kidney bleeding. Preliminary CT angiography studies were evaluated, as well as the angiographic findings. Materials used, procedural data, and clinical outcomes, including complications, were recorded. Results: The diagnostic angiography showed a single source of bleeding in 20 cases (66.7%), two bleeding vessels in 4 cases (13.3%), and multiple hemorrhagic sources in 6 cases (20%). All bleeding sources were successfully embolized; in 12 patients (40%), complete embolization was achieved with coils and 18 patients (60%) with hemostatic sponges. In one case, a second embolization procedure was performed for the persistence of hemodynamic instability. No major post-procedural complications were recorded. The mean procedure duration was 65.1 minutes. Conclusions: Based on our experience and literature data, the treatment of endovascular embolization in acute abdominal bleeding of hepatic and renal origin represents the treatment of choice, as it can provide complete therapeutic success in hemodynamically stable patients. (www.actabiomedica.it)
Collapse
Affiliation(s)
- Ferruccio Sgalambro
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy.
| | - Aldo Victor Giordano
- Interventional Radiology and Neuroradiology, San Salvatore Hospital, L'Aquila, Italy.
| | - Sergio Carducci
- Interventional Radiology and Neuroradiology, San Salvatore Hospital, L'Aquila, Italy.
| | - Marco Varrassi
- Interventional Radiology and Neuroradiology, San Salvatore Hospital, L'Aquila, Italy.
| | - Marco Perri
- Diagnostic and Interventional Radiology, SS. Filippo e Nicola Hospital, Avezzano (AQ), Italy.
| | - Francesco Arrigoni
- Department of Emergency and Interventional Radiology, San Salvatore Hospital, L'Aquila, Italy.
| | - Pierpaolo Palumbo
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy and Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy .
| | - Federico Bruno
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy and Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy.
| | - Luca Bardi
- Dipartimento di Scienze Biomediche Avanzate, Università Federico II, Napoli .
| | | | - Ginevra Danti
- Azienda Ospedaliero Universitaria Careggi, Firenze, Italy.
| | - Francesco Gentili
- Section of Radiology, Unit of Surgical Sciences, University of Parma, Parma, Italy.
| | - Maria Antonietta Mazzei
- Department of Medical, Surgical and Neuro Sciences, University of Siena, Department of Radiological Sciences, Unit of Diagnostic Imaging, Azienda Ospedaliera Universitaria Senese, Siena, Italy .
| | - Ernesto Di Cesare
- Department of Life, Health and Enviromental Sciences, University of L'Aquila, Italy.
| | - Alessandra Splendiani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy.
| | - Carlo Masciocchi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy.
| | - Antonio Barile
- Department of Biotechnological and Applied Clinical Science, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
7
|
Hoter A, Rizk S, Naim HY. The Multiple Roles and Therapeutic Potential of Molecular Chaperones in Prostate Cancer. Cancers (Basel) 2019; 11:cancers11081194. [PMID: 31426412 PMCID: PMC6721600 DOI: 10.3390/cancers11081194] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer (PCa) is one of the most common cancer types in men worldwide. Heat shock proteins (HSPs) are molecular chaperones that are widely implicated in the pathogenesis, diagnosis, prognosis, and treatment of many cancers. The role of HSPs in PCa is complex and their expression has been linked to the progression and aggressiveness of the tumor. Prominent chaperones, including HSP90 and HSP70, are involved in the folding and trafficking of critical cancer-related proteins. Other members of HSPs, including HSP27 and HSP60, have been considered as promising biomarkers, similar to prostate-specific membrane antigen (PSMA), for PCa screening in order to evaluate and monitor the progression or recurrence of the disease. Moreover, expression level of chaperones like clusterin has been shown to correlate directly with the prostate tumor grade. Hence, targeting HSPs in PCa has been suggested as a promising strategy for cancer therapy. In the current review, we discuss the functions as well as the role of HSPs in PCa progression and further evaluate the approach of inhibiting HSPs as a cancer treatment strategy.
Collapse
Affiliation(s)
- Abdullah Hoter
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Sandra Rizk
- School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Hassan Y Naim
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany.
| |
Collapse
|
8
|
Giannattasio S, Megiorni F, Di Nisio V, Del Fattore A, Fontanella R, Camero S, Antinozzi C, Festuccia C, Gravina GL, Cecconi S, Dominici C, Di Luigi L, Ciccarelli C, De Cesaris P, Riccioli A, Zani BM, Lenzi A, Pestell RG, Filippini A, Crescioli C, Tombolini V, Marampon F. Testosterone-mediated activation of androgenic signalling sustains in vitro the transformed and radioresistant phenotype of rhabdomyosarcoma cell lines. J Endocrinol Invest 2019; 42:183-197. [PMID: 29790086 DOI: 10.1007/s40618-018-0900-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/07/2018] [Indexed: 01/01/2023]
Abstract
PURPOSE Rhabdomyosarcoma (RMS), the most common soft-tissue sarcoma in childhood, rarely affects adults, preferring male. RMS expresses the receptor for androgen (AR) and responds to androgen; however, the molecular action of androgens on RMS is unknown. METHODS Herein, testosterone (T) effects were tested in embryonal (ERMS) and alveolar (ARMS) RMS cell lines, by performing luciferase reporter assay, RT-PCR, and western blotting experiments. RNA interference experiments or bicalutamide treatment was performed to assess the specific role of AR. Radiation treatment was delivered to characterise the effects of T treatment on RMS intrinsic radioresistance. RESULTS Our study showed that RMS cells respond to sub-physiological levels of T stimulation, finally promoting AR-dependent genomic and non-genomic effects, such as the transcriptional regulation of several oncogenes, the phosphorylation-mediated post-transductional modifications of AR and the activation of ERK, p38 and AKT signal transduction pathway mediators that, by physically complexing or not with AR, participate in regulating its transcriptional activity and the expression of T-targeted genes. T chronic daily treatment, performed as for the hormone circadian rhythm, did not significantly affect RMS cell growth, but improved RMS clonogenic and radioresistant potential and increased AR mRNA both in ERMS and ARMS. AR protein accumulation was evident in ERMS, this further developing an intrinsic T-independent AR activity. CONCLUSIONS Our results suggest that androgens sustain and improve RMS transformed and radioresistant phenotype, and therefore, their therapeutic application should be avoided in RMS post puberal patients.
Collapse
Affiliation(s)
- S Giannattasio
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - F Megiorni
- Department of Paediatrics, Sapienza University of Rome, Rome, Italy
| | - V Di Nisio
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - A Del Fattore
- Multi-Factorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - R Fontanella
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - S Camero
- Department of Paediatrics, Sapienza University of Rome, Rome, Italy
| | - C Antinozzi
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - C Festuccia
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio 1, 67100, L'Aquila, Coppito, Italy
| | - G L Gravina
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio 1, 67100, L'Aquila, Coppito, Italy
| | - S Cecconi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - C Dominici
- Department of Paediatrics, Sapienza University of Rome, Rome, Italy
| | - L Di Luigi
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - C Ciccarelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio 1, 67100, L'Aquila, Coppito, Italy
| | - P De Cesaris
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio 1, 67100, L'Aquila, Coppito, Italy
| | - A Riccioli
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - B M Zani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio 1, 67100, L'Aquila, Coppito, Italy
| | - A Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - R G Pestell
- Pennsylvania Center for Cancer and Regenerative Medicine, Wynnewood, PA, 19096, USA
| | - A Filippini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - C Crescioli
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - V Tombolini
- Department of Radiotherapy, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - F Marampon
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio 1, 67100, L'Aquila, Coppito, Italy.
- Department of Radiotherapy, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
9
|
Sun S, Zhang Y, Zheng J, Duan B, Cui J, Chen Y, Deng W, Ye B, Liu L, Chen Y, Du J, Gu L. HDAC6 inhibitor TST strengthens the antiproliferative effects of PI3K/mTOR inhibitor BEZ235 in breast cancer cells via suppressing RTK activation. Cell Death Dis 2018; 9:929. [PMID: 30206202 PMCID: PMC6134008 DOI: 10.1038/s41419-018-0931-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/30/2018] [Accepted: 07/31/2018] [Indexed: 11/23/2022]
Abstract
NVP-BEZ235 (BEZ235), an available dual PI3K/mTOR inhibitor, showed antitumor effect and provided a therapy strategy in carcinomas. However, the acquired upregulation of multiple receptor tyrosine kinases (RTKs) by NVP-BEZ235 in tumors limits its clinical efficacy. HDAC6, a class II histone deacetylase, is associated with expressions of multiple RTKs. The aim of this study was to detect whether co-treatment with HDAC6 inhibitor Tubastatin A (TST) would enhance the anticancer effects of BEZ235 in breast cancer cells. In this study, we described that treatment of breast cancer cell lines (T47D, BT474, and MDA-MB-468) with BEZ235 significantly triggered PI3K/mTOR signaling inactivation and increased multiple RTK expression, including EGFR, HER2, HER3, IGF-1 receptor, insulin receptor, and their phosphorylation levels. The adding of TST destabilized these RTKs in those breast cancer cells. Co-treatment with BEZ235 and TST reduced cell proliferative rate by strengthening Akt inactivation. In addition, the combination of these two drugs also cooperatively arrested cell cycle and DNA synthesis. In conclusion, the co-treatment with PI3K/mTOR inhibitor BEZ235 and HDAC6 inhibitor TST displayed additive antiproliferative effects on breast cancer cells through inactivating RTKs and established a rationable combination therapy to treat breast cancer.
Collapse
Affiliation(s)
- Shixiu Sun
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yujie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jianchao Zheng
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, Guangdong, 518083, China
| | - Biao Duan
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jie Cui
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yan Chen
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Wenjie Deng
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Bixing Ye
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Lei Liu
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yongchang Chen
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Jun Du
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Luo Gu
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China. .,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
10
|
Ciccarelli C, Di Rocco A, Gravina GL, Mauro A, Festuccia C, Del Fattore A, Berardinelli P, De Felice F, Musio D, Bouché M, Tombolini V, Zani BM, Marampon F. Disruption of MEK/ERK/c-Myc signaling radiosensitizes prostate cancer cells in vitro and in vivo. J Cancer Res Clin Oncol 2018; 144:1685-1699. [PMID: 29959569 DOI: 10.1007/s00432-018-2696-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 06/26/2018] [Indexed: 12/31/2022]
Abstract
PURPOSE Prostate cancer (PCa) cell radioresistance causes the failure of radiation therapy (RT) in localized or locally advanced disease. The aberrant accumulation of c-Myc oncoprotein, known to promote PCa onset and progression, may be due to the control of gene transcription and/or MEK/ERK-regulated protein stabilization. Here, we investigated the role of MEK/ERK signaling in PCa. METHODS LnCAP, 22Rv1, DU145, and PC3 PCa cell lines were used in in vitro and in vivo experiments. U0126, trametinib MEK/ERK inhibitors, and c-Myc shRNAs were used. Radiation was delivered using an x-6 MV photon linear accelerator. U0126 in vivo activity alone or in combination with irradiation was determined in murine xenografts. RESULTS Inhibition of MEK/ERK signaling down-regulated c-Myc protein in PCa cell lines to varying extents by affecting expression of RNA and protein, which in turn determined radiosensitization in in vitro and in vivo xenograft models of PCa cells. The crucial role played by c-Myc in the MEK/ERK pathways was demonstrated in 22Rv1 cells by the silencing of c-Myc by means of short hairpin mRNA, which yielded effects resembling the targeting of MEK/ERK signaling. The clinically approved compound trametinib used in vitro yielded the same effects as U0126 on growth and C-Myc expression. Notably, U0126 and trametinib induced a drastic down-regulation of BMX, which is known to prevent apoptosis in cancer cells. CONCLUSIONS The results of our study suggest that signal transduction-based therapy can, by disrupting the MEK/ERK/c-Myc axis, reduce human PCa radioresistance caused by increased c-Myc expression in vivo and in vitro and restores apoptosis signals.
Collapse
Affiliation(s)
- Carmela Ciccarelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy
| | - Agnese Di Rocco
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy
| | - Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy
| | - Annunziata Mauro
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Claudio Festuccia
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy
| | - Andrea Del Fattore
- Multi-Factorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, IRCCS, Viale di San Paolo 15, 00146, Rome, Italy
| | - Paolo Berardinelli
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Francesca De Felice
- Division of Radiotherapy, Department of Radiology, Radiation Oncology and Human Pathology, "Sapienza" University of Rome, Rome, Italy
| | - Daniela Musio
- Division of Radiotherapy, Department of Radiology, Radiation Oncology and Human Pathology, "Sapienza" University of Rome, Rome, Italy
| | - Marina Bouché
- Unit of Histology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Vincenzo Tombolini
- Unit of Histology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Bianca Maria Zani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy.
| | - Francesco Marampon
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy. .,Unit of Histology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
11
|
Marampon F, Antinozzi C, Corinaldesi C, Vannelli GB, Sarchielli E, Migliaccio S, Di Luigi L, Lenzi A, Crescioli C. The phosphodiesterase 5 inhibitor tadalafil regulates lipidic homeostasis in human skeletal muscle cell metabolism. Endocrine 2018; 59:602-613. [PMID: 28786077 DOI: 10.1007/s12020-017-1378-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 07/20/2017] [Indexed: 01/01/2023]
Abstract
PURPOSE Tadalafil seems to ameliorate insulin resistance and glucose homeostasis in humans. We have previously reported that tadalafil targets human skeletal muscle cells with an insulin (I)-like effect. We aim to evaluate in human fetal skeletal muscle cells after tadalafil or I: (i) expression profile of I-regulated genes dedicated to cellular energy control, glycolitic activity or microtubule formation/vesicle transport, as GLUT4, PPARγ, HK2, IRS-1, KIF1C, and KIFAP3; (ii) GLUT4, Flotillin-1, and Caveolin-1 localization, all proteins involved in energy-dependent cell trafficking; (iii) activation of I-targeted paths, as IRS-1, PKB/AKT, mTOR, P70/S6K. Free fatty acids intracellular level was measured. Sildenafil or a cGMP synthetic analog were used for comparison; PDE5 and PDE11 gene expression was evaluated in human fetal skeletal muscle cells. METHODS RTq-PCR, PCR, western blot, free fatty acid assay commercial kit, and lipid stain non-fluorescent assay were used. RESULTS Tadalafil upregulated I-targeted investigated genes with the same temporal pattern as I (GLUT4, PPARγ, and IRS-1 at 3 h; HK2, KIF1C, KIFAP3 at 12 h), re-localized GLUT4 in cell sites positively immune-decorated for Caveolin-1 and Flotillin-1, suggesting the involvement of lipid rafts, induced specific residue phosphorylation of IRS-1/AKT/mTOR complex in association with free fatty acid de novo synthesis. Sildenafil or GMP analog did not affect GLUT4 trafficking or free fatty acid levels. CONCLUSION In human fetal skeletal muscle cells tadalafil likely favors energy storage by modulating lipid homeostasis via IRS-1-mediated mechanisms, involving activation of I-targeted genes and intracellular cascade related to metabolic control. Those data provide some biomolecular evidences explaining, in part, tadalafil-induced favorable control of human metabolism shown by clinical studies.
Collapse
Affiliation(s)
- F Marampon
- Department of Movement, Human and Health Sciences, Università di Roma "Foro Italico", Rome, Italy
| | - C Antinozzi
- Department of Movement, Human and Health Sciences, Università di Roma "Foro Italico", Rome, Italy
| | - C Corinaldesi
- Department of Movement, Human and Health Sciences, Università di Roma "Foro Italico", Rome, Italy
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - G B Vannelli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - E Sarchielli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - S Migliaccio
- Department of Movement, Human and Health Sciences, Università di Roma "Foro Italico", Rome, Italy
| | - L Di Luigi
- Department of Movement, Human and Health Sciences, Università di Roma "Foro Italico", Rome, Italy
| | - A Lenzi
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - C Crescioli
- Department of Movement, Human and Health Sciences, Università di Roma "Foro Italico", Rome, Italy.
| |
Collapse
|
12
|
Festuccia C, Mancini A, Colapietro A, Gravina GL, Vitale F, Marampon F, Delle Monache S, Pompili S, Cristiano L, Vetuschi A, Tombolini V, Chen Y, Mehrling T. The first-in-class alkylating deacetylase inhibitor molecule tinostamustine shows antitumor effects and is synergistic with radiotherapy in preclinical models of glioblastoma. J Hematol Oncol 2018; 11:32. [PMID: 29486795 PMCID: PMC5830080 DOI: 10.1186/s13045-018-0576-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/14/2018] [Indexed: 12/14/2022] Open
Abstract
Background The use of alkylating agents such as temozolomide in association with radiotherapy (RT) is the therapeutic standard of glioblastoma (GBM). This regimen modestly prolongs overall survival, also if, in light of the still dismal prognosis, further improvements are desperately needed, especially in the patients with O6-methylguanine-DNA-methyltransferase (MGMT) unmethylated tumors, in which the benefit of standard treatment is less. Tinostamustine (EDO-S101) is a first-in-class alkylating deacetylase inhibitor (AK-DACi) molecule that fuses the DNA damaging effect of bendamustine with the fully functional pan-histone deacetylase (HDAC) inhibitor, vorinostat, in a completely new chemical entity. Methods Tinostamustine has been tested in models of GBM by using 13 GBM cell lines and seven patient-derived GBM proliferating/stem cell lines in vitro. U87MG and U251MG (MGMT negative), as well as T98G (MGMT positive), were subcutaneously injected in nude mice, whereas luciferase positive U251MG cells and patient-derived GBM stem cell line (CSCs-5) were evaluated the orthotopic intra-brain in vivo experiments. Results We demonstrated that tinostamustine possesses stronger antiproliferative and pro-apoptotic effects than those observed for vorinostat and bendamustine alone and similar to their combination and irrespective of MGMT expression. In addition, we observed a stronger radio-sensitization of single treatment and temozolomide used as control due to reduced expression and increased time of disappearance of γH2AX indicative of reduced signal and DNA repair. This was associated with higher caspase-3 activation and reduction of RT-mediated autophagy. In vivo, tinostamustine increased time-to-progression (TTP) and this was additive/synergistic to RT. Tinostamustine had significant therapeutic activity with suppression of tumor growth and prolongation of DFS (disease-free survival) and OS (overall survival) in orthotopic intra-brain models that was superior to bendamustine, RT and temozolomide and showing stronger radio sensitivity. Conclusions Our data suggest that tinostamustine deserves further investigation in patients with glioblastoma. Electronic supplementary material The online version of this article (10.1186/s13045-018-0576-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Claudio Festuccia
- Laboratory of Radiobiology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy.
| | - Andrea Mancini
- Laboratory of Radiobiology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Alessandro Colapietro
- Laboratory of Radiobiology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Giovanni Luca Gravina
- Laboratory of Radiobiology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy.,Division of Radiotherapy, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Flora Vitale
- Division of Neurosciences, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Francesco Marampon
- Division of Radiotherapy, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Simona Delle Monache
- Division of Applied Biology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Simona Pompili
- Division of Human Anatomy, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Loredana Cristiano
- Laboratory of Applied Biology, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Antonella Vetuschi
- Division of Human Anatomy, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Vincenzo Tombolini
- Division of Radiotherapy, Department of Experimental Medicine, University of Rome "La Sapienza", Rome, Italy
| | - Yi Chen
- Northlake International LLC, Pleasanton, CA, USA
| | | |
Collapse
|
13
|
Gravina GL, Marampon F, Sanità P, Festuccia C, Forcella C, Scarsella L, Jitariuc A, Vetuschi A, Sferra R, Colapietro A, Carosa E, Dolci S, Lenzi A, Jannini EA. Episode-like pulse testosterone supplementation induces tumor senescence and growth arrest down-modulating androgen receptor through modulation of p-ERK1/2, pAR ser81 and CDK1 signaling: biological implications for men treated with testosterone replacement therapy. Oncotarget 2017; 8:113792-113806. [PMID: 29371946 PMCID: PMC5768363 DOI: 10.18632/oncotarget.22776] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 11/01/2017] [Indexed: 12/21/2022] Open
Abstract
Despite the growing body of knowledge showing that testosterone (T) may not significantly affect tumor progression in hypogonadal patients treated for prostate cancer (Pca), the use of this hormone in this population still remains controversial. The effects of continuous or pulsed T stimulation were tested in vitro and in vivo on androgen-sensitive Pca cell lines in order to assess the differential biological properties of these two treatment modalities. Pulsed T treatment resulted in a greater inhibition than continuous T supplementation of tumor growth in vitro and in vivo. The effects of pulsed T treatment on tumor growth inhibition, G0/G1 cell cycle arrest, and tumor senescence was more pronounced than those obtained upon continuous T treatments. Mechanistic studies revealed that G0/G1 arrest and tumor senescence upon pulsed T treatment were associated with a marked decrease in cyclin D1, c-Myc and SKp2, CDK4 and p-Rb levels and upregulation of p27 and p-ERK1/2. Pulsed, but not continuous, T supplementation decreased the expression levels of AR, p-ARser81 and CDK1 in both cellular models. The in vitro results were confirmed in an in vivo xenografts, providing evidence of a greater inhibitory activity of pulsed supraphysiological T supplementation than continuous treatment, both in terms of tumor volume and decreased AR, p-ARser81, PSA and CDK1 staining. The rapid cycling from hypogonadal to physiological or supra-physiological T intraprostatic concentrations results in cytostatic and senescence effects in preclinical models of androgen-sensitive Pca. Our preclinical evidence provides relevant new insights in the biology of Pca response to pulsed T supplementation.
Collapse
Affiliation(s)
- Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Prostate Onco-pathology and Experimental Endocrinology, University of L'Aquila, 67100 L'Aquila, Italy
| | - Francesco Marampon
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Prostate Onco-pathology and Experimental Endocrinology, University of L'Aquila, 67100 L'Aquila, Italy
| | - Patrizia Sanità
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Prostate Onco-pathology and Experimental Endocrinology, University of L'Aquila, 67100 L'Aquila, Italy
| | - Claudio Festuccia
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Chiara Forcella
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Prostate Onco-pathology and Experimental Endocrinology, University of L'Aquila, 67100 L'Aquila, Italy
| | - Luca Scarsella
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Anna Jitariuc
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Antonella Vetuschi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Roberta Sferra
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Alessandro Colapietro
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Eleonora Carosa
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Prostate Onco-pathology and Experimental Endocrinology, University of L'Aquila, 67100 L'Aquila, Italy
| | - Susanna Dolci
- Department of Biomedicine and Prevention, Section of Anatomy, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Chair of Endocrinology, Sapienza University of Rome, 00161 Rome, Italy
| | - Emmanuele A Jannini
- Department of Systems Medicine, Chair of Endocrinology and Medical Sexology (ENDOSEX), Tor Vergata University of Rome, 00133 Rome, Italy
| |
Collapse
|
14
|
Antinozzi C, Marampon F, Corinaldesi C, Vicini E, Sgrò P, Vannelli GB, Lenzi A, Crescioli C, Di Luigi L. Testosterone insulin-like effects: an in vitro study on the short-term metabolic effects of testosterone in human skeletal muscle cells. J Endocrinol Invest 2017; 40:1133-1143. [PMID: 28508346 PMCID: PMC5610223 DOI: 10.1007/s40618-017-0686-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 05/04/2017] [Indexed: 11/01/2022]
Abstract
PURPOSE Testosterone by promoting different metabolic pathways contributes to short-term homeostasis of skeletal muscle, the largest insulin-sensitive tissue and the primary site for insulin-stimulated glucose utilization. Despite evidences indicate a close relationship between testosterone and glucose metabolism, the molecular mechanisms responsible for a possible testosterone-mediated insulin-like effects on skeletal muscle are still unknown. METHODS Here we used undifferentiated proliferating or differentiated human fetal skeletal muscle cells (Hfsmc) to investigate the short-term effects of testosterone on the insulin-mediated biomolecular metabolic machinery. GLUT4 cell expression, localization and the phosphorylation/activation of AKT, ERK, mTOR and GSK3β insulin-related pathways at different time points after treatment with testosterone were analyzed. RESULTS Independently from cells differentiation status, testosterone, with an insulin-like effect, induced Glut4-mRNA expression, GLUT4 protein translocation to the cytoplasmic membrane, while no effect was observed on GLUT4 protein expression levels. Furthermore, testosterone treatment modulated the insulin-dependent signal transduction pathways inducing a rapid and persistent activation of AKT, ERK and mTOR, and a transient inhibition of GSK3β. T-related effects were shown to be androgen receptor dependent. CONCLUSION All together our data indicate that testosterone through the activation of non-genomic pathways, participates in skeletal muscle glucose metabolism by inducing insulin-related effects.
Collapse
Affiliation(s)
- C Antinozzi
- Department of Movement, Human and Health Sciences Section of Health Sciences, Unit of Endocrinology, Università degli Studi di Roma "Foro Italico", 00135, Rome, Italy
| | - F Marampon
- Department of Movement, Human and Health Sciences Section of Health Sciences, Unit of Endocrinology, Università degli Studi di Roma "Foro Italico", 00135, Rome, Italy
| | - C Corinaldesi
- Department of Movement, Human and Health Sciences Section of Health Sciences, Unit of Endocrinology, Università degli Studi di Roma "Foro Italico", 00135, Rome, Italy
| | - E Vicini
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences-Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - P Sgrò
- Department of Movement, Human and Health Sciences Section of Health Sciences, Unit of Endocrinology, Università degli Studi di Roma "Foro Italico", 00135, Rome, Italy
| | - G B Vannelli
- Department of Experimental and Clinical Medicine, University of Florence, 50134, Florence, Italy
| | - A Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - C Crescioli
- Department of Movement, Human and Health Sciences Section of Health Sciences, Unit of Endocrinology, Università degli Studi di Roma "Foro Italico", 00135, Rome, Italy.
| | - L Di Luigi
- Department of Movement, Human and Health Sciences Section of Health Sciences, Unit of Endocrinology, Università degli Studi di Roma "Foro Italico", 00135, Rome, Italy
| |
Collapse
|
15
|
Mehndiratta S, Wang RS, Huang HL, Su CJ, Hsu CM, Wu YW, Pan SL, Liou JP. 4-Indolyl-N-hydroxyphenylacrylamides as potent HDAC class I and IIB inhibitors in vitro and in vivo. Eur J Med Chem 2017; 134:13-23. [PMID: 28395150 DOI: 10.1016/j.ejmech.2017.03.079] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/28/2017] [Accepted: 03/30/2017] [Indexed: 11/16/2022]
Abstract
A series of 4,5-indolyl-N-hydroxyphenylacrylamides, as HDAC inhibitors, has been synthesized and evaluated in vitro and in vivo. 4-Indolyl compounds 13 and 17 functions as potent inhibitors of HDAC1 (IC50 1.28 nM and 1.34 nM) and HDAC 2 (IC50 0.90 and 0.53 nM). N-Hydroxy-3-{4-[2-(1H-indol-4-yl)-ethylsulfamoyl]-phenyl}-acrylamide (13) inhibited the human cancer cell growth of PC3, A549, MDA-MB-231 and AsPC-1 with a GI50 of 0.14, 0.25, 0.32, and 0.24 μM, respectively. In in vivo evaluations bearing prostate PC3 xenografts nude mice model, compound 13 suppressed tumor growth with a tumor growth inhibition (TGI) of 62.2%. Immunohistochemistry of protein expressions, in PC-3 xenograft model indicated elevated acetyl-histone 3 and prominently inhibited HDAC2 protein expressions. Therefore, compound 13 could be a suitable lead for further investigation and the development of selective HDAC 2 inhibitors as potent anti-cancer compounds.
Collapse
Affiliation(s)
- Samir Mehndiratta
- School of Pharmacy, College of Pharmacy, Taipei Medical University (TMU), Taiwan
| | - Ruei-Shian Wang
- PhD Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, TMU, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taiwan
| | - Han-Li Huang
- PhD Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, TMU, Taiwan
| | - Chih-Jou Su
- PhD Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, TMU, Taiwan
| | - Chia-Ming Hsu
- PhD Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, TMU, Taiwan
| | - Yi-Wen Wu
- PhD Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, TMU, Taiwan
| | - Shiow-Lin Pan
- PhD Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, TMU, Taiwan; Department of Pharmacology, College of Medicine, Taipei Medical University, Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University (TMU), Taiwan.
| |
Collapse
|
16
|
Faleiro I, Leão R, Binnie A, de Mello RA, Maia AT, Castelo-Branco P. Epigenetic therapy in urologic cancers: an update on clinical trials. Oncotarget 2017; 8:12484-12500. [PMID: 28036257 PMCID: PMC5355359 DOI: 10.18632/oncotarget.14226] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 12/13/2016] [Indexed: 01/06/2023] Open
Abstract
Epigenetic dysregulation is one of many factors that contribute to cancer development and progression. Numerous epigenetic alterations have been identified in urologic cancers including histone modifications, DNA methylation changes, and microRNA expression. Since these changes are reversible, efforts are being made to develop epigenetic drugs that restore the normal epigenetic patterns of cells, and many clinical trials are already underway to test their clinical potential. In this review we analyze multiple clinical trials (n=51) that test the efficacy of these drugs in patients with urologic cancers. The most frequently used epigenetic drugs were histone deacetylase inhibitors followed by antisense oligonucleotides, DNA methyltransferase inhibitors and histone demethylase inhibitors, the last of which are only being tested in prostate cancer. In more than 50% of the clinical trials considered, epigenetic drugs were used as part of combination therapy, which achieved the best results. The epigenetic regulation of some cancers is still matter of research but will undoubtedly open a window to new therapeutic approaches in the era of personalized medicine. The future of therapy for urological malignancies is likely to include multidrug regimens in which epigenetic modifying drugs will play an important role.
Collapse
Affiliation(s)
- Inês Faleiro
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal
- Centre for Biomedical Research, University of Algarve, Faro, Portugal
- Algarve Biomedical Center, Campus Gambelas, Edificio 2. Faro, Portugal
| | - Ricardo Leão
- Department of Surgery, Princess Margaret Cancer Center, Division of Urology, University of Toronto, Toronto, Canada
- Renal Transplantation and Urology Service, Coimbra University Hospital Center EPE, Faculty of Medicine, University of Coimbra, Portugal
| | - Alexandra Binnie
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal
- Centre for Biomedical Research, University of Algarve, Faro, Portugal
- Algarve Biomedical Center, Campus Gambelas, Edificio 2. Faro, Portugal
| | - Ramon Andrade de Mello
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal
- Centre for Biomedical Research, University of Algarve, Faro, Portugal
- Algarve Biomedical Center, Campus Gambelas, Edificio 2. Faro, Portugal
| | - Ana-Teresa Maia
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal
- Centre for Biomedical Research, University of Algarve, Faro, Portugal
- Algarve Biomedical Center, Campus Gambelas, Edificio 2. Faro, Portugal
| | - Pedro Castelo-Branco
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal
- Centre for Biomedical Research, University of Algarve, Faro, Portugal
- Algarve Biomedical Center, Campus Gambelas, Edificio 2. Faro, Portugal
| |
Collapse
|
17
|
Bailey H, McPherson JP, Bailey EB, Werner TL, Gupta S, Batten J, Reddy G, Bhat G, Sharma S, Agarwal N. A phase I study to determine the pharmacokinetics and urinary excretion of belinostat and metabolites in patients with advanced solid tumors. Cancer Chemother Pharmacol 2016; 78:1059-1071. [PMID: 27744565 DOI: 10.1007/s00280-016-3167-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/06/2016] [Indexed: 11/27/2022]
Abstract
PURPOSE Belinostat is an inhibitor of histone deacetylase enzymes, resulting in DNA repair inhibition and apoptosis. Present data are lacking to provide dosing recommendations in renal insufficiency. The purpose of this trial was to assess the pharmacokinetics (PK) of belinostat and belinostat metabolites in plasma and urine. METHODS This was a phase I, single-center, open-label, two-part study. In Part I, patients received single-agent belinostat 1000 mg/m2. Blood and urine samples were collected at pre-specified time points to determine PK of belinostat and metabolites and their elimination in urine. In Part II, patients were permitted to continue belinostat in 21-day cycles on Days 1 through 5 until disease progression, unacceptable toxicity, or according to patient preference. RESULTS A total of nine patients with advanced solid tumors were treated. Median t max for belinostat was observed 10 min after the start of infusion. Concentrations of belinostat rapidly declined with a t 1/2 of 2.9 h. The mean fraction of belinostat excreted unchanged in urine was 0.926 %. The metabolites belinostat glucuronide and 3-ASBA represented the largest fractions of belinostat dose excreted in urine (30.5 and 4.61 %, respectively), while renal excretion appeared to be a minor route of elimination for the parent belinostat (<1 %). The most common adverse events were nausea, fatigue, and diarrhea. One Grade 3 adverse event (constipation) was thought to be treatment related. CONCLUSIONS Urinary elimination of parent belinostat was minimal, although a combined 36.7 % of belinostat metabolites were excreted in urine. Since these metabolites are primarily inactive, belinostat may not require dosage adjustment in renal dysfunction.
Collapse
Affiliation(s)
- Hanna Bailey
- Huntsman Cancer Institute, The University of Utah, 2000 Circle of Hope, Ste 2123, Salt Lake City, UT, 84112, USA
| | - Jordan P McPherson
- Huntsman Cancer Institute, The University of Utah, 2000 Circle of Hope, Ste 2123, Salt Lake City, UT, 84112, USA
| | - Erin B Bailey
- Huntsman Cancer Institute, The University of Utah, 2000 Circle of Hope, Ste 2123, Salt Lake City, UT, 84112, USA
| | - Theresa L Werner
- Huntsman Cancer Institute, The University of Utah, 2000 Circle of Hope, Ste 2123, Salt Lake City, UT, 84112, USA
| | - Sumati Gupta
- Huntsman Cancer Institute, The University of Utah, 2000 Circle of Hope, Ste 2123, Salt Lake City, UT, 84112, USA
| | - Julia Batten
- Huntsman Cancer Institute, The University of Utah, 2000 Circle of Hope, Ste 2123, Salt Lake City, UT, 84112, USA
| | - Guru Reddy
- Spectrum Pharmaceuticals, Irvine, CA, USA
| | | | - Sunil Sharma
- Huntsman Cancer Institute, The University of Utah, 2000 Circle of Hope, Ste 2123, Salt Lake City, UT, 84112, USA
| | - Neeraj Agarwal
- Huntsman Cancer Institute, The University of Utah, 2000 Circle of Hope, Ste 2123, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
18
|
Rosati R, Chen B, Patki M, McFall T, Ou S, Heath E, Ratnam M, Qin Z. Hybrid Enzalutamide Derivatives with Histone Deacetylase Inhibitor Activity Decrease Heat Shock Protein 90 and Androgen Receptor Levels and Inhibit Viability in Enzalutamide-Resistant C4-2 Prostate Cancer Cells. Mol Pharmacol 2016; 90:225-37. [PMID: 27382012 DOI: 10.1124/mol.116.103416] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/30/2016] [Indexed: 11/22/2022] Open
Abstract
Histone deacetylase inhibitors (HDACIs) can disrupt the viability of prostate cancer (PCa) cells through modulation of the cytosolic androgen receptor (AR) chaperone protein heat shock protein 90 (HSP90). However, toxicities associated with their pleiotropic effects could contribute to the ineffectiveness of HDACIs in PCa treatment. We designed hybrid molecules containing partial chemical scaffolds of enzalutamide and suberoylanilide hydroxamic acid (SAHA), with weakened intrinsic pan-HDACI activities, to target HSP90 and AR in enzalutamide-resistant PCa cells. The potency of the new molecules, compounds 2-75 [4-(3-(4-cyano-3-(trifluoromethyl)phenyl)-5,5-dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)-2-fluoro-N-(7-(hydroxyamino)-7-oxoheptyl)benzamide] and 1005 [(E)-3-(4-(3-(4-cyano-3-(trifluoromethyl)phenyl)-5,5-dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)-2-fluorophenyl)-N-hydroxyacrylamide], as inhibitors of nuclear and cytosolic histone deacetylases was substantially lower than that of SAHA in cell-free and in situ assays. Compounds 2-75 and 1005 antagonized gene activation by androgen without inducing chromatin association of AR. Enzalutamide had no effect on the levels of AR or HSP90, whereas the hybrid compounds induced degradation of both AR and HSP90, similar to (compound 1005) or more potently than (compound 2-75) SAHA. Similar to SAHA, compounds 2-75 and 1005 decreased the level of HSP90 and induced acetylation in a predicted approximately 55 kDa HSP90 fragment. Compared with SAHA, compound 2-75 induced greater hyperacetylation of the HDAC6 substrate α-tubulin. In contrast with SAHA, neither hybrid molecule caused substantial hyperacetylation of histones H3 and H4. Compounds 2-75 and 1005 induced p21 and caused loss of viability in the enzalutamide-resistant C4-2 cells, with efficacies that were comparable to or better than SAHA. The results suggest the potential of the new compounds as prototype antitumor drugs that would downregulate HSP90 and AR in enzalutamide-resistant PCa cells with weakened effects on nuclear HDACI targets.
Collapse
Affiliation(s)
- Rayna Rosati
- Barbara Ann Karmanos Cancer Institute, Departments of Oncology (R.R., M.P., T.M., E.H., M.R.) and Pharmaceutical Sciences (B.C., S.O., Z.Q.), Wayne State University, Detroit, Michigan
| | - Bailing Chen
- Barbara Ann Karmanos Cancer Institute, Departments of Oncology (R.R., M.P., T.M., E.H., M.R.) and Pharmaceutical Sciences (B.C., S.O., Z.Q.), Wayne State University, Detroit, Michigan
| | - Mugdha Patki
- Barbara Ann Karmanos Cancer Institute, Departments of Oncology (R.R., M.P., T.M., E.H., M.R.) and Pharmaceutical Sciences (B.C., S.O., Z.Q.), Wayne State University, Detroit, Michigan
| | - Thomas McFall
- Barbara Ann Karmanos Cancer Institute, Departments of Oncology (R.R., M.P., T.M., E.H., M.R.) and Pharmaceutical Sciences (B.C., S.O., Z.Q.), Wayne State University, Detroit, Michigan
| | - Siyu Ou
- Barbara Ann Karmanos Cancer Institute, Departments of Oncology (R.R., M.P., T.M., E.H., M.R.) and Pharmaceutical Sciences (B.C., S.O., Z.Q.), Wayne State University, Detroit, Michigan
| | - Elisabeth Heath
- Barbara Ann Karmanos Cancer Institute, Departments of Oncology (R.R., M.P., T.M., E.H., M.R.) and Pharmaceutical Sciences (B.C., S.O., Z.Q.), Wayne State University, Detroit, Michigan
| | - Manohar Ratnam
- Barbara Ann Karmanos Cancer Institute, Departments of Oncology (R.R., M.P., T.M., E.H., M.R.) and Pharmaceutical Sciences (B.C., S.O., Z.Q.), Wayne State University, Detroit, Michigan
| | - Zhihui Qin
- Barbara Ann Karmanos Cancer Institute, Departments of Oncology (R.R., M.P., T.M., E.H., M.R.) and Pharmaceutical Sciences (B.C., S.O., Z.Q.), Wayne State University, Detroit, Michigan
| |
Collapse
|
19
|
CUDC-101, a Novel Inhibitor of Full-Length Androgen Receptor (flAR) and Androgen Receptor Variant 7 (AR-V7) Activity: Mechanism of Action and In Vivo Efficacy. Discov Oncol 2016; 7:196-210. [PMID: 26957440 DOI: 10.1007/s12672-016-0257-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/12/2016] [Indexed: 12/29/2022] Open
Abstract
Castration-resistant prostate cancer (CRPC) is an androgen receptor (AR)-dependent disease expected to cause the death of more than 27,000 Americans in 2015. There are only a few available treatments for CRPC, making the discovery of new drugs an urgent need. We report that CUDC-101 (an inhibitor od HER2/NEU, EGFR and HDAC) inhibits both the full length AR (flAR) and the AR variant AR-V7. This observation prompted experiments to discover which of the known activities of CUDC-101 is responsible for the inhibition of flAR/AR-V7 signaling. We used pharmacologic and genetic approaches, and found that the effect of CUDC-101 on flAR and AR-V7 was duplicated only by other HDAC inhibitors, or by silencing the HDAC isoforms HDAC5 and HDAC10. We observed that CUDC-101 treatment or AR-V7 silencing by RNAi equally reduced transcription of the AR-V7 target gene, PSA, without affecting viability of 22Rv1 cells. However, when cellular proliferation was used as an end point, CUDC-101 was more effective than AR-V7 silencing, raising the prospect that CUDC-101 has additional targets beside AR-V7. In support of this, we found that CUDC-101 increased the expression of the cyclin-dependent kinase inhibitor p21, and decreased that of the oncogene HER2/NEU. To determine if CUDC-101 reduces growth in a xenograft model of prostate cancer, this drug was given for 14 days to castrated male SCID mice inoculated with 22Rv1 cells. Compared to vehicle, CUDC-101 reduced xenograft growth in a statistically significant way, and without macroscopic side effects. These studies demonstrate that CUDC-101 inhibits wtAR and AR-V7 activity and growth of 22Rv1 cells in vitro and in vivo. These effects result from the ability of CUDC-101 to target not only HDAC signaling, which was associated with decreased flAR and AR-V7 activity, but multiple additional oncogenic pathways. These observations raise the possibility that treatment of CRPC may be achieved by using similarly multi-targeted approaches.
Collapse
|
20
|
Agarwal N, McPherson JP, Bailey H, Gupta S, Werner TL, Reddy G, Bhat G, Bailey EB, Sharma S. A phase I clinical trial of the effect of belinostat on the pharmacokinetics and pharmacodynamics of warfarin. Cancer Chemother Pharmacol 2015; 77:299-308. [PMID: 26719074 DOI: 10.1007/s00280-015-2934-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 11/29/2015] [Indexed: 11/30/2022]
Abstract
PURPOSE Belinostat is a potent small molecule inhibitor that exerts its antitumor effect through inhibition of histone deacetylase. The purpose of this study was to evaluate the pharmacokinetics and pharmacodynamics of warfarin (as a reference drug metabolized by CYP2C9) in the presence and absence of belinostat. METHODS We conducted a phase I, single-center, open-label, drug-drug interaction study between belinostat and warfarin. In part I, patients were given warfarin 5 mg orally (day-14 and 3) and belinostat 1000 mg/m(2) (days 1 through 5). Patients receiving benefit continued belinostat on days 1 through 5 every 21 days until disease progression, unacceptable toxicity, or per patient preference. RESULTS A total of 18 patients were treated. With belinostat, the least-squared means for maximum concentration (C max), area under the curve0-∞, and area under the curve0-t of R-warfarin were slightly increased. However, for the more potent S-warfarin isomer, the same parameters were primarily contained within the pre-specified equivalence limits of 0.80 and 1.25, indicating there was no statistically significant interaction between S-warfarin and belinostat. The most common adverse events were nausea, vomiting, and fatigue. Three grade 3 adverse events (diarrhea 5.6 %, nausea 5.6 %, and vomiting 5.6 %) were thought to be treatment related. Progression-free survival ranged from 0.2 to 13.8 months in all patients. CONCLUSIONS Belinostat did not significantly affect the pharmacokinetics and pharmacodynamics of warfarin, indicating no clinically relevant effect on the enzymatic activity of CYP2C9.
Collapse
Affiliation(s)
- Neeraj Agarwal
- Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, 84112, UT, USA.
| | - Jordan P McPherson
- Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, 84112, UT, USA
| | - Hanna Bailey
- Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, 84112, UT, USA
| | - Sumati Gupta
- Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, 84112, UT, USA
| | - Theresa L Werner
- Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, 84112, UT, USA
| | - Guru Reddy
- Spectrum Pharmaceuticals, Irvine, CA, USA
| | | | - Erin B Bailey
- Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, 84112, UT, USA
| | - Sunil Sharma
- Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, 84112, UT, USA
| |
Collapse
|
21
|
4-Hydroxybenzoic acid derivatives as HDAC6-specific inhibitors modulating microtubular structure and HSP90α chaperone activity against prostate cancer. Biochem Pharmacol 2015; 99:31-52. [PMID: 26549368 DOI: 10.1016/j.bcp.2015.11.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 11/03/2015] [Indexed: 01/06/2023]
Abstract
Histone deacetylase (HDAC)6 is a unique isoenzyme targeting specific substrates including α-tubulin and heat shock protein (HSP)90. HDAC6 is involved in protein trafficking and degradation, cell shape and migration. Deregulation of HDAC6 activity is associated with a variety of diseases including cancer leading to a growing interest for developing HDAC6 inhibitors. Here, we identified two new structurally related 4-hydroxybenzoic acids as selective HDAC6 inhibitors reducing proliferation, colony and spheroid formation as well as viability of prostate cancer cells. Both compounds strongly enhanced α-tubulin acetylation leading to remodeling of microtubular organization. Furthermore, 4-hydroxybenzoic acids decreased HSP90α regulation of the human androgen receptor in prostate cancer cells by increasing HSP90α acetylation levels. Collectively, our data support the potential of 4-hydroxybenzoic acid derivatives as HDAC6-specific inhibitors with anti-cancer properties.
Collapse
|
22
|
Chou YW, Lin FF, Muniyan S, Lin FC, Chen CS, Wang J, Huang CC, Lin MF. Cellular prostatic acid phosphatase (cPAcP) serves as a useful biomarker of histone deacetylase (HDAC) inhibitors in prostate cancer cell growth suppression. Cell Biosci 2015; 5:38. [PMID: 26185616 PMCID: PMC4504398 DOI: 10.1186/s13578-015-0033-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 07/06/2015] [Indexed: 01/17/2023] Open
Abstract
Background Prostate cancer (PCa) is the most commonly diagnosed solid tumor and the second leading cancer death in the United States, and also one of the major cancer-related deaths in Chinese. Androgen deprivation therapy (ADT) is the first line treatment for metastatic PCa. PCa ultimately relapses with subsequent ADT treatment failure and becomes castrate-resistant (CR). It is important to develop effective therapies with a surrogate marker towards CR PCa. Method Histone deacetylase (HDAC) inhibitors were examined to determine their effects in androgen receptor (AR)/cellular prostatic acid phosphatase (cPAcP)-positive PCa cells, including LNCaP C-33, C-81, C4-2 and C4-2B and MDA PCa2b androgen-sensitive and androgen-independent cells, and AR/cPAcP-negative PCa cells, including PC-3 and DU 145 cells. Cell growth was determined by cell number counting. Western blot analyses were carried out to determine AR, cPAcP and PSA protein levels. Results cPAcP protein level was increased by HDAC inhibitor treatment. Valproic acid, a HDAC inhibitor, suppressed the growth of AR/cPAcP-positive PCa cells by over 50% in steroid-reduced conditions, higher than on AR/cPAcP-negative PCa cells. Further, HDAC inhibitor pretreatments increased androgen responsiveness as demonstrated by PSA protein level quantitation. Conclusion Our results clearly demonstrate that HDAC inhibitors can induce cPAcP protein level, increase androgen responsiveness, and exhibit higher inhibitory activities on AR/cPAcP-positive PCa cells than on AR/cPAcP-negative PCa cells. Upon HDAC inhibitor pretreatment, PSA level was greatly elevated by androgens. This data indicates the potential clinical importance of cPAcP serving as a useful biomarker in the identification of PCa patient sub-population suitable for HDAC inhibitor treatment.
Collapse
Affiliation(s)
- Yu-Wei Chou
- Tissue Bank and BioBank, Kaohsiung Chang Gung Memorial Hospital, No. 123, Da-Pi Road, Niao-Song District, Kaohsiung, 833 Taiwan, ROC ; Department of Biochemistry and Molecular Biology, College of Medicine, 985870 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198-5870 USA
| | - Fen-Fen Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, 985870 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198-5870 USA
| | - Sakthivel Muniyan
- Department of Biochemistry and Molecular Biology, College of Medicine, 985870 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198-5870 USA
| | - Frank C Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, 985870 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198-5870 USA ; Division of Urology, Department of Surgery, University of Arizona Medical Center, Tucson, AZ USA
| | - Ching-Shih Chen
- Division of Medicinal Chemistry and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH USA
| | - Jue Wang
- Division of Oncology/Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE USA ; University of Arizona Cancer Center, St. Joseph's Hospital and Medical Center, Phoenix, AZ USA
| | - Chao-Cheng Huang
- Tissue Bank and BioBank, Kaohsiung Chang Gung Memorial Hospital, No. 123, Da-Pi Road, Niao-Song District, Kaohsiung, 833 Taiwan, ROC ; Department of Pathology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | - Ming-Fong Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, 985870 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198-5870 USA ; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE USA ; Department of Surgery/Urology, University of Nebraska Medical Center, Omaha, NE USA ; School of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| |
Collapse
|
23
|
Abstract
The androgen receptor (AR), ligand-induced transcription factor, is expressed in primary prostate cancer and in metastases. AR regulates multiple cellular events, proliferation, apoptosis, migration, invasion, and differentiation. Its expression in prostate cancer cells is regulated by steroid and peptide hormones. AR downregulation by various compounds which are contained in fruits and vegetables is considered a chemopreventive strategy for prostate cancer. There is a bidirectional interaction between the AR and micro-RNA (miRNA) in prostate cancer; androgens may upregulate or downregulate the selected miRNA, whereas the AR itself is a target of miRNA. AR mutations have been discovered in prostate cancer, and their incidence may increase with tumor progression. AR mutations and increased expression of selected coactivators contribute to the acquisition of agonistic properties of anti-androgens. Expression of some of the coactivators is enhanced during androgen ablation. AR activity is regulated by peptides such as cytokines or growth factors which reduce the concentration of androgen required for maximal stimulation of the receptor. In prostate cancer, variant ARs which exhibit constitutive activity were detected. Novel therapies which interfere with intracrine synthesis of androgens or inhibit nuclear translocation of the AR have been introduced in the clinic.
Collapse
Affiliation(s)
- Zoran Culig
- Division of Experimental Urology, Department of Urology, Innsbruck Medical University, Anichstrasse 35, 6020, Innsbruck, Austria,
| | | |
Collapse
|
24
|
Gravina GL, Senapedis W, McCauley D, Baloglu E, Shacham S, Festuccia C. Nucleo-cytoplasmic transport as a therapeutic target of cancer. J Hematol Oncol 2014; 7:85. [PMID: 25476752 PMCID: PMC4272779 DOI: 10.1186/s13045-014-0085-1] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 11/09/2014] [Indexed: 12/19/2022] Open
Abstract
Shuttling of specific proteins out of the nucleus is essential for the regulation of the cell cycle and proliferation of both normal and malignant tissues. Dysregulation of this fundamental process may affect many other important cellular processes such as tumor growth, inflammatory response, cell cycle, and apoptosis. It is known that XPO1 (Exportin-1/Chromosome Region Maintenance 1/CRM1) is the main mediator of nuclear export in many cell types. Nuclear proteins exported to the cytoplasm by XPO1 include the drug targets topoisomerase IIα (topo IIα) and BCR-ABL and tumor suppressor proteins such as Rb, APC, p53, p21, and p27. XPO1 can mediate cell proliferation through several pathways: (i) the sub-cellular localization of NES-containing oncogenes and tumor suppressor proteins, (ii) the control of the mitotic apparatus and chromosome segregation, and (iii) the maintenance of nuclear and chromosomal structures. The XPO1 protein is elevated in ovarian carcinoma, glioma, osteosarcoma, pancreatic and cervical cancer. There is a growing body of research indicating that XPO1 may have an important role as a prognostic marker in solid tumors. Because of this, nuclear export inhibition through XPO1 is a potential target for therapeutic intervention in many cancers. The best understood XPO1 inhibitors are the small molecule nuclear export inhibitors (NEIs; Leptomycin B and derivatives, ratjadones, PKF050-638, valtrate, ACA, CBS9106, selinexor/KPT-330, and verdinexor/KPT-335). Selinexor and verdinexor are orally bioavailable, highly potent, small molecules that are classified as Selective Inhibitors of Nuclear Export (SINE). KPT-330 is the only NEI currently in Phase I/II human clinical trials in hematological and solid cancers. Of all the potential targets in nuclear cytoplasmic transport, the nuclear export receptor XPO1 remains the best understood and most advanced therapeutic target for the treatment of cancer.
Collapse
Affiliation(s)
- Giovanni Luca Gravina
- />Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | | | - Dilara McCauley
- />Karyopharm Therapeutics, Inc., 85 Wells Avenue, Newton, MA USA
| | - Erkan Baloglu
- />Karyopharm Therapeutics, Inc., 85 Wells Avenue, Newton, MA USA
| | - Sharon Shacham
- />Karyopharm Therapeutics, Inc., 85 Wells Avenue, Newton, MA USA
| | - Claudio Festuccia
- />Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
25
|
Zhang M, Wu CH, Zhu XL, Wang YJ. Loss of Imprinting of Insulin-Like Growth Factor 2 is Associated with Increased Risk of Primary Lung Cancer in the Central China Region. Asian Pac J Cancer Prev 2014; 15:7799-803. [DOI: 10.7314/apjcp.2014.15.18.7799] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
26
|
Gravina GL, Tortoreto M, Mancini A, Addis A, Di Cesare E, Lenzi A, Landesman Y, McCauley D, Kauffman M, Shacham S, Zaffaroni N, Festuccia C. XPO1/CRM1-selective inhibitors of nuclear export (SINE) reduce tumor spreading and improve overall survival in preclinical models of prostate cancer (PCa). J Hematol Oncol 2014; 7:46. [PMID: 25284315 PMCID: PMC4283114 DOI: 10.1186/1756-8722-7-46] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 06/19/2014] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Exportin 1 (XPO1), also called chromosome region maintenance 1 (CRM1), is the sole exportin mediating transport of many multiple tumor suppressor proteins out of the nucleus. AIM AND METHODS To verify the hypothesis that XPO1 inhibition affects prostate cancer (PCa) metastatic potential, orally available, potent and selective, SINE compounds, Selinexor (KPT- 330) and KPT-251, were tested in preclinical models known to generate bone lesions and systemic tumor spread. RESULTS In vitro, Selinexor reduced both secretion of proteases and ability to migrate and invade of PCa cells. SINEs impaired secretion of pro-angiogenic and pro-osteolytic cytokines and reduced osteoclastogenesis in RAW264.7 cells. In the intra-prostatic growth model, Selinexor reduced DU145 tumor growth by 41% and 61% at the doses of 4 mg/Kg qd/5 days and 10 mg/Kg q2dx3 weeks, respectively, as well as the incidence of macroscopic visceral metastases. In a systemic metastasis model, following intracardiac injection of PCb2 cells, 80% (8/10) of controls, 10% (1/10) Selinexor- and 20% (2/10) KPT-251-treated animals developed radiographic evidence of lytic bone lesions. Similarly, after intra-tibial injection, the lytic areas were higher in controls than in Selinexor and KPT-251 groups. Analogously, the serum levels of osteoclast markers (mTRAP and type I collagen fragment, CTX), were significantly higher in controls than in Selinexor- and KPT-251-treated animals. Importantly, overall survival and disease-free survival were significantly higher in Selinexor- and KPT-251-treated animals when compared to controls. CONCLUSIONS Selective blockade of XPO1-dependent nuclear export represents a completely novel approach for the treatment of advanced and metastatic PCa.
Collapse
Affiliation(s)
- Giovanni Luca Gravina
- />Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L’Aquila, L’Aquila, Italy
- />Department of Experimental Medicine, Pathophysiology Section, Sapienza University of Rome, Rome, Italy
| | - Monica Tortoreto
- />Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Andrea Mancini
- />Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L’Aquila, L’Aquila, Italy
| | - Alessandro Addis
- />Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Ernesto Di Cesare
- />Department of Biotechnological and Applied Clinical Sciences, Division of Radiotherapy, University of L’Aquila, L’Aquila, Italy
| | - Andrea Lenzi
- />Department of Experimental Medicine, Pathophysiology Section, Sapienza University of Rome, Rome, Italy
| | | | | | | | | | - Nadia Zaffaroni
- />Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Claudio Festuccia
- />Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
27
|
Advances in prostate cancer research and treatment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:708383. [PMID: 25215290 PMCID: PMC4151599 DOI: 10.1155/2014/708383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 07/23/2014] [Indexed: 11/17/2022]
|
28
|
|
29
|
Gravina GL, Marampon F, Sherris D, Vittorini F, Di Cesare E, Tombolini V, Lenzi A, Jannini EA, Festuccia C. Torc1/Torc2 inhibitor, Palomid 529, enhances radiation response modulating CRM1-mediated survivin function and delaying DNA repair in prostate cancer models. Prostate 2014; 74:852-68. [PMID: 24715588 DOI: 10.1002/pros.22804] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 02/26/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND P529, a Torc1/Torc2 inhibitor, has demonstrated its potential as a radiosensitizer. However the molecular mechanisms underlying this phenomenon still need to be elucidated. Aim of this study is to dissect molecular mechanisms regulating the radiosensitizing properties of P529 in a wide panel of prostate cancer models. METHODS Six tumor cell lines and xenograft models were used for in vitro and in vivo studies. Clonogenic survival, apoptotic, autophagic, and senescence assays were used to examine the effects of ionizing radiation (IR) alone and in combination with P529. CRM1, survivin, GSK-3β, and DNA-DSBs expression and modulation, upon P529 and RT, were monitored by western blot. In vivo treatment response upon P529, irradiation or combination of P529 with IR was monitored by tumor volume, time to progression (TTP), and immunohistochemical analysis. RESULTS P529 treatment induced significantly more apoptosis and DNA double-strand break (DSB) when combined with radiotherapy resulting in cellular radiosensitization and growth delay of irradiated tumor xenografts. Upon P529 treatment Rad51, DNA-PKcs, and Ku70 protein expression was downregulated, indicating delayed DNA double-strand damage repair. The radiosensitizing properties of P529 were partially linked to GSK-3β, cyclin-D1, and c-myc modulation with associated inhibition of CRM1-mediated nuclear export of survivin. Importantly, autophagy and tumor senescence were involved in the enhanced P529 radioresponse. CONCLUSIONS Impaired DNA double-strand damage repair, inhibition of CRM1-mediated nuclear export of survivin, modulation of cyclin-D1 and c-myc with associated pro-apoptotic and autophagic and senescent events explain the radiosensitizing properties of P529 in preclinical models of prostate cancer.
Collapse
Affiliation(s)
- Giovanni Luca Gravina
- Division of Radiation Oncology, Department of Clinical and Applied Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy; Department of Clinical and Applied Sciences and Biotechnologies, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy; Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy; Department of Clinical and Applied Sciences and Biotechnologies, School of Sexology, University of L'Aquila, L'Aquila, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|