1
|
Sestito S, Cirone I, Sagona S, Runfola M, Raffellini L, La Rocca V, Citi V, Martelli A, Daniele S, Lai M, Calderone V, Martini C, Manera C, Rapposelli S. Design, synthesis and biological evaluation of new H2S-releasing rivastigmine derivatives as neuroprotective molecules. Eur J Med Chem 2025; 283:117175. [PMID: 39705734 DOI: 10.1016/j.ejmech.2024.117175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/11/2024] [Accepted: 12/14/2024] [Indexed: 12/22/2024]
Abstract
Alzheimer's disease (AD) represents one of the main challenges for the 21st century medical research as no disease-modifying agent has been successfully progressed to the market, while the number of people affected by AD is estimated to grow exponentially over the next years. The complex network of triggering factors involved in the insurgence and progression of AD can be rightly addressed as one of the main reasons behind the difficulty in identifying new pharmacological approaches. For this reason, the discovery and development of drugs endowed with pleiotropic activity remain the most valuable, but at the same time challenging, approaches to tackle down AD. Interestingly, the combination of active pharmacophores through molecular hybridization - or Multi-Target Directed Ligand strategy (MTDL) - has not been explored enough for this disease, despite proving to be a successfully strategy in other field, such as oncology. To contribute to the development of new strategies against AD, we decided to explore the hybridization of the marketed drug rivastigmine - prescribed to ameliorate AD symptomatology - with moieties capable to release hydrogen sulfide (H2S), a gasotransmitter with a key role in the neurological physiology of ageing. In particular, we identified compound 1, as a potent small molecule capable of inhibit AChE, preventing inflammation and ROS production in cultured neurons and microglia, triggering autophagy response and blocking Aβ fibrils propagation. Interestingly, the beneficial effects observed in vitro have been confirmed in vivo, since the rivastigmine derivative 1 improved the lifespan in a Caenorhabditis elegans model of AD.
Collapse
Affiliation(s)
- Simona Sestito
- Department of Chemistry and Pharmacy, University of Sassari, via Muroni 23/a, 07100, Sassari, Italy
| | - Italo Cirone
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126, Pisa, Italy
| | - Simona Sagona
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126, Pisa, Italy
| | | | - Lorenzo Raffellini
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126, Pisa, Italy
| | - Veronica La Rocca
- Retrovirus Center, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Valentina Citi
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126, Pisa, Italy
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126, Pisa, Italy; Centre for Instrumentation Sharing, University of Pisa (CISUP), Italy
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126, Pisa, Italy
| | - Michele Lai
- Retrovirus Center, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy; Centre for Instrumentation Sharing, University of Pisa (CISUP), Italy
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126, Pisa, Italy; Centre for Instrumentation Sharing, University of Pisa (CISUP), Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126, Pisa, Italy
| | - Clementina Manera
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126, Pisa, Italy; Centre for Instrumentation Sharing, University of Pisa (CISUP), Italy
| | - Simona Rapposelli
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126, Pisa, Italy; Centre for Instrumentation Sharing, University of Pisa (CISUP), Italy.
| |
Collapse
|
2
|
Lawler J. Counter Regulation of Tumor Angiogenesis by Vascular Endothelial Growth Factor and Thrombospondin-1. Semin Cancer Biol 2022; 86:126-135. [PMID: 36191900 DOI: 10.1016/j.semcancer.2022.09.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 10/31/2022]
Abstract
Considerable progress has been made in our understanding of the process of angiogenesis in the context of normal and tumor tissue over the last fifty years. Angiogenesis, like most physiological processes, is carefully controlled by dynamic and opposing effects of positive factors, such as vascular endothelial growth factor (VEGF), and negative factors, such as thrombospondin-1. In most cases, the progression of a small mass of cancerous cells to a life-threatening tumor depends upon the initiation of angiogenesis and involves the dysregulation of the angiogenic balance. Whereas our newfound appreciation for the role of angiogenesis in cancer has opened up new avenues for treatment, the success of these treatments, which have focused almost exclusively on antagonizing the VEGF pathway, has been limited to date. It is anticipated that this situation will improve as more therapeutics that target other pathways are developed, more strategies for combination therapies are advanced, more detailed stratification of patient populations occurs, and a better understanding of resistance to anti-angiogenic therapy is gained.
Collapse
Affiliation(s)
- Jack Lawler
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, The Center for Vascular Biology Research, 99 Brookline Ave, Boston MA 02215, United States.
| |
Collapse
|
3
|
Song ZL, Zhao L, Ma T, Osama A, Shen T, He Y, Fang J. Progress and perspective on hydrogen sulfide donors and their biomedical applications. Med Res Rev 2022; 42:1930-1977. [PMID: 35657029 DOI: 10.1002/med.21913] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/18/2022] [Accepted: 05/23/2022] [Indexed: 12/22/2022]
Abstract
Following the discovery of nitric oxide (NO) and carbon monoxide (CO), hydrogen sulfide (H2 S) has been identified as the third gasotransmitter in humans. Increasing evidence have shown that H2 S is of preventive or therapeutic effects on diverse pathological complications. As a consequence, it is of great significance to develop suitable approaches of H2 S-based therapeutics for biomedical applications. H2 S-releasing agents (H2 S donors) play important roles in exploring and understanding the physiological functions of H2 S. More importantly, accumulating studies have validated the theranostic potential of H2 S donors in extensive repertoires of in vitro and in vivo disease models. Thus, it is imperative to summarize and update the literatures in this field. In this review, first, the background of H2 S on its chemical and biological aspects is concisely introduced. Second, the studies regarding the H2 S-releasing compounds are categorized and described, and accordingly, their H2 S-donating mechanisms, biological applications, and therapeutic values are also comprehensively delineated and discussed. Necessary comparisons between related H2 S donors are presented, and the drawbacks of many typical H2 S donors are analyzed and revealed. Finally, several critical challenges encountered in the development of multifunctional H2 S donors are discussed, and the direction of their future development as well as their biomedical applications is proposed. We expect that this review will reach extensive audiences across multiple disciplines and promote the innovation of H2 S biomedicine.
Collapse
Affiliation(s)
- Zi-Long Song
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China.,Botanical Agrochemicals Research & Development Center, Lanzhou Jiaotong University, Lanzhou, Gansu, China
| | - Lanning Zhao
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China.,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Tao Ma
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Alsiddig Osama
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Tong Shen
- Botanical Agrochemicals Research & Development Center, Lanzhou Jiaotong University, Lanzhou, Gansu, China
| | - Yilin He
- Botanical Agrochemicals Research & Development Center, Lanzhou Jiaotong University, Lanzhou, Gansu, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China.,School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, China
| |
Collapse
|
4
|
The Role of Hydrogen Sulfide in Respiratory Diseases. Biomolecules 2021; 11:biom11050682. [PMID: 34062820 PMCID: PMC8147381 DOI: 10.3390/biom11050682] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 02/08/2023] Open
Abstract
Respiratory diseases are leading causes of death and disability around the globe, with a diverse range of health problems. Treatment of respiratory diseases and infections has been verified to be thought-provoking because of the increasing incidence and mortality rate. Hydrogen sulfide (H2S) is one of the recognized gaseous transmitters involved in an extensive range of cellular functions, and physiological and pathological processes in a variety of diseases, including respiratory diseases. Recently, the therapeutic potential of H2S for respiratory diseases has been widely investigated. H2S plays a vital therapeutic role in obstructive respiratory disease, pulmonary fibrosis, emphysema, pancreatic inflammatory/respiratory lung injury, pulmonary inflammation, bronchial asthma and bronchiectasis. Although the therapeutic role of H2S has been extensively studied in various respiratory diseases, a concrete literature review will have an extraordinary impact on future therapeutics. This review provides a comprehensive overview of the effective role of H2S in respiratory diseases. Besides, we also summarized H2S production in the lung and its metabolism processes in respiratory diseases.
Collapse
|
5
|
Zhang P, Yu Y, Wang P, Shen H, Ling X, Xue X, Yang Q, Zhang Y, Xiao J, Wang Z. Role of Hydrogen Sulfide in Myocardial Ischemia-Reperfusion Injury. J Cardiovasc Pharmacol 2021; 77:130-141. [PMID: 33165141 DOI: 10.1097/fjc.0000000000000943] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 10/13/2020] [Indexed: 12/28/2022]
Abstract
ABSTRACT Hydrogen sulfide (H2S), generally known as a new gas signal molecule after nitric oxide and carbon monoxide, has been found as an important endogenous gasotransmitter in the last few decades, and it plays a significant role in the cardiovascular system both pathologically and physiologically. In recent years, there is growing evidence that H2S provides myocardial protection against myocardial ischemia-reperfusion injury (MIRI), which resulted in an ongoing focus on the possible mechanisms of action accounting for the H2S cardioprotective effect. At present, lots of mechanisms of action have been verified through in vitro and in vivo models of I/R injury, such as S-sulfhydrated modification, antiapoptosis, effects on microRNA, bidirectional effect on autophagy, antioxidant stress, or interaction with NO and CO. With advances in understanding of the molecular pathogenesis of MIRI and pharmacology studies, the design, the development, and the pharmacological characterization of H2S donor drugs have made great important progress. This review summarizes the latest research progress on the role of H2S in MIRI, systematically explains the molecular mechanism of H2S affecting MIRI, and provides a new idea for the formulation of a myocardial protection strategy in the future.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Yue Yu
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Pei Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Hua Shen
- Department of Cardiovascular Surgery, Chinese PLA General Hospital, Beijing, China
| | - Xinyu Ling
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Xiaofei Xue
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Qian Yang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Yufeng Zhang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Jian Xiao
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Zhinong Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| |
Collapse
|
6
|
Yu L, Wang M, Yang Y, Xu F, Zhang X, Xie F, Gao L, Li X. Predicting therapeutic drugs for hepatocellular carcinoma based on tissue-specific pathways. PLoS Comput Biol 2021; 17:e1008696. [PMID: 33561121 PMCID: PMC7920387 DOI: 10.1371/journal.pcbi.1008696] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 03/01/2021] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a significant health problem worldwide with poor prognosis. Drug repositioning represents a profitable strategy to accelerate drug discovery in the treatment of HCC. In this study, we developed a new approach for predicting therapeutic drugs for HCC based on tissue-specific pathways and identified three newly predicted drugs that are likely to be therapeutic drugs for the treatment of HCC. We validated these predicted drugs by analyzing their overlapping drug indications reported in PubMed literature. By using the cancer cell line data in the database, we constructed a Connectivity Map (CMap) profile similarity analysis and KEGG enrichment analysis on their related genes. By experimental validation, we found securinine and ajmaline significantly inhibited cell viability of HCC cells and induced apoptosis. Among them, securinine has lower toxicity to normal liver cell line, which is worthy of further research. Our results suggested that the proposed approach was effective and accurate for discovering novel therapeutic options for HCC. This method also could be used to indicate unmarked drug-disease associations in the Comparative Toxicogenomics Database. Meanwhile, our method could also be applied to predict the potential drugs for other types of tumors by changing the database.
Collapse
Affiliation(s)
- Liang Yu
- School of Computer Science and Technology, Xidian University, Shaanxi, China
| | - Meng Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, 72, Jimo District, Qingdao, Shandong, China
| | - Yang Yang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, 72, Jimo District, Qingdao, Shandong, China
| | - Fengdan Xu
- School of Computer Science and Technology, Xidian University, Shaanxi, China
| | - Xu Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, 72, Jimo District, Qingdao, Shandong, China
| | - Fei Xie
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, 72, Jimo District, Qingdao, Shandong, China
| | - Lin Gao
- School of Computer Science and Technology, Xidian University, Shaanxi, China
| | - Xiangzhi Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Advanced Medical Research Institute, Shandong University, 72, Jimo District, Qingdao, Shandong, China
| |
Collapse
|
7
|
Avallone A, Piccirillo MC, Di Gennaro E, Romano C, Calabrese F, Roca MS, Tatangelo F, Granata V, Cassata A, Cavalcanti E, Maurea N, Maiolino P, Silvestro L, De Stefano A, Giuliani F, Rosati G, Tamburini E, Aprea P, Vicario V, Nappi A, Vitagliano C, Casaretti R, Leone A, Petrillo A, Botti G, Delrio P, Izzo F, Perrone F, Budillon A. Randomized phase II study of valproic acid in combination with bevacizumab and oxaliplatin/fluoropyrimidine regimens in patients with RAS-mutated metastatic colorectal cancer: the REVOLUTION study protocol. Ther Adv Med Oncol 2020; 12:1758835920929589. [PMID: 32849914 PMCID: PMC7425244 DOI: 10.1177/1758835920929589] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/04/2020] [Indexed: 01/30/2023] Open
Abstract
Background Despite effective treatments, metastatic colorectal cancer (mCRC) prognosis is still poor, mostly in RAS-mutated tumors, thus suggesting the need for novel combinatorial therapies. Epigenetic alterations play an important role in initiation and progression of cancers, including CRC. Histone-deacetylase inhibitors (HDACi) have shown activity in combination with chemotherapy in the treatment of solid tumors. Owing to its HDACi activity and its safe use for epileptic disorders, valproic acid (VPA) is a good candidate for anticancer therapy that we have largely explored preclinically translating our findings in currently ongoing clinical studies. We have shown in CRC models that HDACi, including VPA, induces synergistic antitumor effects in combination with fluoropyrimidines. Furthermore, unpublished results from our group demonstrated that VPA induces differentiation and sensitization of CRC stem cells to oxaliplatin. Moreover, preclinical and clinical data suggest that HDACi may prevent/reverse anti-angiogenic resistance. Methods/Design A randomized, open-label, two-arm, multicenter phase-II study will be performed to explore whether the addition of VPA to first line bevacizumab/oxaliplatin/fluoropyrimidine regimens (mFOLFOX-6/mOXXEL) might improve progression-free survival (PFS) in RAS-mutated mCRC patients. A sample size of 200 patients was calculated under the hypothesis that the addition of VPA to chemotherapy/bevacizumab can improve PFS from 9 to 12 months, with one-sided alpha of 0.20 and a power of 0.80. Secondary endpoints are overall survival, objective response rate, metastases resection rate, toxicity, and quality of life. Moreover, the study will explore several prognostic and predictive biomarkers on blood samples, primary tumors, and on resected metastases. Discussion The "Revolution" study aims to improve the treatment efficacy of RAS-mutated mCRC through an attractive strategy evaluating the combination of VPA with standard cancer treatment. Correlative studies could identify novel biomarkers and could add new insight in the mechanism of interaction between VPA, fluoropyrimidine, oxaliplatin, and bevacizumab. Trial Registration EudraCT: 2018-001414-15; ClinicalTrials.gov identifier: NCT04310176.
Collapse
Affiliation(s)
- Antonio Avallone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Via M. Semmola, Napoli, 80131, Italy
| | | | - Elena Di Gennaro
- Experimental Pharmacology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Carmela Romano
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Filomena Calabrese
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Maria Serena Roca
- Experimental Pharmacology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Fabiana Tatangelo
- Pathology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Vincenza Granata
- Radiology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Antonio Cassata
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Ernesta Cavalcanti
- Laboratory Medicine Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Nicola Maurea
- Cardiology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Piera Maiolino
- Pharmacy Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Lucrezia Silvestro
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Alfonso De Stefano
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | | | - Gerardo Rosati
- Medical Oncology Unit, S. Carlo Hospital, Potenza, Italy
| | - Emiliano Tamburini
- Dipartimento di Oncologia e Cure Palliative, Azienda Ospedaliera Cardinale G. Panico, Tricase-Lecce, Italy
| | - Pasquale Aprea
- Vascular Access Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Valeria Vicario
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Anna Nappi
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Carlo Vitagliano
- Experimental Pharmacology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Rossana Casaretti
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Alessandra Leone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Antonella Petrillo
- Radiology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Paolo Delrio
- Colorectal Oncological Surgery, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Francesco Izzo
- Hepatobiliary Surgery Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Francesco Perrone
- Clinical Trials Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Via M. Semmola, Napoli, 80131, Italy
| |
Collapse
|
8
|
Dach K, Yaghoobi B, Schmuck MR, Carty DR, Morales KM, Lein PJ. Teratological and Behavioral Screening of the National Toxicology Program 91-Compound Library in Zebrafish (Danio rerio). Toxicol Sci 2019; 167:77-91. [PMID: 30364989 DOI: 10.1093/toxsci/kfy266] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
To screen the tens of thousands of chemicals for which no toxicity data currently exists, it is necessary to move from in vivo rodent models to alternative models, such as zebrafish. Here, we used dechorionated Tropical 5D wild-type zebrafish embryos to screen a 91-compound library provided by the National Toxicology Program (NTP) for developmental toxicity. This library contained 86 unique chemicals that included negative controls, flame retardants, polycyclic aromatic hydrocarbons (PAHs), drugs, industrial chemicals, and pesticides. Fish were exposed to 5 concentrations of each chemical or an equal amount of vehicle (0.5% DMSO) in embryo medium from 6 h post-fertilization (hpf) to 5 days post-fertilization (dpf). Fish were examined daily for mortality and teratogenic effects and photomotor behavior was assessed at 4 and 5 dpf. Of the 5 negative control compounds in the library, none caused mortality/teratogenesis, but two altered behavior. Chemicals provided in duplicate produced similar outcomes. Overall, 13 compounds caused mortality/teratology but not behavioral abnormalities, 24 only affected behavior, and 18 altered both endpoints, with behavior affected at concentrations that did not cause mortality/teratology (55/86 hits). Of the compounds that affected behavior, 52% caused behavioral abnormalities at either 4 or 5 dpf. Compounds within the same functional group caused different behavioral abnormalities, while similar behavioral patterns were caused by compounds from different groups. Our data suggest that behavior is a sensitive endpoint for developmental toxicity screening that integrates multiple modes of toxic action and is influenced by the age of the larval fish at the time of testing.
Collapse
Affiliation(s)
- Katharina Dach
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, Davis, CA California 95616
| | - Bianca Yaghoobi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, Davis, CA California 95616
| | - Martin R Schmuck
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, Davis, CA California 95616
| | - Dennis R Carty
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, Davis, CA California 95616
| | - Kelly M Morales
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, Davis, CA California 95616
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, Davis, CA California 95616
| |
Collapse
|
9
|
Murugavel S, Bugyei-Twum A, Matkar PN, Al-Mubarak H, Chen HH, Adam M, Jain S, Narang T, Abdin RM, Qadura M, Connelly KA, Leong-Poi H, Singh KK. Valproic Acid Induces Endothelial-to-Mesenchymal Transition-Like Phenotypic Switching. Front Pharmacol 2018; 9:737. [PMID: 30050438 PMCID: PMC6050396 DOI: 10.3389/fphar.2018.00737] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 06/18/2018] [Indexed: 12/14/2022] Open
Abstract
Valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, is a widely used anticonvulsant drug that is currently undergoing clinical evaluation for anticancer therapy due to its anti-angiogenic potential. Endothelial cells (ECs) can transition into mesenchymal cells and this form of EC plasticity is called endothelial-to-mesenchymal transition (EndMT), which is widely implicated in several pathologies including cancer and organ fibrosis. However, the effect of VPA on EC plasticity and EndMT remains completely unknown. We report herein that VPA-treatment significantly inhibits tube formation, migration, nitric oxide production, proliferation and migration in ECs. A microscopic evaluation revealed, and qPCR, immunofluorescence and immunoblotting data confirmed EndMT-like phenotypic switching as well as an increased expression of pro-fibrotic genes in VPA-treated ECs. Furthermore, our data confirmed important and regulatory role played by TGFβ-signaling in VPA-induced EndMT. Our qPCR array data performed for 84 endothelial genes further supported our findings and demonstrated 28 significantly and differentially regulated genes mainly implicated in angiogenesis, endothelial function, EndMT and fibrosis. We, for the first time report that VPA-treatment associated EndMT contributes to the VPA-associated loss of endothelial function. Our data also suggest that VPA based therapeutics may exacerbate endothelial dysfunction and EndMT-related phenotype in patients undergoing anticonvulsant or anticancer therapy, warranting further investigation.
Collapse
Affiliation(s)
| | - Antoinette Bugyei-Twum
- Division of Cardiology, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Pratiek N Matkar
- Division of Cardiology, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Husain Al-Mubarak
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Hao H Chen
- Division of Cardiology, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Mohamed Adam
- Division of Cardiology, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Shubha Jain
- Vascular Surgery, Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada
| | - Tanya Narang
- Faculty of Science, York University, Toronto, ON, Canada
| | - Rawand M Abdin
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Mohammad Qadura
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Vascular Surgery, Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada
| | - Kim A Connelly
- Division of Cardiology, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Howard Leong-Poi
- Division of Cardiology, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Krishna K Singh
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Vascular Surgery, Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada.,Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
10
|
Dithiolethiones: a privileged pharmacophore for anticancer therapy and chemoprevention. Future Med Chem 2018; 10:1241-1260. [DOI: 10.4155/fmc-2017-0281] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dithiolethiones are five-membered sulfur-containing cyclic scaffolds that exhibit antioxidative, anti-inflammatory, antithrombic and chemotherapeutic activities. Dithiolethiones display the chemopreventive and cytoprotective effects by activating the antioxidant response element and mounting the transcription of cytoprotective phase II enzymatic machinery. In addition, several classes of dithiolethiones efficiently modulate the activities of proteins that play crucial roles in normal and cancer cells, including glutathione S-transferase, cyclooxygenases and master regulator NF-κB. The present paper summarizes synthetic aspects, pharmacological potentials and biological attributes of dithiolethiones and its derivatives. Additionally, this review concludes with a discussion on how the current state-of-the-art technologies may help in defining a structure–activity relationship of dithiolethiones, thereby facilitating the design and synthesis of potent drug candidates.
Collapse
|
11
|
Gabriele E, Brambilla D, Ricci C, Regazzoni L, Taguchi K, Ferri N, Asai A, Sparatore A. New sulfurated derivatives of cinnamic acids and rosmaricine as inhibitors of STAT3 and NF-κB transcription factors. J Enzyme Inhib Med Chem 2017; 32:1012-1028. [PMID: 28738705 PMCID: PMC6009881 DOI: 10.1080/14756366.2017.1350658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/30/2017] [Accepted: 06/30/2017] [Indexed: 12/30/2022] Open
Abstract
A set of new sulfurated drug hybrids, mainly derived from caffeic and ferulic acids and rosmaricine, has been synthesized and their ability to inhibit both STAT3 and NF-κB transcription factors have been evaluated. Results showed that most of the new hybrid compounds were able to strongly and selectively bind to STAT3, whereas the parent drugs were devoid of this ability at the tested concentrations. Some of them were also able to inhibit the NF-κB transcriptional activity in HCT-116 cell line and inhibited HCT-116 cell proliferation in vitro with IC50 in micromolar range, thus suggesting a potential anticancer activity. Taken together, our study described the identification of new derivatives with dual STAT3/NF-κB inhibitory activity, which may represent hit compounds for developing multi-target anticancer agents.
Collapse
Affiliation(s)
- Elena Gabriele
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milano, Italy
| | - Dario Brambilla
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milano, Italy
| | - Chiara Ricci
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Luca Regazzoni
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milano, Italy
| | - Kyoko Taguchi
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Nicola Ferri
- Department of Pharmaceutical and Pharmacological Sciences, Università degli Studi di Padova, Largo Egidio Meneghetti, Padova, Italy
| | - Akira Asai
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Anna Sparatore
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
12
|
Hammers MD, Singh L, Montoya LA, Moghaddam AD, Pluth MD. Synthesis of Amino-ADT Provides Access to Hydrolytically Stable Amide-Coupled Hydrogen Sulfide Releasing Drug Targets. Synlett 2016; 27:1349-1353. [PMID: 27397970 PMCID: PMC4936786 DOI: 10.1055/s-0035-1560603] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
As additional physiological functions of hydrogen sulfide (H2S) are discovered, developing practical methods for exogenous H2S delivery is important. In particular, nonsteroidal anti-inflammatory drugs (NSAIDs) functionalized with H2S-releasing anethole dithiolethione (ADT-OH) through ester bonds are being investigated for their combined anti-inflammatory and antioxidant potential. The chemical robustness of the connection between drug and H2S-delivery components, however, is a key and controllable linkage in these compounds. Because esters are susceptible to hydrolysis, particularly under acidic conditions such as stomach acid in oral drug delivery applications, we report here a simple synthesis of amino-ADT (ADT-NH2 ) and provide conditions for successful ADT-NH2 derivatization with the drugs naproxen and valproic acid. Using UV-vis spectroscopy and HPLC analysis, we demonstrate that amide-functionalized ADT derivatives are significantly more resistant to hydrolysis than ester-functionalized ADT derivatives.
Collapse
Affiliation(s)
- Matthew D. Hammers
- Department of Chemistry and Biochemistry, Institute of Molecular Biology, Materials Science Institute, University of Oregon, Eugene, OR 97403-1253, USA
| | - Loveprit Singh
- Department of Chemistry and Biochemistry, Institute of Molecular Biology, Materials Science Institute, University of Oregon, Eugene, OR 97403-1253, USA
| | - Leticia A. Montoya
- Department of Chemistry and Biochemistry, Institute of Molecular Biology, Materials Science Institute, University of Oregon, Eugene, OR 97403-1253, USA
| | - Alan D. Moghaddam
- Department of Chemistry and Biochemistry, Institute of Molecular Biology, Materials Science Institute, University of Oregon, Eugene, OR 97403-1253, USA
| | - Michael D. Pluth
- Department of Chemistry and Biochemistry, Institute of Molecular Biology, Materials Science Institute, University of Oregon, Eugene, OR 97403-1253, USA
| |
Collapse
|
13
|
Abstract
Hydrogen sulfide (H(2)S) is a gasomediator synthesized from L- and D-cysteine in various tissues. It is involved in a number of physiological and pathological processes. H(2)S exhibits antiatherosclerotic, vasodilator, and proangiogenic properties, and protects the kidney and heart from damage following ischemia/reperfusion injury. H(2)S donors may be natural or synthetic, and may be used for the safe treatment of a wide range of diseases. This review article summarizes the current state of knowledge of the therapeutic function of H(2)S.
Collapse
Affiliation(s)
- Beata Olas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland.
| |
Collapse
|
14
|
Zhao Y, You W, Zheng J, Chi Y, Tang W, Du R. Valproic acid inhibits the angiogenic potential of cervical cancer cells via HIF-1α/VEGF signals. Clin Transl Oncol 2016; 18:1123-1130. [DOI: 10.1007/s12094-016-1494-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 02/20/2016] [Indexed: 01/14/2023]
|
15
|
Popov SA, Kornaukhova LM, Shpatov AV, Grigoŕev IA. Synthesis of Conjugate Esters of 5-(4-Hydroxyphenyl)-3H-1,2-Dithiole-3-Thione and Ursolic Acid 3-O-ACYL Derivatives. Chem Nat Compd 2016. [DOI: 10.1007/s10600-016-1588-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
16
|
Bos EM, van Goor H, Joles JA, Whiteman M, Leuvenink HGD. Hydrogen sulfide: physiological properties and therapeutic potential in ischaemia. Br J Pharmacol 2016; 172:1479-93. [PMID: 25091411 DOI: 10.1111/bph.12869] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Revised: 06/19/2014] [Accepted: 07/27/2014] [Indexed: 12/19/2022] Open
Abstract
Hydrogen sulfide (H2 S) has become a molecule of high interest in recent years, and it is now recognized as the third gasotransmitter in addition to nitric oxide and carbon monoxide. In this review, we discuss the recent literature on the physiology of endogenous and exogenous H2 S, focusing upon the protective effects of hydrogen sulfide in models of hypoxia and ischaemia.
Collapse
Affiliation(s)
- Eelke M Bos
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
17
|
Feng W, Teo XY, Novera W, Ramanujulu PM, Liang D, Huang D, Moore PK, Deng LW, Dymock BW. Discovery of New H2S Releasing Phosphordithioates and 2,3-Dihydro-2-phenyl-2-sulfanylenebenzo[d][1,3,2]oxazaphospholes with Improved Antiproliferative Activity. J Med Chem 2015; 58:6456-80. [DOI: 10.1021/acs.jmedchem.5b00848] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Wei Feng
- Department of Pharmacy, National University of Singapore, Block S4A Level 3, 18 Science Drive 4, Singapore 117543
- Life
Sciences Institute, Centre for Life Sciences, National University of Singapore, Level 5, 28 Medical Drive, Singapore 117456
| | - Xin-Yi Teo
- Life
Sciences Institute, Centre for Life Sciences, National University of Singapore, Level 5, 28 Medical Drive, Singapore 117456
| | - Wisna Novera
- Department
of Biochemistry, National University of Singapore, Block MD7
04-06, 8 Medical Drive, Singapore 117596
| | - Pondy Murugappan Ramanujulu
- Department of Pharmacy, National University of Singapore, Block S4A Level 3, 18 Science Drive 4, Singapore 117543
- Life
Sciences Institute, Centre for Life Sciences, National University of Singapore, Level 5, 28 Medical Drive, Singapore 117456
| | - Dong Liang
- Department of Chemistry, National University of Singapore, Block S8 Level 3, Science Drive 3, Singapore 117543
| | - Dejian Huang
- Department of Chemistry, National University of Singapore, Block S8 Level 3, Science Drive 3, Singapore 117543
| | - Philip K. Moore
- Life
Sciences Institute, Centre for Life Sciences, National University of Singapore, Level 5, 28 Medical Drive, Singapore 117456
- Department of Pharmacology, National University of Singapore, UHL Level 5-02R, Lee Kong Chian Wing, 21 Lower Kent Ridge Road, Singapore 119077
| | - Lih-Wen Deng
- Department
of Biochemistry, National University of Singapore, Block MD7
04-06, 8 Medical Drive, Singapore 117596
| | - Brian W. Dymock
- Department of Pharmacy, National University of Singapore, Block S4A Level 3, 18 Science Drive 4, Singapore 117543
| |
Collapse
|
18
|
Role of Hydrogen Sulfide in Ischemia-Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:186908. [PMID: 26064416 PMCID: PMC4443900 DOI: 10.1155/2015/186908] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/10/2014] [Accepted: 12/10/2014] [Indexed: 12/13/2022]
Abstract
Ischemia-reperfusion (I/R) injury is one of the major causes of high morbidity, disability, and mortality in the world. I/R injury remains a complicated and unresolved situation in clinical practice, especially in the field of solid organ transplantation. Hydrogen sulfide (H2S) is the third gaseous signaling molecule and plays a broad range of physiological and pathophysiological roles in mammals. H2S could protect against I/R injury in many organs and tissues, such as heart, liver, kidney, brain, intestine, stomach, hind-limb, lung, and retina. The goal of this review is to highlight recent findings regarding the role of H2S in I/R injury. In this review, we present the production and metabolism of H2S and further discuss the effect and mechanism of H2S in I/R injury.
Collapse
|
19
|
Abstract
Hydrogen sulfide (H2S) is now recognized as the so called "third gasotransmitter" taking its place alongside nitric oxide and carbon monoxide. In recent years, H2S has been reported to exhibit a diverse range of pharmacological effects in biological systems. Much of this evidence is derived from a combination of conventional pharmacological and genetic approaches coupled with the use of chemical compounds such as sodium hydrosulfide, a rapid H2S releasing donor. Developments in the design of new drug entities which attempt to take into account physicochemical properties, targeting to specific cellular organelles, triggering of H2S release upon specific chemical reactions in the cell, and controlling the release of H2S over extended periods of time have been described. For most of these molecules, little or no work has been conducted to determine their biological activity or possible therapeutic effects. It is therefore not clear whether such molecules have therapeutic potential which highlights the need for further in vivo studies. One exception to the general rule is GYY4137 (morpholin-4-ium 4-methoxyphenyl(morpholino) phosphinodithioate), a slow releasing H2S donor, which has been evaluated for activity in a range of pharmacological models both in vitro and in vivo. GYY4137 was first reported to release H2S and exhibit vasodilator activity over 5 years ago and, to date, GYY4137 is becoming increasingly employed as a pharmacological "tool" to explore the biological functions of H2S.
Collapse
Affiliation(s)
- Peter Rose
- University of Lincoln, Lincoln, Lincolnshire, United Kingdom
| | - Brian W Dymock
- Department of Pharmacy, National University of Singapore, Singapore
| | - Philip K Moore
- Neurobiology Program, Life Science Institute and Department of Pharmacology, National University of Singapore, Singapore.
| |
Collapse
|
20
|
Abstract
Pharmacological concentrations of H2S donors inhibit some T cell functions by inhibiting mitochondrial function, but evidence is also emerging that H2S at physiological concentrations produced via chemical sources and endogenously is a positive physiological mediator of T cell function. Expression of the H2S biosynthetic enzymes cystathionine γ-lyase (CSE) and cystathionine β-synthase (CBS) is induced in response to T cell receptor signaling. Inhibiting the induction of these enzymes limits T cell activation and proliferation, which can be overcome by exposure to exogenous H2S at submicromolar concentrations. Exogenous H2S at physiological concentrations increases the ability of T cells to form an immunological synapse by altering cytoskeletal actin dynamics and increasing the reorientation of the microtubule-organizing center. Downstream, H2S enhances T cell receptor-dependent induction of CD69, CD25, and Interleukin-2 (IL-2) gene expression. The T cell stimulatory activity of H2S is enhanced under hypoxic conditions that limit its oxidative metabolism by mitochondrial and nonenzymatic processes. Studies of the receptor CD47 have revealed the first endogenous inhibitory signaling pathway that regulates H2S signaling in T cells. Binding of the secreted protein thrombospondin-1 to CD47 elicits signals that block the stimulatory activity of exogenous H2S on T cell activation and limit the induction of CSE and CBS gene expression. CD47 signaling thereby inhibits T cell receptor-mediated T cell activation.
Collapse
|
21
|
Sparatore A, Santus G, Giustarini D, Rossi R, Del Soldato P. Therapeutic potential of new hydrogen sulfide-releasing hybrids. Expert Rev Clin Pharmacol 2014; 4:109-21. [DOI: 10.1586/ecp.10.122] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
22
|
Pharmacological characterization of the vascular effects of aryl isothiocyanates: Is hydrogen sulfide the real player? Vascul Pharmacol 2014; 60:32-41. [DOI: 10.1016/j.vph.2013.11.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 11/12/2013] [Accepted: 11/16/2013] [Indexed: 12/16/2022]
|
23
|
Martelli A, Testai L, Citi V, Marino A, Pugliesi I, Barresi E, Nesi G, Rapposelli S, Taliani S, Da Settimo F, Breschi MC, Calderone V. Arylthioamides as H2S Donors: l-Cysteine-Activated Releasing Properties and Vascular Effects in Vitro and in Vivo. ACS Med Chem Lett 2013; 4:904-8. [PMID: 24900583 DOI: 10.1021/ml400239a] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 08/08/2013] [Indexed: 12/18/2022] Open
Abstract
A small library of arylthioamides 1-12 was easily synthesized, and their H2S-releasing properties were evaluated both in the absence or in the presence of an organic thiol such as l-cysteine. A number of arylthioamides (1-3 and 7) showed a slow and l-cysteine-dependent H2S-releasing mechanism, similar to that exhibited by the reference slow H2S-releasing agents, such as diallyl disulfide (DADS) and the phosphinodithioate derivative GYY 4137. Compound 1 strongly abolished the noradrenaline-induced vasoconstriction in isolated rat aortic rings and hyperpolarized the membranes of human vascular smooth muscle cells in a concentration-dependent fashion. Finally, a significant reduction of the systolic blood pressure of anesthetized normotensive rats was observed after its oral administration. Altogether these results highlighted the potential of arylthioamides 1-3 and 7 as H2S-donors for basic studies, and for the rational design/development of promising pharmacotherapeutic agents to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Alma Martelli
- Dipartimento
di Farmacia, Università di Pisa,
via Bonanno, 6, I-56126 Pisa, Italy
| | - Lara Testai
- Dipartimento
di Farmacia, Università di Pisa,
via Bonanno, 6, I-56126 Pisa, Italy
| | - Valentina Citi
- Dipartimento
di Farmacia, Università di Pisa,
via Bonanno, 6, I-56126 Pisa, Italy
| | - Alice Marino
- Dipartimento
di Farmacia, Università di Pisa,
via Bonanno, 6, I-56126 Pisa, Italy
| | - Isabella Pugliesi
- Dipartimento
di Farmacia, Università di Pisa,
via Bonanno, 6, I-56126 Pisa, Italy
| | - Elisabetta Barresi
- Dipartimento
di Farmacia, Università di Pisa,
via Bonanno, 6, I-56126 Pisa, Italy
| | - Giulia Nesi
- Dipartimento
di Farmacia, Università di Pisa,
via Bonanno, 6, I-56126 Pisa, Italy
| | - Simona Rapposelli
- Dipartimento
di Farmacia, Università di Pisa,
via Bonanno, 6, I-56126 Pisa, Italy
| | - Sabrina Taliani
- Dipartimento
di Farmacia, Università di Pisa,
via Bonanno, 6, I-56126 Pisa, Italy
| | - Federico Da Settimo
- Dipartimento
di Farmacia, Università di Pisa,
via Bonanno, 6, I-56126 Pisa, Italy
| | - Maria C. Breschi
- Dipartimento
di Farmacia, Università di Pisa,
via Bonanno, 6, I-56126 Pisa, Italy
| | - Vincenzo Calderone
- Dipartimento
di Farmacia, Università di Pisa,
via Bonanno, 6, I-56126 Pisa, Italy
| |
Collapse
|
24
|
Si J, Zhang H, Wang Z, Wu Z, Lu J, Di C, Zhou X, Wang X. Effects of (12)C(6+) ion radiation and ferulic acid on the zebrafish (Danio rerio) embryonic oxidative stress response and gene expression. Mutat Res 2013; 745-746:26-33. [PMID: 23535216 DOI: 10.1016/j.mrfmmm.2013.03.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 03/12/2013] [Accepted: 03/15/2013] [Indexed: 06/02/2023]
Abstract
The effects of carbon ion irradiation and ferulic acid (FA) on the induction of oxidative stress and alteration of gene expression were studied in zebrafish (Danio rerio) embryos. Zebrafish embryos at 8 hpf were divided into seven groups: the control group; the 1Gy, 3Gy and 7Gy irradiation groups; and three FA-pre-treated irradiation groups. In the irradiated groups, a significant increase in the teratogenesis of the zebrafish embryos and oxidative stress was accompanied by increased malondialdehyde (MDA) content, decreased glutathione (GSH) content and alterations in antioxidant enzyme activities (such as catalase [CAT] and superoxide dismutase [SOD]). Moreover, the mRNA levels for Cu/Zn-sod, Mn-sod, cat and gpx, the genes encoding these antioxidant proteins, were altered significantly. However, the mRNA expression patterns were not in accordance with those of the antioxidant enzymes and were more sensitive under low-dose irradiation. In addition, we detected the mRNA expression of ucp-2 and bcl-2, which are located at the mitochondrial inner membrane and related to reactive oxidative species (ROS) production. In the irradiated groups, the mRNA level of ucp-2 was significantly increased, whereas the mRNA level of bcl-2 was significantly decreased. Supplementation with FA, an antioxidant, was better able to reduce the irradiation-induced oxidative damage marked by changes in mortality, morphology, antioxidant enzyme activities and the MDA and GSH content, as well as in the mRNA expression levels. Overall, this study provided helpful information about the transcriptional effects of irradiation to better understand the mechanism of carbon ion-induced oxidative stress and FA-induced radioprotective effects.
Collapse
Affiliation(s)
- Jing Si
- Department of Heavy Ion Radiation Medicine, Chinese Academy of Sciences, Lanzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Quezada M, Alvarez M, Peña OA, Henríquez S, d' Alençon CA, Lange S, Oliva B, Owen GI, Allende ML. Antiangiogenic, antimigratory and antiinflammatory effects of 2-methoxyestradiol in zebrafish larvae. Comp Biochem Physiol C Toxicol Pharmacol 2013; 157:141-9. [PMID: 23142146 DOI: 10.1016/j.cbpc.2012.10.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Revised: 10/30/2012] [Accepted: 10/31/2012] [Indexed: 12/13/2022]
Abstract
2-Methoxyestradiol (2ME), an endogenous metabolite of 17β-estradiol, has been previously reported to possess antiangiogenic and antitumor properties. Herein, we demonstrate that the effects of this antiangiogenic steroid can be readily assayed in live zebrafish, introducing a convenient and robust new model system as a screening tool for both single cell and collective cell migration assays. Using the in vitro mammalian endothelial cell line EA.hy926, we first show that cell migration and angiogenesis, as estimated by wound assay and tube formation respectively, are antagonized by 2ME. In zebrafish (Danio rerio) larvae, dose-dependent exposure to 2ME diminishes (1) larval angiogenesis, (2) leukocyte recruitment to damaged lateral line neuromasts and (3) retards the lateral line primordium in its migration along the body. Our results indicate that 2ME has an effect on collective cell migration in vivo as well as previously reported anti-tumorigenic activity and suggests that the molecular mechanisms governing cell migration in a variety of contexts are conserved between fish and mammals. Moreover, we exemplify the versatility of the zebrafish larvae for testing diverse physiological processes and screening for antiangiogenic and antimigratory drugs in vivo.
Collapse
Affiliation(s)
- Marisol Quezada
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wang B, Yang R, Wu Y, Li H, Hu Z, Chen Y, Zou S. Sodium valproate inhibits the growth of human cholangiocarcinoma in vitro and in vivo. Gastroenterol Res Pract 2013; 2013:374593. [PMID: 24324485 PMCID: PMC3845332 DOI: 10.1155/2013/374593] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 08/27/2013] [Indexed: 01/08/2023] Open
Abstract
Background. None of treatment options for Cholangiocarcinoma (CCA), including surgery, adjuvant radiotherapy and chemotherapy, and ultimately liver transplantation, have been shown to substantially improve the survival rate in patients with CCA. Valproic acid (VPA), a histone deacetylase inhibitor, has been shown to display potent antitumor effects. In this study, sodium valproate, the clinically available form of VPA, was tested for its ability to inhibit the growth of cholangiocarcinoma cells, both in vitro and in vivo. Materials and Methods. Cholangiocarcinoma cells (TFK-1, QBC939, and CCLP1) of different origins were treated with sodium valproate to determine their effects on cell proliferation and differentiation, cell cycle regulation, apoptosis, and autophagy. The in vivo effects of sodium valproate on cholangiocarcinoma growth were assessed using a xenograft mouse model injected with TFK-1 cells. Results. Sodium valproate inhibited cholangiocarcinoma cell growth by inducing cell cycle arrest, cell differentiation, and apoptosis; sodium valproate effects were independent of autophagy. Tumor growth inhibition was also observed in vivo using TFK-1 xenografts. Conclusion. The in vitro and in vivo outcomes provide preclinical rationale for clinical evaluation of sodium valproate, alone or in combination with other drugs, to improve patient outcome in cholangiocarcinoma.
Collapse
Affiliation(s)
- Bing Wang
- Department of General Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rui Yang
- Department of General Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yue Wu
- Department of General Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongbo Li
- Department of General Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zouxiao Hu
- Department of General Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yongjun Chen
- Department of General Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shengquan Zou
- Department of General Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
- *Shengquan Zou:
| |
Collapse
|
27
|
Recent Progress in 1,2-Dithiole-3-thione Chemistry. ADVANCES IN HETEROCYCLIC CHEMISTRY 2013. [DOI: 10.1016/b978-0-12-407777-5.00001-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
|
28
|
Pruvot B, Quiroz Y, Voncken A, Jeanray N, Piot A, Martial JA, Muller M. A panel of biological tests reveals developmental effects of pharmaceutical pollutants on late stage zebrafish embryos. Reprod Toxicol 2012; 34:568-83. [DOI: 10.1016/j.reprotox.2012.07.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 07/09/2012] [Accepted: 07/13/2012] [Indexed: 01/10/2023]
|
29
|
Yu X, Tong Y, Kwok HF, Sze SCW, Zhong L, Lau CBS, Ge W. Anti-angiogenic activity of Erxian Decoction, a traditional Chinese herbal formula, in zebrafish. Biol Pharm Bull 2012; 35:2119-27. [PMID: 23018578 DOI: 10.1248/bpb.b12-00130] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Erxian Decoction (EXD), a traditional Chinese herbal formula, has been used to treat menopausal symptoms and other aging diseases for several decades. Recently, our laboratory found that EXD could inhibit the proliferation of breast cancer cells. This activity may be mediated by anti-angiogenic action. To investigate the anti-angiogenic activity of EXD, its inhibitory effect on blood vessel formation was evaluated using both wild type and transgenic zebrafish embryos with fluorescent vasculature in vivo. Both semi-quantitative and real-time quantitative polymerase chain reaction (qPCR) were carried out to evaluate the effect of EXD on the expression of several genes closely associated with angiogenesis in zebrafish. EXD was found to inhibit vessel formation in zebrafish embryos in a dose- and time-dependent manner. Furthermore, it reduced the mRNA expression of vascular endothelial growth factor A (VEGF-A) and the protein level of hypoxia inducible factor 1α (HIF-1α) in the embryos, suggesting the involvement of HIF-1 mediated VEGF-A signaling pathway in the anti-angiogenic action of EXD. The anti-angiogenic activity of EXD provides new insights to its clinical application and may in the future lead to the development of potential drugs for treating various cancers, especially in menopausal period.
Collapse
Affiliation(s)
- Xiaobin Yu
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, China
| | | | | | | | | | | | | |
Collapse
|
30
|
Whiteman M, Winyard PG. Hydrogen sulfide and inflammation: the good, the bad, the ugly and the promising. Expert Rev Clin Pharmacol 2012; 4:13-32. [PMID: 22115346 DOI: 10.1586/ecp.10.134] [Citation(s) in RCA: 240] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Hydrogen sulfide is rapidly gaining ground as a physiological mediator of inflammation, but there is no clear consensus as to its precise role in inflammatory signaling. This article discusses the disparate anti-inflammatory ('the good') and proinflammatory ('the bad') effects of endogenous and pharmacological H(2)S in disparate animal model and cell culture systems. We also discuss 'the ugly', such as problems of using wholly specific inhibitors of enzymatic H(2)S synthesis, and the use of pharmacological donor compounds, which release H(2)S too quickly to be physiologically representative of endogenous H(2)S synthesis. Furthermore, recently developed slow-release H(2)S donors, which offer a more physiological approach to understanding the complex role of H(2)S in acute and chronic inflammation ('the promising') are discussed.
Collapse
Affiliation(s)
- Matthew Whiteman
- Peninsula Medical School, University of Exeter, St Luke's Campus, Magdalen Road, Exeter, Devon, EX1 2LU, UK.
| | | |
Collapse
|
31
|
Wang R. Physiological implications of hydrogen sulfide: a whiff exploration that blossomed. Physiol Rev 2012; 92:791-896. [PMID: 22535897 DOI: 10.1152/physrev.00017.2011] [Citation(s) in RCA: 1421] [Impact Index Per Article: 109.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The important life-supporting role of hydrogen sulfide (H(2)S) has evolved from bacteria to plants, invertebrates, vertebrates, and finally to mammals. Over the centuries, however, H(2)S had only been known for its toxicity and environmental hazard. Physiological importance of H(2)S has been appreciated for about a decade. It started by the discovery of endogenous H(2)S production in mammalian cells and gained momentum by typifying this gasotransmitter with a variety of physiological functions. The H(2)S-catalyzing enzymes are differentially expressed in cardiovascular, neuronal, immune, renal, respiratory, gastrointestinal, reproductive, liver, and endocrine systems and affect the functions of these systems through the production of H(2)S. The physiological functions of H(2)S are mediated by different molecular targets, such as different ion channels and signaling proteins. Alternations of H(2)S metabolism lead to an array of pathological disturbances in the form of hypertension, atherosclerosis, heart failure, diabetes, cirrhosis, inflammation, sepsis, neurodegenerative disease, erectile dysfunction, and asthma, to name a few. Many new technologies have been developed to detect endogenous H(2)S production, and novel H(2)S-delivery compounds have been invented to aid therapeutic intervention of diseases related to abnormal H(2)S metabolism. While acknowledging the challenges ahead, research on H(2)S physiology and medicine is entering an exponential exploration era.
Collapse
Affiliation(s)
- Rui Wang
- Department of Biology, Lakehead University, Thunder Bay, Ontario, Canada.
| |
Collapse
|
32
|
Frantzias J, Logan JG, Mollat P, Sparatore A, Del Soldato P, Ralston SH, Idris AI. Hydrogen sulphide-releasing diclofenac derivatives inhibit breast cancer-induced osteoclastogenesis in vitro and prevent osteolysis ex vivo. Br J Pharmacol 2012; 165:1914-1925. [PMID: 21955294 DOI: 10.1111/j.1476-5381.2011.01704.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Hydrogen sulphide (H(2)S) and prostaglandins are both involved in inflammation, cancer and bone turnover, and non-steroidal anti-inflammatory drugs (NSAIDs) and H(2)S donors exhibit anti-inflammatory and anti-tumour properties. H(2)S-releasing diclofenac (S-DCF) derivatives are a novel class of NSAIDs combining the properties of a H(2)S donor with those of a conventional NSAID. EXPERIMENTAL APPROACH We studied the effects of the S-DCF derivatives ACS15 and ACS32 on osteoclast and osteoblast differentiation and activity in vitro, human and mouse breast cancer cells support for osteoclast formation and signalling in vitro, and osteolysis ex vivo. KEY RESULTS The S-diclofenac derivatives ACS15 and ACS32 inhibited the increase in osteoclast formation induced by human MDA-MB-231 and MCF-7 and mouse 4T1 breast cancer cells without affecting breast cancer cell viability. Conditioned media from human MDA-MB-231 cells enhanced IκB phosphorylation and osteoclast formation and these effects were significantly inhibited following treatment by ACS15 and ACS32, whereas the parent compound diclofenac had no effects. ACS15 and ACS32 inhibited receptor activator of NFκB ligand-induced osteoclast formation and resorption, and caused caspase-3 activation and apoptosis in mature osteoclasts via a mechanism dependent on IKK/NFκB inhibition. In calvaria organ culture, human MDA-MB-231 cells caused osteolysis, and this effect was completely prevented following treatment with ACS15 and ACS32. CONCLUSIONS AND IMPLICATIONS S-diclofenac derivatives inhibit osteoclast formation and activity, suppress breast cancer cell support for osteoclastogenesis and prevent osteolysis. This suggests that H(2)S-releasing diclofenac derivatives exhibit anti-resorptive properties, which might be of clinical value in the treatment of osteolytic bone disease.
Collapse
Affiliation(s)
- J Frantzias
- The Centre for Molecular Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UKEdinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UKGalapagos SASU, Romainville, FranceDepartment of Pharmaceutical Sciences 'P. Pratesi', Università degli Studi di Milano, Milan, ItalyCTG Pharma S.r.l., Milan, Italy
| | - J G Logan
- The Centre for Molecular Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UKEdinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UKGalapagos SASU, Romainville, FranceDepartment of Pharmaceutical Sciences 'P. Pratesi', Università degli Studi di Milano, Milan, ItalyCTG Pharma S.r.l., Milan, Italy
| | - P Mollat
- The Centre for Molecular Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UKEdinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UKGalapagos SASU, Romainville, FranceDepartment of Pharmaceutical Sciences 'P. Pratesi', Università degli Studi di Milano, Milan, ItalyCTG Pharma S.r.l., Milan, Italy
| | - A Sparatore
- The Centre for Molecular Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UKEdinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UKGalapagos SASU, Romainville, FranceDepartment of Pharmaceutical Sciences 'P. Pratesi', Università degli Studi di Milano, Milan, ItalyCTG Pharma S.r.l., Milan, Italy
| | - P Del Soldato
- The Centre for Molecular Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UKEdinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UKGalapagos SASU, Romainville, FranceDepartment of Pharmaceutical Sciences 'P. Pratesi', Università degli Studi di Milano, Milan, ItalyCTG Pharma S.r.l., Milan, Italy
| | - S H Ralston
- The Centre for Molecular Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UKEdinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UKGalapagos SASU, Romainville, FranceDepartment of Pharmaceutical Sciences 'P. Pratesi', Università degli Studi di Milano, Milan, ItalyCTG Pharma S.r.l., Milan, Italy
| | - A I Idris
- The Centre for Molecular Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UKEdinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UKGalapagos SASU, Romainville, FranceDepartment of Pharmaceutical Sciences 'P. Pratesi', Università degli Studi di Milano, Milan, ItalyCTG Pharma S.r.l., Milan, Italy
| |
Collapse
|
33
|
Paramasivam A, Kalaimangai M, Sambantham S, Anandan B, Jayaraman G. Anti-angiogenic activity of thymoquinone by the down-regulation of VEGF using zebrafish (Danio rerio) model. ACTA ACUST UNITED AC 2012. [DOI: 10.1016/j.bionut.2012.03.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
34
|
Abstract
SIGNIFICANCE An abundance of experimental evidence suggests that hydrogen sulfide (H(2)S) plays a prominent role in physiology and pathophysiology. Many targets exist for H(2)S therapy. The molecular targets of H(2)S include proteins, enzymes, transcription factors, and membrane ion channels. RECENT ADVANCES Novel H(2)S precursors are being synthesized and discovered that are capable of releasing H(2)S in a slow and sustained manner. This presents a novel and advantageous approach to H(2)S therapy for treatment of chronic conditions associated with a decline in endogenous H(2)S, such as diabetes and cardiovascular disease. CRITICAL ISSUES While H(2)S is cytoprotective at physiological concentrations, it is not universally cytoprotective, as it appears to have pro-apoptotic actions in cancer cells and is well known to be toxic at supraphysiological concentrations. Many of the pleiotropic effects of H(2)S on health are associated with the inhibition of inflammation and upregulation of prosurvival pathways. The powerful anti-inflammatory, cytoprotective, immunomodulating, and trophic effects of H(2)S on the vast majority of normal cells seem to be mediated mainly by its actions as an extremely versatile direct and indirect antioxidant and free radical scavenger. While the overall effects of H(2)S on transformed (i.e., malignant) cells can be characterized as pro-oxidant and pro-apoptotic, they contrast sharply with the cytoprotective effects on most normal cells. FUTURE DIRECTIONS H(2)S has become a molecule of great interest, and several slow-releasing H(2)S prodrugs are currently under development. We believe that additional agents regulating H(2)S bioavailability will be developed during the next 10 years.
Collapse
Affiliation(s)
- Benjamin Lee Predmore
- Department of Surgery-Cardiothoracic Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | |
Collapse
|
35
|
Tesei A, Brigliadori G, Carloni S, Fabbri F, Ulivi P, Arienti C, Sparatore A, Del Soldato P, Pasini A, Amadori D, Silvestrini R, Zoli W. Organosulfur derivatives of the HDAC inhibitor valproic acid sensitize human lung cancer cell lines to apoptosis and to cisplatin cytotoxicity. J Cell Physiol 2012; 227:3389-96. [DOI: 10.1002/jcp.24039] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
36
|
Favoni RE, Daga A, Malatesta P, Florio T. Preclinical studies identify novel targeted pharmacological strategies for treatment of human malignant pleural mesothelioma. Br J Pharmacol 2012; 166:532-53. [PMID: 22289125 PMCID: PMC3417486 DOI: 10.1111/j.1476-5381.2012.01873.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 12/01/2011] [Accepted: 12/20/2011] [Indexed: 12/22/2022] Open
Abstract
The incidence of human malignant pleural mesothelioma (hMPM) is still increasing worldwide. hMPM prognosis is poor even if the median survival time has been slightly improved after the introduction of the up-to-date chemotherapy. Nevertheless, large phase II/III trials support the combination of platinum derivatives and pemetrexed or raltitrexed, as preferred first-line schedule. Better understanding of the molecular machinery of hMPM will lead to the design and synthesis of novel compounds targeted against pathways identified as crucial for hMPM cell proliferation and spreading. Among them, several receptors tyrosine kinase show altered activity in subsets of hMPM. This observation suggests that these kinases might represent novel therapeutic targets in this chemotherapy-resistant disease. Over these foundations, several promising studies are ongoing at preclinical level and novel molecules are currently under evaluation as well. Yet, established tumour cell lines, used for decades to investigate the efficacy of anticancer agents, although still the main source of drug efficacy studies, after long-term cultures tend to biologically diverge from the original tumour, limiting the predictive potential of in vivo efficacy. Cancer stem cells (CSCs), a subpopulation of malignant cells capable of self-renewal and multilineage differentiation, are believed to play an essential role in cancer initiation, growth, metastasization and relapse, being responsible of chemo- and radiotherapy refractoriness. According to the current carcinogenesis theory, CSCs represent the tumour-initiating cell (TIC) fraction, the only clonogenic subpopulation able to originate a tumour mass. Consequently, the recently described isolation of TICs from hMPM, the proposed main pharmacological target for novel antitumoural drugs, may contribute to better dissect the biology and multidrug resistance pathways controlling hMPM growth.
Collapse
Affiliation(s)
- Roberto E Favoni
- IRCCS A.O.U. San Martino-IST, Laboratory of Gene Transfer, Genoa, Italy.
| | | | | | | |
Collapse
|
37
|
Zagouri F, Sergentanis TN, Chrysikos D, Filipits M, Bartsch R. Molecularly targeted therapies in cervical cancer. A systematic review. Gynecol Oncol 2012; 126:291-303. [PMID: 22504292 DOI: 10.1016/j.ygyno.2012.04.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 04/03/2012] [Accepted: 04/04/2012] [Indexed: 12/22/2022]
Abstract
Cervical cancer represents the third most common cause of female cancer mortality. Even with the best currently available treatment, a significant proportion of patients will experience recurrence and eventually die. Evidently, there is a clear need for the development of new agents with novel mechanisms of action in this disease. A number of biological agents modulating different signal transduction pathways are currently in clinical development, inhibiting angiogenesis, targeting epidermal growth factor receptor, cell cycle, histone deacetylases, cyclooxygenase-2 (COX-2), or mammalian target of rapamycin (mTOR). This is the first systematic review of the literature to synthesize all available data emerging from trials and evaluate the efficacy and safety of molecularly targeted drugs in cervical cancer. However, it should be stressed that relatively fewer molecularly targeted agents have been tested in cervical cancer in comparison with other cancer types; of note, no related phase 3 trials have been published and consequently no agent has been approved for use in clinical practice. Nevertheless, the promising results of bevacizumab in therapeutic trials for cervical cancer have shown that targeting the VEGF pathway is an attractive therapeutic strategy. As knowledge accumulates on the molecular mechanisms underlying carcinogenesis in the cervix, the anticipated development and assessment of molecularly targeted agents may offer a promising perspective for cervical cancer.
Collapse
Affiliation(s)
- Flora Zagouri
- Comprehensive Cancer Center Vienna, Department of Medicine I/Division of Oncology, Medical University of Vienna, Austria.
| | | | | | | | | |
Collapse
|
38
|
Wu M, Khan IA, Dasmahapatra AK. Valproate-induced teratogenesis in Japanese rice fish (Oryzias latipes) embryogenesis. Comp Biochem Physiol C Toxicol Pharmacol 2012; 155:528-37. [PMID: 22249148 DOI: 10.1016/j.cbpc.2012.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Revised: 01/04/2012] [Accepted: 01/05/2012] [Indexed: 01/18/2023]
Abstract
Fertilized eggs of Japanese rice fish (medaka) at three developmental stages (Iwamatsu stages 4-30) were exposed to waterborne valproic acid (VPA) (0-80 mM) in hatching solution for 48 h. The amount of valproate to cause 50% mortality (IC(50)) is found to be developmental stage-specific. The embryos were more sensitive to valproate at early stages of development (Iwamatsu stages 4-10) than in the embryos in late stages (Iwamatsu stages 17-30). Valproate exposed embryos have microcephaly and disrupted cardiovasculature with delayed vessel circulation, thrombus formation, and slow heart rate. The hatching efficiency is also reduced by valproate exposure due to developmental delay. The mRNA analysis of nine genes belong to oxidative stress (catalase, gsr, gst), neurogenesis (iro3, wnt1, shh, otx2, nlgn3b) and cell cycle regulation (ccna2) have been done. It was observed that the genes belong to oxidative stress remained unaltered after valproate exposure. However, some of the genes belong to neurogenesis (wnt1,shh, otx2 and nlgn3b) and cell cycle (ccna2) showed developmental stage-specific alteration after valproate exposure. This study indicates that valproate is able to induce some of the phenotypic features which are analogous to human fetal valproate syndrome (FVS). Modulation of genes expressed in neural tissues indicates that this fish can be used to analyze the mechanisms of many neurobehavioral disorders like Autism spectrum disorder (ASD) in human.
Collapse
Affiliation(s)
- Mengmeng Wu
- National Center for Natural Products Research, University of Mississippi, University, Mississippi 38677, USA
| | | | | |
Collapse
|
39
|
Switzer CH, Cheng RYS, Ridnour LA, Murray MC, Tazzari V, Sparatore A, Del Soldato P, Hines HB, Glynn SA, Ambs S, Wink DA. Dithiolethiones inhibit NF-κB activity via covalent modification in human estrogen receptor-negative breast cancer. Cancer Res 2012; 72:2394-404. [PMID: 22436383 DOI: 10.1158/0008-5472.can-11-3115] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The NF-κB transcription factor family influences breast cancer outcomes by regulating genes involved in tumor progression, angiogenesis, and metastasis. Dithiolethiones, a class of naturally occurring compounds with cancer chemoprevention effects that have become clinically available, have been found to inhibit NF-κB activity. However, the mechanism of this inhibition has not been identified, and the influence of dithiolethines on NF-κB pathway in breast cancer cells has not been examined. Here, we investigated the chemical and biochemical effects of dithiolethione on NF-κB and downstream effector molecules in estrogen receptor-negative breast cancer cells and murine tumor xenografts. The dithiolethiones ACS-1 and ACS-2 inhibited NF-κB transcriptional activity. Interestingly, this inhibition was not due to H(2)S release or protein phosphatase 2A activation, which are key properties of dithiolethiones, but occurred via a covalent reaction with the NF-κB p50 and p65 subunits to inhibit DNA binding. Dithiolethione-mediated inhibition of NF-κB-regulated genes resulted in the inhibition of interleukin (IL)-6, IL-8, urokinase-type plasminogen activator, and VEGF production. ACS-1 also inhibited matrix metalloproteinase-9 activity, cellular migration, and invasion, and ACS-2 reduced tumor burden and resulted in increased tumor host interactions. Together, our findings suggest that dithiolethiones show potential clinical use for estrogen negative breast cancer as a chemotherapeutic or adjuvant therapy.
Collapse
|
40
|
Stewart AM, Desmond D, Kyzar E, Gaikwad S, Roth A, Riehl R, Collins C, Monnig L, Green J, Kalueff AV. Perspectives of zebrafish models of epilepsy: What, how and where next? Brain Res Bull 2012; 87:135-43. [DOI: 10.1016/j.brainresbull.2011.11.020] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 11/20/2011] [Accepted: 11/25/2011] [Indexed: 10/14/2022]
|
41
|
Emerging role of hydrogen sulfide in health and disease: critical appraisal of biomarkers and pharmacological tools. Clin Sci (Lond) 2011; 121:459-88. [PMID: 21843150 DOI: 10.1042/cs20110267] [Citation(s) in RCA: 245] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
H2S (hydrogen sulfide) is a well known and pungent gas recently discovered to be synthesized enzymatically in mammalian and human tissues. In a relatively short period of time, H2S has attracted substantial interest as an endogenous gaseous mediator and potential target for pharmacological manipulation. Studies in animals and humans have shown H2S to be involved in diverse physiological and pathophysiological processes, such as learning and memory, neurodegeneration, regulation of inflammation and blood pressure, and metabolism. However, research is limited by the lack of specific analytical and pharmacological tools which has led to considerable controversy in the literature. Commonly used inhibitors of endogenous H2S synthesis have been well known for decades to interact with other metabolic pathways or even generate NO (nitric oxide). Similarly, commonly used H2S donors release H2S far too quickly to be physiologically relevant, but may have therapeutic applications. In the present review, we discuss the enzymatic synthesis of H2S and its emerging importance as a mediator in physiology and pathology. We also critically discuss the suitability of proposed 'biomarkers' of H2S synthesis and metabolism, and highlight the complexities of the currently used pharmacological H2S 'donor' molecules and 'specific' H2S synthesis inhibitors in their application to studying the role of H2S in human disease.
Collapse
|
42
|
Karén J, Rodriguez A, Friman T, Dencker L, Sundberg C, Scholz B. Effects of the histone deacetylase inhibitor valproic acid on human pericytes in vitro. PLoS One 2011; 6:e24954. [PMID: 21966390 PMCID: PMC3178576 DOI: 10.1371/journal.pone.0024954] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 08/25/2011] [Indexed: 11/18/2022] Open
Abstract
Microvascular pericytes are of key importance in neoformation of blood vessels, in stabilization of newly formed vessels as well as maintenance of angiostasis in resting tissues. Furthermore, pericytes are capable of differentiating into pro-fibrotic collagen type I producing fibroblasts. The present study investigates the effects of the histone deacetylase (HDAC) inhibitor valproic acid (VPA) on pericyte proliferation, cell viability, migration and differentiation. The results show that HDAC inhibition through exposure of pericytes to VPA in vitro causes the inhibition of pericyte proliferation and migration with no effect on cell viability. Pericyte exposure to the potent HDAC inhibitor Trichostatin A caused similar effects on pericyte proliferation, migration and cell viability. HDAC inhibition also inhibited pericyte differentiation into collagen type I producing fibroblasts. Given the importance of pericytes in blood vessel biology a qPCR array focusing on the expression of mRNAs coding for proteins that regulate angiogenesis was performed. The results showed that HDAC inhibition promoted transcription of genes involved in vessel stabilization/maturation in human microvascular pericytes. The present in vitro study demonstrates that VPA influences several aspects of microvascular pericyte biology and suggests an alternative mechanism by which HDAC inhibition affects blood vessels. The results raise the possibility that HDAC inhibition inhibits angiogenesis partly through promoting a pericyte phenotype associated with stabilization/maturation of blood vessels.
Collapse
Affiliation(s)
- Jakob Karén
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Alejandro Rodriguez
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Tomas Friman
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Lennart Dencker
- Department of Pharmaceutical Bioscience, Uppsala University, Uppsala, Sweden
| | - Christian Sundberg
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Department of Women and Children's Health, Uppsala University Hospital, Uppsala, Sweden
- * E-mail:
| | - Birger Scholz
- Department of Pharmaceutical Bioscience, Uppsala University, Uppsala, Sweden
| |
Collapse
|
43
|
Martelli A, Testai L, Breschi MC, Blandizzi C, Virdis A, Taddei S, Calderone V. Hydrogen sulphide: novel opportunity for drug discovery. Med Res Rev 2010; 32:1093-130. [PMID: 23059761 DOI: 10.1002/med.20234] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Hydrogen sulphide (H(2)S) is emerging as an important endogenous modulator, which exhibits the beneficial effects of nitric oxide (NO) on the cardiovascular (CV) system, without producing toxic metabolites. H(2)S is biosynthesized in mammalian tissues by cystathionine-β-synthase and cystathionine-γ-lyase. H(2)S exhibits the antioxidant properties of inorganic and organic sulphites, behaving as a scavenger of reactive oxygen species. There is also clear evidence that H(2)S triggers other important effects, mainly mediated by the activation of ATP-sensitive potassium channels (K(ATP)). This mechanism accounts for the vasorelaxing and cardioprotective effects of H(2)S. Furthermore, H(2)S inhibits smooth muscle proliferation and platelet aggregation. In non-CV systems, H(2)S regulates the functions of the central nervous system, as well as respiratory, gastroenteric, and endocrine systems. Conversely, H(2)S deficiency contributes to the pathogenesis of hypertension. Likewise, impairment of H(2)S biosynthesis is involved in CV complications associated with diabetes mellitus. There is also evidence of a cross-talk between the H(2)S and the endothelial NO pathways. In particular, recent observations indicate a possible pathogenic link between deficiencies of H(2 S activity and the progress of endothelial dysfunction. These biological aspects of endogenous H(2)S have led several authors to look at this mediator as "the new NO" that has given attractive opportunities to develop innovative classes of drugs. In this review, the main biological actions of H(2)S are discussed. Moreover, some examples of H(2)S-donors are shown, as well as some hybrids, in which H(2)S-releasing moieties are added to well-known drugs, for improving their pharmacodynamic profile or reducing the potential for adverse effects, are reported.
Collapse
Affiliation(s)
- Alma Martelli
- Dipartimento di Psichiatria, Neurobiologia, Farmacologia e Biotecnologie, Università di Pisa, Pisa, Italy
| | | | | | | | | | | | | |
Collapse
|
44
|
Valera VA, Li-Ning-T E, Walter BA, Roberts DD, Linehan WM, Merino MJ. Protein expression profiling in the spectrum of renal cell carcinomas. J Cancer 2010; 1:184-96. [PMID: 20975849 PMCID: PMC2962428 DOI: 10.7150/jca.1.184] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 10/14/2010] [Indexed: 11/05/2022] Open
Abstract
In this study, we aimed to evaluate the protein expression profile of a spectrum of renal cell carcinomas (RCC) to find potential biomarkers for disease onset and progression and therefore, prospective therapeutic targets. A 2D-gel based proteomic analysis was used to outline differences in protein levels among different subtypes of renal cell carcinomas, including clear cell carcinomas, papillary lesions, chromophobe tumors and renal oncocytomas. Spot pattern was compared to the corresponding normal kidney from the same patients and distinctive, differentially expressed proteins were characterized by mass spectrometry. Twenty-one protein spots were found differentially expressed between clear cell RCC and normal tissue and 38 spots were found expressed in chromophobe tumors. Eleven proteins were identified, with most differentially expressed -by fold change- between clear cell tumors and the corresponding normal tissue. Two of the identified proteins, Triosephosphate isomerase 1 (TPI-1) and Heat Shock protein 27 (Hsp27), were further validated in a separate set of tumors by immunohistochemistry and expression levels were correlated with clinicopathologic features of the patients. Hsp27 was highly expressed in 82% of the tumors used for validation, and all cases showed strong immunoreactivity for TPI-1. In both Hsp27 and TPI-1, protein expression positively correlated with histologic features of the disease. Our results suggest that the subjacent cytogenetic abnormalities seen in different histological types of RCC are followed by specific changes in protein expression. From these changes, Hsp27 and TPI-1 emerged as potential candidates for the differentiation and prognosis in RCC.
Collapse
Affiliation(s)
- Vladimir A Valera
- 1. Surgical Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, USA
| | - Elsa Li-Ning-T
- 1. Surgical Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, USA
| | - Beatriz A Walter
- 1. Surgical Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, USA
| | - David D. Roberts
- 2. Biochemical Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, USA
| | - W M Linehan
- 3. Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Maria J Merino
- 1. Surgical Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, USA
| |
Collapse
|
45
|
Predmore BL, Lefer DJ. Development of hydrogen sulfide-based therapeutics for cardiovascular disease. J Cardiovasc Transl Res 2010; 3:487-98. [PMID: 20628909 DOI: 10.1007/s12265-010-9201-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 06/21/2010] [Indexed: 10/19/2022]
Abstract
The physiological role of the gaseous signaling molecule hydrogen sulfide (H(2)S) was first realized in the mid-1990s with the work of Abe and Kimura. Since then, it has become evident that this endogenous gas is extremely important in the homeostasis of the cardiovascular system and the pathogenesis of cardiovascular disease. Several biotechnology companies have developed and are developing H(2)S-based therapeutic compounds, and there are ongoing clinical trials investigating the therapeutic potential of H(2)S. Several organic and chemical compounds that are known H(2)S donors have the potential to be developed into effective H(2)S-based therapeutic agents. This review will provide a historical and current perspective on the role(s) of H(2)S in the cardiovascular system and the current state of development and future outlook of H(2)S-based therapies for cardiovascular disease.
Collapse
Affiliation(s)
- Benjamin L Predmore
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, 550 Peachtree Street, NE, Atlanta, GA 30308, USA
| | | |
Collapse
|
46
|
Moody TW, Switzer C, Santana-Flores W, Ridnour LA, Berna M, Thill M, Jensen RT, Sparatore A, Del Soldato P, Yeh GC, Roberts DD, Giaccone G, Wink DA. Dithiolethione modified valproate and diclofenac increase E-cadherin expression and decrease proliferation of non-small cell lung cancer cells. Lung Cancer 2010; 68:154-60. [PMID: 19628293 PMCID: PMC3835159 DOI: 10.1016/j.lungcan.2009.06.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Revised: 06/10/2009] [Accepted: 06/14/2009] [Indexed: 10/20/2022]
Abstract
The effects of dithiolethione modified valproate, diclofenac and sulindac on non-small cell lung cancer (NSCLC) cells were investigated. Sulfur(S)-valproate and S-diclofenac at 1 microg/ml concentrations significantly reduced prostaglandin (PG)E(2) levels in NSCLC cell lines A549 and NCI-H1299 as did the COX-2 inhibitor DuP-697. In vitro, S-valproate, S-diclofenac and S-sulindac half-maximally inhibited the clonal growth of NCI-H1299 cells at 6, 6 and 15 microg/ml, respectively. Using the MTT assay, 10 microg/ml S-valproate, NO-aspirin and Cay10404, a selective COX-2 inhibitor, but not SC-560, a selective COX-1 inhibitor, inhibited the growth of A549 cells. In vivo, 18mg/kg i.p. of S-valproate and S-diclofenac, but not S-sulindac, significantly inhibited A549 or NCI-H1299 xenograft proliferation in nude mice, but had no effect on the nude mouse body weight. The mechanism by which S-valproate and S-diclofenac inhibited the growth of NSCLC cells was investigated. Nitric oxide-aspirin but not S-valproate caused apoptosis of NSCLC cells. By Western blot, S-valproate and S-diclofenac increased E-cadherin but reduced vimentin and ZEB1 (a transcriptional suppressor of E-cadherin) protein expression in NSCLC cells. Because S-valproate and S-diclofenac inhibit the growth of NSCLC cells and reduce PGE(2) levels, they may prove beneficial in the chemoprevention and/or therapy of NSCLC.
Collapse
Affiliation(s)
- Terry W Moody
- National Cancer Institute, Office of the Director, Center for Cancer Research, Bethesda, MD 20892-1500, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Jia Y, Wu SL, Isenberg JS, Dai S, Sipes JM, Field L, Zeng B, Bandle RW, Ridnour LA, Wink DA, Ramchandran R, Karger BL, Roberts DD. Thiolutin inhibits endothelial cell adhesion by perturbing Hsp27 interactions with components of the actin and intermediate filament cytoskeleton. Cell Stress Chaperones 2010; 15:165-81. [PMID: 19579057 PMCID: PMC2866983 DOI: 10.1007/s12192-009-0130-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Revised: 06/12/2009] [Accepted: 06/17/2009] [Indexed: 10/20/2022] Open
Abstract
Thiolutin is a dithiole synthesized by Streptomyces sp. that inhibits endothelial cell adhesion and tumor growth. We show here that thiolutin potently inhibits developmental angiogenesis in zebrafish and vascular outgrowth from tissue explants in 3D cultures. Thiolutin is a potent and selective inhibitor of endothelial cell adhesion accompanied by rapid induction of HSPB1 (Hsp27) phosphorylation. The inhibitory effects of thiolutin on endothelial cell adhesion are transient, potentially due to a compensatory increase in Hsp27 protein levels. Accordingly, heat shock induction of Hsp27 limits the anti-adhesive activity of thiolutin. Thiolutin treatment results in loss of actin stress fibers, increased cortical actin as cells retract, and decreased cellular F-actin. Mass spectrometric analysis of Hsp27 binding partners following immunoaffinity purification identified several regulatory components of the actin cytoskeleton that associate with Hsp27 in a thiolutin-sensitive manner including several components of the Arp2/3 complex. Among these, ArpC1a is a direct binding partner of Hsp27. Thiolutin treatment induces peripheral localization of phosphorylated Hsp27 and Arp2/3. Hsp27 also associates with the intermediate filament components vimentin and nestin. Thiolutin treatment specifically ablates Hsp27 interaction with nestin and collapses nestin filaments. These results provide new mechanistic insights into regulation of cell adhesion and cytoskeletal dynamics by Hsp27.
Collapse
Affiliation(s)
- Yifeng Jia
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - Shiaw-Lin Wu
- Barnett Institute, Northeastern University, Boston, MA 02115 USA
| | - Jeff S. Isenberg
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
- Hemostasis and Vascular Biology Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261 USA
| | - Shujia Dai
- Barnett Institute, Northeastern University, Boston, MA 02115 USA
| | - John M. Sipes
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - Lyndsay Field
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - Bixi Zeng
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - Russell W. Bandle
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - Lisa A. Ridnour
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - David A. Wink
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - Ramani Ramchandran
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
- Department of Pediatrics, Children’s Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - Barry L. Karger
- Barnett Institute, Northeastern University, Boston, MA 02115 USA
| | - David D. Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
- NIH, Building 10 Room 2A33, 10 Center Dr MSC1500, Bethesda, MD 20892-1500 USA
| |
Collapse
|
48
|
Brannen KC, Panzica-Kelly JM, Danberry TL, Augustine-Rauch KA. Development of a zebrafish embryo teratogenicity assay and quantitative prediction model. ACTA ACUST UNITED AC 2010; 89:66-77. [DOI: 10.1002/bdrb.20223] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
49
|
Kothari V, Joshi G, Nama S, Somasundaram K, Mulherkar R. HDAC inhibitor valproic acid enhances tumor cell kill in adenovirus-HSVtk mediated suicide gene therapy in HNSCC xenograft mouse model. Int J Cancer 2010; 126:733-42. [PMID: 19569045 DOI: 10.1002/ijc.24700] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Safety, efficacy and enhanced transgene expression are the primary concerns while using any vector for gene therapy. One of the widely used vectors in clinical trials is adenovirus which provides a safe way to deliver the therapeutic gene. However, adenovirus has poor transduction efficiency in vivo since most tumor cells express low coxsackie and adenovirus receptors. Similarly transgene expression remains low, possibly because of the chromatization of adenoviral genome upon infection in eukaryotic cells, an effect mediated by histone deacetylases (HDACs). Using a recombinant adenovirus (Ad-HSVtk) carrying the herpes simplex thymidine kinase (HSVtk) and GFP genes we demonstrate that HDAC inhibitor valproic acid can bring about an increase in CAR expression on host cells and thereby enhanced Ad-HSVtk infectivity. It also resulted in an increase in transgene (HSVtk and GFP) expression. This, in turn, resulted in increased cell kill of HNSCC cells, following ganciclovir treatment in vitro as well as in vivo in a xenograft nude mouse model.
Collapse
Affiliation(s)
- Vishal Kothari
- Department of Genetic Engineering Unit, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra, India
| | | | | | | | | |
Collapse
|
50
|
Naumov P, Petruševski G, Jovanovski G. Vibrational spectra of calcium, strontium and barium valproates. Self-assembly of valproate nanostrands in aqueous solution and in the solid state. CrystEngComm 2010. [DOI: 10.1039/c000488j] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|