1
|
Samanta S, Noda H, Watanabe T, Cui J, Shibasaki M. Direct Catalytic Asymmetric Conjugate Addition of Benzofuran-3(2H)-Ones to α,β-Unsaturated Thioamides: Stereodivergent Synthesis of Rocaglaol. Angew Chem Int Ed Engl 2025; 64:e202415805. [PMID: 39351614 DOI: 10.1002/anie.202415805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Indexed: 11/10/2024]
Abstract
Rocaglaol, a representative flavagline, has attracted significant attention because of its unique chemical structure and biological activities. This paper reports a mild and scalable copper-catalyzed enantioselective conjugate addition of benzofuran-3(2H)-ones to α,β-unsaturated thioamides. This method allows for the concise synthesis of all possible stereoisomers of a key intermediate of rocaglaol and its derivatives in a highly diastereo- and enantioselective manner using different chiral phosphine ligands. Theoretical insights into the reaction mechanism and the origin of ligand-dependent diastereodivergence were obtained using density functional theory calculations.
Collapse
Affiliation(s)
- Sadhanendu Samanta
- Institute of Microbial Chemistry (BIKAKEN), Tokyo, 3-14-23 Kamiosaki Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Hidetoshi Noda
- Institute of Microbial Chemistry (BIKAKEN), Tokyo, 3-14-23 Kamiosaki Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Takumi Watanabe
- Institute of Microbial Chemistry (BIKAKEN), Tokyo, 3-14-23 Kamiosaki Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Jin Cui
- Institute of Microbial Chemistry (BIKAKEN), Tokyo, 3-14-23 Kamiosaki Shinagawa-ku, Tokyo, 141-0021, Japan
- Center for Innovative Drug Discovery, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Pharmacy, Fudan University, Shanghai, P. R. China
| | - Masakatsu Shibasaki
- Institute of Microbial Chemistry (BIKAKEN), Tokyo, 3-14-23 Kamiosaki Shinagawa-ku, Tokyo, 141-0021, Japan
| |
Collapse
|
2
|
Peron G, Mastinu A, Peña-Corona SI, Hernández-Parra H, Leyva-Gómez G, Calina D, Sharifi-Rad J. Silvestrol, a potent anticancer agent with unfavourable pharmacokinetics: Current knowledge on its pharmacological properties and future directions for the development of novel drugs. Biomed Pharmacother 2024; 177:117047. [PMID: 38959604 DOI: 10.1016/j.biopha.2024.117047] [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: 04/09/2024] [Revised: 06/14/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024] Open
Abstract
Cancer remains a leading cause of death, with increasing incidence. Conventional treatments offer limited efficacy and cause significant side effects, hence novel drugs with improved pharmacological properties and safety are required. Silvestrol (SLV) is a flavagline derived from some plants of the Aglaia genus that has shown potent anticancer effects, warranting further study. Despite its efficacy in inhibiting the growth of several types of cancer cells, SLV is characterized by an unfavorable pharmacokinetics that hamper its use as a drug. A consistent research over the recent years has led to develop novel SLV derivatives with comparable pharmacodynamics and an ameliorated pharmacokinetic profile, demonstrating potential applications in the clinical management of cancer. This comprehensive review aims to highlight the most recent data available on SLV and its synthetic derivatives, addressing their pharmacological profile and therapeutic potential in cancer treatment. A systematic literature review of both in vitro and in vivo studies focusing on anticancer effects, pharmacodynamics, and pharmacokinetics of these compounds is presented. Overall, literature data highlight that rationale chemical modifications of SLV are critical for the development of novel drugs with high efficacy on a broad variety of cancers and improved bioavailability in vivo. Nevertheless, SLV analogues need to be further studied to better understand their mechanisms of action, which can be partially different to SLV. Furthermore, clinical research is still required to assess their efficacy in humans and their safety.
Collapse
Affiliation(s)
- Gregorio Peron
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia 25123, Italy.
| | - Andrea Mastinu
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia 25123, Italy
| | - Sheila I Peña-Corona
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Hector Hernández-Parra
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico; Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova 200349, Romania.
| | - Javad Sharifi-Rad
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea; Centro de Estudios Tenológicos y Universitarios del Golfo, Veracruz, Mexico.
| |
Collapse
|
3
|
Todosenko N, Yurova K, Vulf M, Khaziakhmatova O, Litvinova L. Prohibitions in the meta-inflammatory response: a review. Front Mol Biosci 2024; 11:1322687. [PMID: 38813101 PMCID: PMC11133639 DOI: 10.3389/fmolb.2024.1322687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 05/01/2024] [Indexed: 05/31/2024] Open
Abstract
Prohibitins are the central regulatory element of cellular homeostasis, especially by modulating the response at different levels: Nucleus, mitochondria and membranes. Their localization and interaction with various proteins, homons, transcription and nuclear factors, and mtDNA indicate the globality and complexity of their pleiotropic properties, which remain to be investigated. A more detailed deciphering of cellular metabolism in relation to prohibitins under normal conditions and in various metabolic diseases will allow us to understand the precise role of prohibitins in the signaling cascades of PI3K/Akt, Raf/MAP/ERK, STAT3, p53, and others and to fathom their mutual influence. A valuable research perspective is to investigate the role of prohibitins in the molecular and cellular interactions between the two major players in the pathogenesis of obesity-adipocytes and macrophages - that form the basis of the meta-inflammatory response. Investigating the subtle intercellular communication and molecular cascades triggered in these cells will allow us to propose new therapeutic strategies to eliminate persistent inflammation, taking into account novel molecular genetic approaches to activate/inactivate prohibitins.
Collapse
Affiliation(s)
- Natalia Todosenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Kristina Yurova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Maria Vulf
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Olga Khaziakhmatova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Larisa Litvinova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
- Laboratory of Cellular and Microfluidic Technologies, Siberian State Medical University, Tomsk, Russia
| |
Collapse
|
4
|
Najem A, Krayem M, Sabbah S, Pesetti M, Journe F, Awada A, Désaubry L, Ghanem GE. Targeting Prohibitins to Inhibit Melanoma Growth and Overcome Resistance to Targeted Therapies. Cells 2023; 12:1855. [PMID: 37508519 PMCID: PMC10378173 DOI: 10.3390/cells12141855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/19/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Despite important advances in the treatment of metastatic melanoma with the development of MAPK-targeted agents and immune checkpoint inhibitors, the majority of patients either do not respond to therapies or develop acquired resistance. Furthermore, there is no effective targeted therapy currently available for BRAF wild-type melanomas (approximately 50% of cutaneous melanoma). Thus, there is a compelling need for new efficient targeted therapies. Prohibitins (PHBs) are overexpressed in several types of cancers and implicated in the regulation of signaling networks that promote cell invasion and resistance to cell apoptosis. Herein, we show that PHBs are highly expressed in melanoma and are associated with not only poor survival but also with resistance to BRAFi/MEKi. We designed and identified novel specific PHB inhibitors that can inhibit melanoma cell growth in 3D spheroid models and a large panel of representative cell lines with different molecular subtypes, including those with intrinsic and acquired resistance to MAPKi, by significantly moderating both MAPK (CRAF-ERK axis) and PI3K/AKT pathways, and inducing apoptosis through the mitochondrial pathway and up-regulation of p53. In addition, autophagy inhibition enhances the antitumor efficacy of these PHB ligands. More important, these ligands can act in synergy with MAPKi to more efficiently inhibit cell growth and overcome drug resistance in both BRAF wild-type and mutant melanoma. In conclusion, targeting PHBs represents a very promising therapeutic strategy in melanoma, regardless of mutational status.
Collapse
Affiliation(s)
- Ahmad Najem
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Mohammad Krayem
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Serena Sabbah
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Matilde Pesetti
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Fabrice Journe
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Ahmad Awada
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
- Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Laurent Désaubry
- Center of Research in Biomedicine of Strasbourg, Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, 67000 Strasbourg, France
| | - Ghanem E Ghanem
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
| |
Collapse
|
5
|
Montiel-Dávalos A, Ayala Y, Hernández G. The dark side of mRNA translation and the translation machinery in glioblastoma. Front Cell Dev Biol 2023; 11:1086964. [PMID: 36994107 PMCID: PMC10042294 DOI: 10.3389/fcell.2023.1086964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/03/2023] [Indexed: 03/14/2023] Open
Abstract
Among the different types of cancer affecting the central nervous system (CNS), glioblastoma (GB) is classified by the World Health Organization (WHO) as the most common and aggressive CNS cancer in adults. GB incidence is more frequent among persons aged 45–55 years old. GB treatments are based on tumor resection, radiation, and chemotherapies. The current development of novel molecular biomarkers (MB) has led to a more accurate prediction of GB progression. Moreover, clinical, epidemiological, and experimental studies have established genetic variants consistently associated with the risk of suffering GB. However, despite the advances in these fields, the survival expectancy of GB patients is still shorter than 2 years. Thus, fundamental processes inducing tumor onset and progression remain to be elucidated. In recent years, mRNA translation has been in the spotlight, as its dysregulation is emerging as a key cause of GB. In particular, the initiation phase of translation is most involved in this process. Among the crucial events, the machinery performing this phase undergoes a reconfiguration under the hypoxic conditions in the tumor microenvironment. In addition, ribosomal proteins (RPs) have been reported to play translation-independent roles in GB development. This review focuses on the research elucidating the tight relationship between translation initiation, the translation machinery, and GB. We also summarize the state-of-the-art drugs targeting the translation machinery to improve patients’ survival. Overall, the recent advances in this field are shedding new light on the dark side of translation in GB.
Collapse
|
6
|
dos Santos IV, Borges RS, Silva GM, de Lima LR, Bastos RS, Ramos RS, Silva LB, da Silva CHTP, dos Santos CBR. Hierarchical Virtual Screening Based on Rocaglamide Derivatives to Discover New Potential Anti-Skin Cancer Agents. Front Mol Biosci 2022; 9:836572. [PMID: 35720115 PMCID: PMC9201829 DOI: 10.3389/fmolb.2022.836572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/26/2022] [Indexed: 12/04/2022] Open
Abstract
Skin Cancer (SC) is among the most common type of cancers worldwide. The search for SC therapeutics using molecular modeling strategies as well as considering natural plant-derived products seems to be a promising strategy. The phytochemical Rocaglamide A (Roc-A) and its derivatives rise as an interesting set of reference compounds due to their in vitro cytotoxic activity with SC cell lines. In view of this, we performed a hierarchical virtual screening study considering Roc-A and its derivatives, with the aim to find new chemical entities with potential activity against SC. For this, we selected 15 molecules (Roc-A and 14 derivatives) and initially used them in docking studies to predict their interactions with Checkpoint kinase 1 (Chk1) as a target for SC. This allowed us to compile and use them as a training set to build robust pharmacophore models, validated by Pearson’s correlation (p) values and hierarchical cluster analysis (HCA), subsequentially submitted to prospective virtual screening using the Molport® database. Outputted compounds were then selected considering their similarities to Roc-A, followed by analyses of predicted toxicity and pharmacokinetic properties as well as of consensus molecular docking using three software. 10 promising compounds were selected and analyzed in terms of their properties and structural features and, also, considering their previous reports in literature. In this way, the 10 promising virtual hits found in this work may represent potential anti-SC agents and further investigations concerning their biological tests shall be conducted.
Collapse
Affiliation(s)
- Igor V.F. dos Santos
- Modeling and Computational Chemistry Laboratory, Federal University of Amapá, Macapá, Brazil
- Graduate Program in Biotechnology and Biodiversity-Network BIONORTE, Federal University of Amapá, Macapá, Brazil
| | - Rosivaldo S. Borges
- Modeling and Computational Chemistry Laboratory, Federal University of Amapá, Macapá, Brazil
- Graduate Program in Medicinal Chemistry and Molecular Modeling, Federal University of Pará, Belém, Brazil
| | - Guilherme M. Silva
- Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto - Universidade de São Paulo, Ribeirão Preto, Brazil
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Lúcio R. de Lima
- Modeling and Computational Chemistry Laboratory, Federal University of Amapá, Macapá, Brazil
- Graduate Program in Medicinal Chemistry and Molecular Modeling, Federal University of Pará, Belém, Brazil
| | - Ruan S. Bastos
- Modeling and Computational Chemistry Laboratory, Federal University of Amapá, Macapá, Brazil
- Graduate Program in Medicinal Chemistry and Molecular Modeling, Federal University of Pará, Belém, Brazil
| | - Ryan S. Ramos
- Modeling and Computational Chemistry Laboratory, Federal University of Amapá, Macapá, Brazil
- Graduate Program in Biotechnology and Biodiversity-Network BIONORTE, Federal University of Amapá, Macapá, Brazil
| | - Luciane B. Silva
- Modeling and Computational Chemistry Laboratory, Federal University of Amapá, Macapá, Brazil
- Graduate Program in Medicinal Chemistry and Molecular Modeling, Federal University of Pará, Belém, Brazil
| | - Carlos H. T. P. da Silva
- Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto - Universidade de São Paulo, Ribeirão Preto, Brazil
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Cleydson B. R. dos Santos
- Modeling and Computational Chemistry Laboratory, Federal University of Amapá, Macapá, Brazil
- Graduate Program in Biotechnology and Biodiversity-Network BIONORTE, Federal University of Amapá, Macapá, Brazil
- Graduate Program in Medicinal Chemistry and Molecular Modeling, Federal University of Pará, Belém, Brazil
- *Correspondence: Cleydson B. R. dos Santos,
| |
Collapse
|
7
|
Stereodivergent total synthesis of rocaglaol initiated by synergistic dual-metal-catalyzed asymmetric allylation of benzofuran-3(2H)-one. Chem 2022. [DOI: 10.1016/j.chempr.2022.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
8
|
Abstract
This review deals with the synthesis of naturally occurring alkaloids containing partially or completely saturated pyrimidine nuclei. The interest in these compounds is associated with their structural diversity, high biological activity and toxicity. The review is divided into four parts, each of which describes a number of synthetic methodologies toward structurally different naturally occurring alkaloids containing saturated cyclic six-membered amidine, guanidine, aminal and urea (thiourea) moieties, respectively. The development of various synthetic strategies for the preparation of these compounds has remarkably increased during the past few decades. This is primarily due to the fact that some of these compounds are isolated only in limited quantities, which makes it practically impossible to study their full structural characteristics and biological activity.
Collapse
|
9
|
Antifungal Secondary Metabolites Against Blast Fungus Magnaporthe oryzae. Fungal Biol 2022. [DOI: 10.1007/978-3-031-04805-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
10
|
Greger H. Comparative phytochemistry of flavaglines (= rocaglamides), a group of highly bioactive flavolignans from Aglaia species (Meliaceae). PHYTOCHEMISTRY REVIEWS : PROCEEDINGS OF THE PHYTOCHEMICAL SOCIETY OF EUROPE 2022; 21:725-764. [PMID: 34104125 PMCID: PMC8176878 DOI: 10.1007/s11101-021-09761-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/17/2021] [Indexed: 05/07/2023]
Abstract
Flavaglines are formed by cycloaddition of a flavonoid nucleus with a cinnamic acid moiety representing a typical chemical character of the genus Aglaia of the family Meliaceae. Based on biosynthetic considerations 148 derivatives are grouped together into three skeletal types representing 77 cyclopenta[b]benzofurans, 61 cyclopenta[bc]benzopyrans, and 10 benzo[b]oxepines. Apart from different hydroxy, methoxy, and methylenedioxy groups of the aromatic rings, important structural variation is created by different substitutions and stereochemistries of the central cyclopentane ring. Putrescine-derived bisamides constitute important building blocks occurring as cyclic 2-aminopyrrolidines or in an open-chained form, and are involved in the formation of pyrimidinone flavaglines. Regarding the central role of cinnamic acid in the formation of the basic skeleton, rocagloic acid represents a biosynthetic precursor from which aglafoline- and rocaglamide-type cyclopentabenzofurans can be derived, while those of the rocaglaol-type are the result of decarboxylation. Broad-based comparison revealed characteristic substitution trends which contribute as chemical markers to natural delimitation and grouping of taxonomically problematic Aglaia species. A wide variety of biological activities ranges from insecticidal, antifungal, antiprotozoal, and anti-inflammatory properties, especially to pronounced anticancer and antiviral activities. The high insecticidal activity of flavaglines is comparable with that of the well-known natural insecticide azadirachtin. Comparative feeding experiments informed about structure-activity relationships and exhibited different substitutions of the cyclopentane ring essential for insecticidal activity. Parallel studies on the antiproliferative activity of flavaglines in various tumor cell lines revealed similar structural prerequisites that let expect corresponding molecular mechanisms. An important structural modification with very high cytotoxic potency was found in the benzofuran silvestrol characterized by an unusual dioxanyloxy subunit. It possessed comparable cytotoxicity to that of the natural anticancer compounds paclitaxel (Taxol®) and camptothecin without effecting normal cells. The primary effect was the inhibition of protein synthesis by binding to the translation initiation factor eIF4A, an ATP-dependent DEAD-box RNA helicase. Flavaglines were also shown to bind to prohibitins (PHB) responsible for regulation of important signaling pathways, and to inhibit the transcriptional factor HSF1 deeply involved in metabolic programming, survival, and proliferation of cancer cells. Flavaglines were shown to be not only promising anticancer agents but gained now also high expectations as agents against emerging RNA viruses like SARS-CoV-2. Targeting the helicase eIF4A with flavaglines was recently described as pan-viral strategy for minimizing the impact of future RNA virus pandemics.
Collapse
Affiliation(s)
- Harald Greger
- Chemodiversity Research Group, Faculty of Life Sciences, University of Vienna, Rennweg 14, 1030 Wien, Austria
| |
Collapse
|
11
|
Lu HH, Cao MY. Enantioselective Palladium-Catalyzed Decarboxylative Dearomative Asymmetric Allylic Alkylation of Benzofurans: Diversity-Oriented Synthesis of Flavaglines. Synlett 2021. [DOI: 10.1055/a-1650-4266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
AbstractWith the introduction of new Trost-type bisphosphine ligands bearing a chiral cycloalkane framework, the highly efficient and enantioselective palladium-catalyzed decarboxylative dearomative asymmetric allylic alkylation (AAA) of benzofurans was achieved. This enabled a diversity-oriented synthesis (DOS) of previously unreachable flavaglines, which features two diversification stages. A new avenue for developing flavagline-based drugs was thus established.1 Introduction2 The Dearomative Asymmetric Allylic Alkylation of Benzofurans3 Synthesis of Flavaglines4 Conclusion and Outlook
Collapse
Affiliation(s)
- Hai-Hua Lu
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University
- Institute of Natural Sciences, Westlake Institute for Advanced Study
- Department of Chemistry, Zhejiang University
- Institute of Advanced Synthesis (IAS), Nanjing Tech University
| | - Meng-Yue Cao
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University
- Institute of Natural Sciences, Westlake Institute for Advanced Study
- Department of Chemistry, Zhejiang University
| |
Collapse
|
12
|
Belser M, Walker DW. Role of Prohibitins in Aging and Therapeutic Potential Against Age-Related Diseases. Front Genet 2021; 12:714228. [PMID: 34868199 PMCID: PMC8636131 DOI: 10.3389/fgene.2021.714228] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/21/2021] [Indexed: 12/16/2022] Open
Abstract
A decline in mitochondrial function has long been associated with age-related health decline. Several lines of evidence suggest that interventions that stimulate mitochondrial autophagy (mitophagy) can slow aging and prolong healthy lifespan. Prohibitins (PHB1 and PHB2) assemble at the mitochondrial inner membrane and are critical for mitochondrial homeostasis. In addition, prohibitins (PHBs) have diverse roles in cell and organismal biology. Here, we will discuss the role of PHBs in mitophagy, oxidative phosphorylation, cellular senescence, and apoptosis. We will also discuss the role of PHBs in modulating lifespan. In addition, we will review the links between PHBs and diseases of aging. Finally, we will discuss the emerging concept that PHBs may represent an attractive therapeutic target to counteract aging and age-onset disease.
Collapse
Affiliation(s)
- Misa Belser
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - David W. Walker
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
13
|
Nishida Y, Zhao R, Heese LE, Akiyama H, Patel S, Jaeger AM, Jacamo RO, Kojima K, Ma MCJ, Ruvolo VR, Chachad D, Devine W, Lindquist S, Davis RE, Porco JA, Whitesell L, Andreeff M, Ishizawa J. Inhibition of translation initiation factor eIF4a inactivates heat shock factor 1 (HSF1) and exerts anti-leukemia activity in AML. Leukemia 2021; 35:2469-2481. [PMID: 34127794 PMCID: PMC8764661 DOI: 10.1038/s41375-021-01308-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 04/01/2021] [Accepted: 05/21/2021] [Indexed: 01/31/2023]
Abstract
Eukaryotic initiation factor 4A (eIF4A), the enzymatic core of the eIF4F complex essential for translation initiation, plays a key role in the oncogenic reprogramming of protein synthesis, and thus is a putative therapeutic target in cancer. As important component of its anticancer activity, inhibition of translation initiation can alleviate oncogenic activation of HSF1, a stress-inducible transcription factor that enables cancer cell growth and survival. Here, we show that primary acute myeloid leukemia (AML) cells exhibit the highest transcript levels of eIF4A1 compared to other cancer types. eIF4A inhibition by the potent and specific compound rohinitib (RHT) inactivated HSF1 in these cells, and exerted pronounced in vitro and in vivo anti-leukemia effects against progenitor and leukemia-initiating cells, especially those with FLT3-internal tandem duplication (ITD). In addition to its own anti-leukemic activity, genetic knockdown of HSF1 also sensitized FLT3-mutant AML cells to clinical FLT3 inhibitors, and this synergy was conserved in FLT3 double-mutant cells carrying both ITD and tyrosine kinase domain mutations. Consistently, the combination of RHT and FLT3 inhibitors was highly synergistic in primary FLT3-mutated AML cells. Our results provide a novel therapeutic rationale for co-targeting eIF4A and FLT3 to address the clinical challenge of treating FLT3-mutant AML.
Collapse
Affiliation(s)
- Yuki Nishida
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ran Zhao
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lauren E. Heese
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hiroki Akiyama
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shreya Patel
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alex M. Jaeger
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Rodrigo O. Jacamo
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kensuke Kojima
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Department of Hematology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Man Chun John Ma
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vivian R. Ruvolo
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dhruv Chachad
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William Devine
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, USA
| | - Susan Lindquist
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - R. Eric Davis
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John A. Porco
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, USA
| | - Luke Whitesell
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA,Present address: Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Michael Andreeff
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jo Ishizawa
- Department of Leukemia, Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
14
|
Chakraborty M, Mahmud NU, Ullah C, Rahman M, Islam T. Biological and biorational management of blast diseases in cereals caused by Magnaporthe oryzae. Crit Rev Biotechnol 2021; 41:994-1022. [PMID: 34006149 DOI: 10.1080/07388551.2021.1898325] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Blast diseases, caused by the fungal pathogen Magnaporthe oryzae, are among the most destructive diseases that occur on at least 50 species of grasses, including cultivated cereals wheat, and rice. Although fungicidal control of blast diseases has widely been researched, development of resistance of the pathogen against commercially available products makes this approach unreliable. Novel approaches such as the application of biopesticides against the blast fungus are needed for sustainable management of this economically important disease. Antagonistic microorganisms, such as fungi and probiotic bacteria from diverse taxonomic genera were found to suppress blast fungi both in vitro and in vivo. Various classes of secondary metabolites, such as alkaloids, phenolics, and terpenoids of plant and microbial origin significantly inhibit fungal growth and may also be effective in managing blast diseases. Common modes of action of microbial biocontrol agents include: antibiosis, production of lytic enzymes, induction of systemic resistance in host plant, and competition for nutrients or space. However, the precise mechanism of biocontrol of the blast fungus by antagonistic microorganisms and/or their bioactive secondary metabolites is not well understood. Commercial formulations of biocontrol agents and bioactive natural products could be cost-effective and sustainable but their availability at this time is extremely limited. This review updates our knowledge on the infection pathway of the wheat blast fungus, catalogs naturally occurring biocontrol agents that may be effective against blast diseases, and discusses their role in sustainable management of the disease.
Collapse
Affiliation(s)
- Moutoshi Chakraborty
- Institute of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh
| | - Nur Uddin Mahmud
- Institute of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh
| | - Chhana Ullah
- Department of Biochemistry, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Mahfuzur Rahman
- WVU Extension Service, West Virginia University, Morgantown, WV, USA
| | - Tofazzal Islam
- Institute of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh
| |
Collapse
|
15
|
Désaubry L, Abou-Hamdan H. Unexpected Inversion of Configuration During the Carbamoylation of 1-Azaflavaglines. Synlett 2020. [DOI: 10.1055/s-0040-1707277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AbstractThe acylation of 8-demethoxy-1-azaflavaglines by dimethylcarbamoyl chloride was found to operate with an inversion of configuration, which is rationalized by the occurrence of styrylurea intermediate. The configuration-reversed products were not observed when the substrate was substituted by a methoxy in position 8, suggesting that an overstabilization of the carbocationic intermediate prevents this reaction to take place.
Collapse
|
16
|
Alula KM, Delgado-Deida Y, Jackson DN, Venuprasad K, Theiss AL. Nuclear partitioning of Prohibitin 1 inhibits Wnt/β-catenin-dependent intestinal tumorigenesis. Oncogene 2020; 40:369-383. [PMID: 33144683 PMCID: PMC7856018 DOI: 10.1038/s41388-020-01538-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 10/09/2020] [Accepted: 10/21/2020] [Indexed: 12/19/2022]
Abstract
The Wnt/β-catenin signaling pathway is aberrantly activated in the majority of colorectal cancer cases due to somatic mutations in the adenomatous polyposis coli (APC) gene. Prohibitin 1 (PHB1) serves pleiotropic cellular functions with dynamic subcellular trafficking facilitating signaling crosstalk between organelles. Nuclear-localized PHB1 is an important regulator of gene transcription. Using mice with inducible intestinal epithelial cell (IEC)-specific deletion of Phb1 (Phb1iΔIEC) and mice with IEC-specific overexpression of Phb1 (Phb1Tg), we demonstrate that IEC-specific PHB1 combats intestinal tumorigenesis in the ApcMin/+ mouse model by inhibiting Wnt/β-catenin signaling. Forced nuclear accumulation of PHB1 in human RKO or SW48 CRC cell lines increased AXIN1 expression and decreased cell viability. PHB1 deficiency in CRC cells decreased AXIN1 expression and increased β-catenin activation that was abolished by XAV939, a pharmacological AXIN stabilizer. These results define a role of PHB1 in inhibiting the Wnt/β-catenin pathway to influence the development of intestinal tumorigenesis. Induction of nuclear PHB1 trafficking provides a novel therapeutic option to influence AXIN1 expression and the β-catenin destruction complex in Wnt-driven intestinal tumorigenesis.
Collapse
Affiliation(s)
- Kibrom M Alula
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Yaritza Delgado-Deida
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Dakota N Jackson
- Department of Internal Medicine, Division of Gastroenterology, Baylor Scott & White Research Institute, Baylor University Medical Center, Dallas, TX, USA
| | - K Venuprasad
- University of Texas Southwestern Medical Center, College of Medicine, Dallas, TX, USA
| | - Arianne L Theiss
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado, Aurora, CO, USA.
| |
Collapse
|
17
|
Tabti R, Lamoureux F, Charrier C, Ory B, Heymann D, Bentouhami E, Désaubry L. Development of prohibitin ligands against osteoporosis. Eur J Med Chem 2020; 210:112961. [PMID: 33129591 DOI: 10.1016/j.ejmech.2020.112961] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/17/2020] [Accepted: 10/17/2020] [Indexed: 10/23/2022]
Abstract
Current therapeutic approaches to osteoporosis display some potential adverse effects and a limited efficacy on non-vertebral fracture reduction. Some sulfonylamidines targeting the scaffold proteins prohibitins-1 and 2 (PHB1/2) have been showed to inhibit the formation of osteoclasts in charge of bone resorption. Herein, we report the development of a second generation of anti-osteoclastic PHB ligands. The most potent compound, IN45, showed 88% inhibition at the low concentration of 5 μM, indicates that it might serve as a basis for the development of new antiosteoporotic drugs.
Collapse
Affiliation(s)
- Redouane Tabti
- Laboratory of Regenerative Nanomedicine (RNM), INSERM U 1260, CRBS, Rue Eugène Boeckel, 67000, Strasbourg, France; LCIMN Laboratory, Faculty of Technology, University Ferhat Abbas, Sétif, Algeria
| | - François Lamoureux
- INSERM, UMR 1238, Nantes, France; Université de Nantes, Nantes Atlantique Universités, Laboratoire « Sarcome Osseux et Remodelage des Tissus Osseux Calcifiés », Faculté de Médecine, Nantes, France
| | - Céline Charrier
- INSERM, UMR 1238, Nantes, France; Université de Nantes, Nantes Atlantique Universités, Laboratoire « Sarcome Osseux et Remodelage des Tissus Osseux Calcifiés », Faculté de Médecine, Nantes, France
| | - Benjamin Ory
- INSERM, UMR 1238, Nantes, France; Université de Nantes, Nantes Atlantique Universités, Laboratoire « Sarcome Osseux et Remodelage des Tissus Osseux Calcifiés », Faculté de Médecine, Nantes, France
| | - Dominique Heymann
- Institut de Cancérologie de L'Ouest, Site René Gauducheau, Boulevard Professeur Jacques Monod, Saint-Herblain, France; Université de Nantes, Faculty of Medicine, Nantes, France; University of Sheffield, Dept of Oncology and Metabolism, Sheffield, UK
| | - Embarek Bentouhami
- LCIMN Laboratory, Faculty of Technology, University Ferhat Abbas, Sétif, Algeria
| | - Laurent Désaubry
- Laboratory of Regenerative Nanomedicine (RNM), INSERM U 1260, CRBS, Rue Eugène Boeckel, 67000, Strasbourg, France; Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
| |
Collapse
|
18
|
Harmouch E, Seitlinger J, Chaddad H, Ubeaud-Sequier G, Barths J, Saidu S, Désaubry L, Grandemange S, Massfelder T, Fuhrmann G, Fioretti F, Dontenwill M, Benkirane-Jessel N, Idoux-Gillet Y. Flavagline synthetic derivative induces senescence in glioblastoma cancer cells without being toxic to healthy astrocytes. Sci Rep 2020; 10:13750. [PMID: 32792639 PMCID: PMC7426813 DOI: 10.1038/s41598-020-70820-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/30/2020] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive types of cancer, which begins within the brain. It is the most invasive type of glioma developed from astrocytes. Until today, Temozolomide (TMZ) is the only standard chemotherapy for patients with GBM. Even though chemotherapy extends the survival of patients, there are many undesirable side effects, and most cases show resistance to TMZ. FL3 is a synthetic flavagline which displays potent anticancer activities, and is known to inhibit cell proliferation, by provoking cell cycle arrest, and leads to apoptosis in a lot of cancer cell lines. However, the effect of FL3 in glioblastoma cancer cells has not yet been examined. Hypoxia is a major problem for patients with GBM, resulting in tumor resistance and aggressiveness. In this study, we explore the effect of FL3 in glioblastoma cells under normoxia and hypoxia conditions. Our results clearly indicate that this synthetic flavagline inhibits cell proliferation and induced senescence in glioblastoma cells cultured under both conditions. In addition, FL3 treatment had no effect on human brain astrocytes. These findings support the notion that the FL3 molecule could be used in combination with other chemotherapeutic agents or other therapies in glioblastoma treatments.
Collapse
Affiliation(s)
- Ezeddine Harmouch
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 11 Rue Humann, 67000, Strasbourg, France
- Faculté de Chirurgie Dentaire, Université de Strasbourg, 67000, Strasbourg, France
| | - Joseph Seitlinger
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 11 Rue Humann, 67000, Strasbourg, France
- Faculté de Chirurgie Dentaire, Université de Strasbourg, 67000, Strasbourg, France
- Hôpitaux Universitaire de Strasbourg (HUS), 67000, Strasbourg, France
| | - Hassan Chaddad
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 11 Rue Humann, 67000, Strasbourg, France
- Faculté de Chirurgie Dentaire, Université de Strasbourg, 67000, Strasbourg, France
| | - Geneviève Ubeaud-Sequier
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 11 Rue Humann, 67000, Strasbourg, France
- Faculté de Chirurgie Dentaire, Université de Strasbourg, 67000, Strasbourg, France
| | - Jochen Barths
- Core Facility for Flow Cytometry, Cell Sorting and EliSpot, UMR 1110, INSERM, Strasbourg, France
| | - Sani Saidu
- CNRS UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Strasbourg, France
| | - Laurent Désaubry
- Laboratory of Cardio-Oncology and Medicinal Chemistry (FRE 2033), CNRS, Institut Le Bel, Strasbourg, France
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Stéphanie Grandemange
- CNRS, UMR 7039 CRAN, Université de Lorraine, Campus Sciences, 30 bvd des Aiguillettes, 54505, Vandoeuvre les Nancy Cedex, France
| | - Thierry Massfelder
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 11 Rue Humann, 67000, Strasbourg, France
- Faculté de Chirurgie Dentaire, Université de Strasbourg, 67000, Strasbourg, France
| | - Guy Fuhrmann
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 11 Rue Humann, 67000, Strasbourg, France
- Faculté de Chirurgie Dentaire, Université de Strasbourg, 67000, Strasbourg, France
| | - Florence Fioretti
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 11 Rue Humann, 67000, Strasbourg, France
- Faculté de Chirurgie Dentaire, Université de Strasbourg, 67000, Strasbourg, France
- Hôpitaux Universitaire de Strasbourg (HUS), 67000, Strasbourg, France
| | - Monique Dontenwill
- CNRS UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Strasbourg, France
| | - Nadia Benkirane-Jessel
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 11 Rue Humann, 67000, Strasbourg, France.
- Faculté de Chirurgie Dentaire, Université de Strasbourg, 67000, Strasbourg, France.
| | - Ysia Idoux-Gillet
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 11 Rue Humann, 67000, Strasbourg, France.
- Faculté de Chirurgie Dentaire, Université de Strasbourg, 67000, Strasbourg, France.
| |
Collapse
|
19
|
Nilewski C, Michels TD, Xiang AX, Packard GK, Sprengeler PA, Eam B, Fish S, Thompson PA, Wegerski CJ, Ernst JT, Reich SH. Strategic Diastereoselective C1 Functionalization in the Aza-Rocaglamide Scaffold toward Natural Product-Inspired eIF4A Inhibitors. Org Lett 2020; 22:6257-6261. [DOI: 10.1021/acs.orglett.0c01944] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Christian Nilewski
- eFFECTOR Therapeutics, 11180 Roselle Street, Suite A, San Diego, California 92121, United States
| | - Theodore D. Michels
- eFFECTOR Therapeutics, 11180 Roselle Street, Suite A, San Diego, California 92121, United States
| | - Alan X. Xiang
- eFFECTOR Therapeutics, 11180 Roselle Street, Suite A, San Diego, California 92121, United States
| | - Garrick K. Packard
- eFFECTOR Therapeutics, 11180 Roselle Street, Suite A, San Diego, California 92121, United States
| | - Paul A. Sprengeler
- eFFECTOR Therapeutics, 11180 Roselle Street, Suite A, San Diego, California 92121, United States
| | - Boreth Eam
- eFFECTOR Therapeutics, 11180 Roselle Street, Suite A, San Diego, California 92121, United States
| | - Sarah Fish
- eFFECTOR Therapeutics, 11180 Roselle Street, Suite A, San Diego, California 92121, United States
| | - Peggy A. Thompson
- eFFECTOR Therapeutics, 11180 Roselle Street, Suite A, San Diego, California 92121, United States
| | - Christopher J. Wegerski
- eFFECTOR Therapeutics, 11180 Roselle Street, Suite A, San Diego, California 92121, United States
| | - Justin T. Ernst
- eFFECTOR Therapeutics, 11180 Roselle Street, Suite A, San Diego, California 92121, United States
| | - Siegfried H. Reich
- eFFECTOR Therapeutics, 11180 Roselle Street, Suite A, San Diego, California 92121, United States
| |
Collapse
|
20
|
Jackson DN, Alula KM, Delgado-Deida Y, Tabti R, Turner K, Wang X, Venuprasad K, Souza RF, Désaubry L, Theiss AL. The Synthetic Small Molecule FL3 Combats Intestinal Tumorigenesis via Axin1-Mediated Inhibition of Wnt/β-Catenin Signaling. Cancer Res 2020; 80:3519-3529. [PMID: 32665357 DOI: 10.1158/0008-5472.can-20-0216] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/23/2020] [Accepted: 07/09/2020] [Indexed: 12/17/2022]
Abstract
Colorectal cancer exhibits aberrant activation of Wnt/β-catenin signaling. Many inhibitors of the Wnt/β-catenin pathway have been tested for Wnt-dependent cancers including colorectal cancer, but are unsuccessful due to severe adverse reactions. FL3 is a synthetic derivative of natural products called flavaglines, which exhibit anti-inflammatory and cytoprotective properties in intestinal epithelial cells, but has not been previously tested in cell or preclinical models of intestinal tumorigenesis. In vitro studies suggest that flavaglines target prohibitin 1 (PHB1) as a ligand, but this has not been established in the intestine. PHB1 is a highly conserved protein with diverse functions that depend on its posttranslational modifications and subcellular localization. Here, we demonstrate that FL3 combats intestinal tumorigenesis in the azoxymethane-dextran sodium sulfate and ApcMin/+ mouse models and in human colorectal cancer tumor organoids (tumoroids) by inhibiting Wnt/β-catenin signaling via induction of Axin1 expression. FL3 exhibited no change in cell viability in normal intestinal epithelial cells or human matched-normal colonoids. FL3 response was diminished in colorectal cancer cell lines and human colorectal cancer tumoroids harboring a mutation at S45 of β-catenin. PHB1 deficiency in mice or in human colorectal cancer tumoroids abolished FL3-induced expression of Axin1 and drove tumoroid death. In colorectal cancer cells, FL3 treatment blocked phosphorylation of PHB1 at Thr258, resulting in its nuclear translocation and binding to the Axin1 promoter. These results suggest that FL3 inhibits Wnt/β-catenin signaling via PHB1-dependent activation of Axin1. FL3, therefore, represents a novel compound that combats Wnt pathway-dependent cancers, such as colorectal cancer. SIGNIFICANCE: Targeting of PHB1 by FL3 provides a novel mechanism to combat Wnt-driven cancers, with limited intestinal toxicity. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/17/3519/F1.large.jpg.
Collapse
Affiliation(s)
- Dakota N Jackson
- Division of Gastroenterology, Department of Internal Medicine, Baylor Scott & White Research Institute, Baylor University Medical Center, Dallas, Texas
| | - Kibrom M Alula
- Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, Colorado
| | - Yaritza Delgado-Deida
- Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, Colorado
| | - Redouane Tabti
- Laboratory of Regenerative Nanomedicine (UMR 1260), INSERM-University of Strasbourg, Strasbourg, France
| | - Kevin Turner
- University of Texas Southwestern Medical Center, College of Medicine, Dallas, Texas
| | - Xuan Wang
- Division of Gastroenterology, Department of Internal Medicine, Baylor Scott & White Research Institute, Baylor University Medical Center, Dallas, Texas
| | - K Venuprasad
- University of Texas Southwestern Medical Center, College of Medicine, Dallas, Texas
| | - Rhonda F Souza
- Division of Gastroenterology, Department of Internal Medicine, Baylor Scott & White Research Institute, Baylor University Medical Center, Dallas, Texas
| | - Laurent Désaubry
- Laboratory of Regenerative Nanomedicine (UMR 1260), INSERM-University of Strasbourg, Strasbourg, France
| | - Arianne L Theiss
- Division of Gastroenterology, Department of Internal Medicine, Baylor Scott & White Research Institute, Baylor University Medical Center, Dallas, Texas. .,Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
21
|
Cao MY, Ma BJ, Lao ZQ, Wang H, Wang J, Liu J, Xing K, Huang YH, Gan KJ, Gao W, Wang H, Hong X, Lu HH. Optically Active Flavaglines-Inspired Molecules by a Palladium-Catalyzed Decarboxylative Dearomative Asymmetric Allylic Alkylation. J Am Chem Soc 2020; 142:12039-12045. [PMID: 32584568 DOI: 10.1021/jacs.0c05113] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
With the aid of a class of newly discovered Trost-type bisphosphine ligands bearing a chiral cycloalkane framework, the Pd-catalyzed decarboxylative dearomative asymmetric allylic alkylation (AAA) of benzofurans was achieved with high efficiency [0.2-1.0 mol% Pd2(dba)3/L], good generality, and high enantioselectivity (>30 examples, 82-99% yield and 90-96% ee). Moreover, a diversity-oriented synthesis (DOS) of previously unreachable flavaglines is disclosed. It features a reliable and scalable sequence of the freshly developed Tsuji-Trost-Stoltz AAA, a Wacker-Grubbs-Stoltz oxidation, an intra-benzoin condensation, and a conjugate addition, which allows the efficient construction of the challenging and compact cyclopenta[b]benzofuran scaffold with contiguous stereocenters. This strategy offers a new avenue for developing flavagline-based drugs.
Collapse
Affiliation(s)
- Meng-Yue Cao
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, 18 Shilongshan Road, Hangzhou 310024, China.,Institute of Natural Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China.,Department of Chemistry, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Bin-Jie Ma
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, 18 Shilongshan Road, Hangzhou 310024, China.,Institute of Natural Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Zhi-Qi Lao
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, 18 Shilongshan Road, Hangzhou 310024, China.,Institute of Natural Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Hongliang Wang
- Department of Chemistry, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Jing Wang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, 18 Shilongshan Road, Hangzhou 310024, China.,Institute of Natural Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Juan Liu
- Institute of Advanced Synthesis (IAS), Nanjing Tech University, 30 South Puzhu Road, Nanjing 211816, China
| | - Kuan Xing
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, 18 Shilongshan Road, Hangzhou 310024, China.,Institute of Natural Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Yu-Hao Huang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, 18 Shilongshan Road, Hangzhou 310024, China.,Institute of Natural Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Kang-Ji Gan
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, 18 Shilongshan Road, Hangzhou 310024, China.,Institute of Natural Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China.,Department of Chemistry, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Wei Gao
- Institute of Advanced Synthesis (IAS), Nanjing Tech University, 30 South Puzhu Road, Nanjing 211816, China
| | - Huaimin Wang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, 18 Shilongshan Road, Hangzhou 310024, China.,Institute of Natural Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Xin Hong
- Department of Chemistry, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Hai-Hua Lu
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, 18 Shilongshan Road, Hangzhou 310024, China.,Institute of Natural Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China.,Institute of Advanced Synthesis (IAS), Nanjing Tech University, 30 South Puzhu Road, Nanjing 211816, China.,Department of Chemistry, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| |
Collapse
|
22
|
Jin X, Xie J, Zabolocki M, Wang X, Jiang T, Wang D, Désaubry L, Bardy C, Proud CG. The prohibitin-binding compound fluorizoline affects multiple components of the translational machinery and inhibits protein synthesis. J Biol Chem 2020; 295:9855-9867. [PMID: 32430400 DOI: 10.1074/jbc.ra120.012979] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/12/2020] [Indexed: 01/12/2023] Open
Abstract
Fluorizoline (FLZ) binds to prohibitin-1 and -2 (PHB1/2), which are pleiotropic scaffold proteins known to affect signaling pathways involved in several intracellular processes. However, it is not yet clear how FLZ exerts its effect. Here, we show that exposure of three different human cancer cell lines to FLZ increases the phosphorylation of key translation factors, particularly of initiation factor 2 (eIF2) and elongation factor 2 (eEF2), modifications that inhibit their activities. FLZ also impaired signaling through mTOR complex 1, which also regulates the translational machinery, e.g. through the eIF4E-binding protein 4E-BP1. In line with these findings, FLZ potently inhibited protein synthesis. We noted that the first phase of this inhibition involves very rapid eEF2 phosphorylation, which is catalyzed by a dedicated Ca2+-dependent protein kinase, eEF2 kinase (eEF2K). We also demonstrate that FLZ induces a swift and marked rise in intracellular Ca2+ levels, likely explaining the effects on eEF2. Disruption of normal Ca2+ homeostasis can also induce endoplasmic reticulum stress, and our results suggest that induction of this stress response contributes to the increased phosphorylation of eIF2, likely because of activation of the eIF2-modifying kinase PKR-like endoplasmic reticulum kinase (PERK). We show that FLZ induces cancer cell death and that this effect involves contributions from the phosphorylation of both eEF2 and eIF2. Our findings provide important new insights into the biological effects of FLZ and thus the roles of PHBs, specifically in regulating Ca2+ levels, cellular protein synthesis, and cell survival.
Collapse
Affiliation(s)
- Xin Jin
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia.,Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology and School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Jianling Xie
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia
| | - Michael Zabolocki
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia.,Laboratory for Human Neurophysiology and Genetics, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Xuemin Wang
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia.,School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Tao Jiang
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology and School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Dong Wang
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Laurent Désaubry
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.,Laboratory of Medicinal Chemistry and Cardio-oncology, CNRS, Strasbourg, France
| | - Cedric Bardy
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia.,Laboratory for Human Neurophysiology and Genetics, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Christopher G Proud
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia .,School of Biological Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
23
|
Nijhawans P, Behl T, Bhardwaj S. Angiogenesis in obesity. Biomed Pharmacother 2020; 126:110103. [PMID: 32200253 DOI: 10.1016/j.biopha.2020.110103] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/03/2020] [Accepted: 03/11/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Angiogenesis is considered as a major progenitor in the progression of obesity. The current manuscript enumerates the extrinsic role of angiogenesis in obesity. RESULT High caloric diet and lack of physical exercise are the most common causes of obesity and related metabolic conditions. A grossly elevated levels of fat in adipose tissue escalate certain complications which further worsen the state of obesity. Enlargement of white adipose tissue (WAT), deposition of fat mass, proliferation of endothelial cells, production of inflammatory cytokines induces the formation of denovo capillaries from parent microvasculature. Also, several intracellular signaling pathways precipitate obesity. Though, angiostatic molecules (endostatin, angiostatin and TNP-470) have been designed to combat obesity and associated complications. CONCLUSION Adipose tissue trigger growth of blood capillaries, and in turn adipose tissue endothelial cells promote pre-adipocyte proliferation. Modulation of angiogenesis and treatment with angiostatic substances may have the potential to impair the progression of obesity.
Collapse
Affiliation(s)
- Priya Nijhawans
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | | |
Collapse
|
24
|
Elderwish S, Audebrand A, Nebigil CG, Désaubry L. Discovery of 3,3'-pyrrolidinyl-spirooxindoles as cardioprotectant prohibitin ligands. Eur J Med Chem 2019; 186:111859. [PMID: 31735574 DOI: 10.1016/j.ejmech.2019.111859] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 01/08/2023]
Abstract
The scaffold proteins prohibitins-1 and 2 (PHB1/2) play many important roles in coordinating many cell signaling pathways and represent emerging targets in cardiology and oncology. We previously reported that a family of natural products derivatives, flavaglines, binds to PHB1/2 to exert cardioprotectant and anti-cancer effects. However, flavaglines also target the initiation factor of translation eIF4A, which doesn't contribute to cardioprotection and may even induce some adverse effects. Herein, we report the development of a convenient and robust synthesis of the new PHB2 ligand 2'-phenylpyrrolidinyl-spirooxindole, and its analogues. We discovered that these compounds displays cardioprotective effect against doxorubicin mediated cardiotoxicity and uncovered the structural requirement for this activity. We identified in particular some analogues that are more cardioprotectant than flavaglines. Pull-down experiments demonstrated that these compounds bind not only to PHB2 but also PHB1. These novel PHB ligands may provide the basis for the development of new drugs candidates to protect the heart against the adverse effects of anticancer treatments.
Collapse
Affiliation(s)
- Sabria Elderwish
- Laboratory of Medicinal Chemistry and Cardio-oncology, CNRS, 4 rue Blaise Pascal, 67081, Strasbourg, France
| | - Anaïs Audebrand
- Laboratory of Medicinal Chemistry and Cardio-oncology, CNRS, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Canan G Nebigil
- Laboratory of Medicinal Chemistry and Cardio-oncology, CNRS, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Laurent Désaubry
- Laboratory of Medicinal Chemistry and Cardio-oncology, CNRS, 4 rue Blaise Pascal, 67081, Strasbourg, France.
| |
Collapse
|
25
|
Bentayeb H, Aitamer M, Petit B, Dubanet L, Elderwish S, Désaubry L, de Gramont A, Raymond E, Olivrie A, Abraham J, Jauberteau MO, Troutaud D. Prohibitin (PHB) expression is associated with aggressiveness in DLBCL and flavagline-mediated inhibition of cytoplasmic PHB functions induces anti-tumor effects. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:450. [PMID: 31684984 PMCID: PMC6830009 DOI: 10.1186/s13046-019-1440-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/04/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Diffuse large B-cell lymphomas (DLBCLs) are aggressive lymphomas accounting for approximately a third of non-Hodgkin lymphomas. Prohibitin 1 (PHB1) and prohibitin 2 (PHB2) are scaffold proteins that promote mitochondria homeostasis and consequently cell survival, but biological functions of cytoplasmic PHBs remain largely unknown in DLBCL. METHODS PHB expression was analyzed in 82 DLBCL biopsies and five DLBCL cell lines by immunohistochemistry (IHC) and Western blotting. Pharmacological inhibition of PHB using the synthetic flavagline FL3 was realized in vitro to gain insight PHB cellular functions. Effects of FL3 on DLBCL cell line viability, apoptosis, C-Raf-ERK-MNK-eIF4E signaling pathway and eIF4F complex formation and activity were evaluated by XTT assay, annexin V-FITC/PI dual staining and Western blotting respectively. Subcutaneous DLBCL xenograft model in SCID mice was also performed to determine in vivo FL3 effect. RESULTS As in DLBCL cell lines, PHB1 and PHB2 were expressed in germinal center B-cell-like (GCB) and activated B-cell-like (ABC) subtypes. In patient samples, high PHB levels were associated with higher serum LDH (PHB1 and PHB2), IPIaa (PHB2), and Ki-67 (PHB2) expression. Higher PHB1 expression tends to be associated with shorter event-free survival (EFS) in patients, especially in male patients. FL3 induced apoptosis of DLBCL cell lines that was associated with inhibition of the ERK-MNK-eIF4E signaling pathway, including aggressive double/triple-hit DLBCL cell lines. This resulted in altered eIF4F complex formation and activity leading to a reduction of Bcl-2 and c-Myc expression levels. Moreover, FL3 strongly downregulated DLBCL cellular levels of Akt protein and AKT mRNA. FL3 antitumor activity was also confirmed in vivo in a murine xenograft model. CONCLUSION Our data indicate that PHB overexpression is associated with markers of tumor aggressiveness in DLBCL, and that targeting PHBs may be a therapeutic option, notably in aggressive subtypes.
Collapse
Affiliation(s)
| | | | - Barbara Petit
- Laboratoire d'Anatomie-Pathologique, CHU de Limoges, Limoges, France
| | | | | | - Laurent Désaubry
- UMR 7203, CNRS - Université Paris Sorbonne, Paris, France.,Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | | | - Eric Raymond
- AFR Oncology, 1 place Paul Verlaine, Boulogne-Billancourt, France.,Groupe Hospitalier Saint-Joseph, Paris, France
| | - Agnès Olivrie
- Service d'Hématologie Clinique, CHU de Limoges, Limoges, France
| | - Julie Abraham
- Service d'Hématologie Clinique, CHU de Limoges, Limoges, France
| | - Marie-Odile Jauberteau
- EA3842, Université de Limoges, Limoges, France.,Service d'Immunologie, CHU Limoges, Limoges, France
| | | |
Collapse
|
26
|
The synthetic flavagline FL3 spares normal human skin cells from its cytotoxic effect via an activation of Bad. Toxicol In Vitro 2019; 60:27-35. [DOI: 10.1016/j.tiv.2019.04.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/08/2019] [Accepted: 04/23/2019] [Indexed: 12/18/2022]
|
27
|
Ortiz MV, Ahmed S, Burns M, Henssen AG, Hollmann TJ, MacArthur I, Gunasekera S, Gaewsky L, Bradwin G, Ryan J, Letai A, He Y, Naranjo A, Chi YY, LaQuaglia M, Heaton T, Cifani P, Dome JS, Gadd S, Perlman E, Mullen E, Steen H, Kentsis A. Prohibitin is a prognostic marker and therapeutic target to block chemotherapy resistance in Wilms' tumor. JCI Insight 2019; 4:127098. [PMID: 31391345 DOI: 10.1172/jci.insight.127098] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 07/09/2019] [Indexed: 01/05/2023] Open
Abstract
Wilms' tumor is the most common type of childhood kidney cancer. To improve risk stratification and identify novel therapeutic targets for patients with Wilms' tumor, we used high-resolution mass spectrometry proteomics to identify urine tumor markers associated with Wilms' tumor relapse. We determined the urine proteomes at diagnosis of 49 patients with Wilms' tumor, non-Wilms' tumor renal tumors, and age-matched controls, leading to the quantitation of 6520 urine proteins. Supervised analysis revealed specific urine markers of renal rhabdoid tumors, kidney clear cell sarcomas, renal cell carcinomas as well as those detected in patients with cured and relapsed Wilms' tumor. In particular, urine prohibitin was significantly elevated at diagnosis in patients with relapsed as compared with cured Wilms' tumor. In a validation cohort of 139 patients, a specific urine prohibitin ELISA demonstrated that prohibitin concentrations greater than 998 ng/mL at diagnosis were significantly associated with ultimate Wilms' tumor relapse. Immunohistochemical analysis revealed that prohibitin was highly expressed in primary Wilms' tumor specimens and associated with disease stage. Using functional genetic experiments, we found that prohibitin was required for the growth and survival of Wilms' tumor cells. Overexpression of prohibitin was sufficient to block intrinsic mitochondrial apoptosis and to cause resistance to diverse chemotherapy drugs, at least in part by dysregulating factors that control apoptotic cytochrome c release from mitochondrial cristae. Thus, urine prohibitin may improve therapy stratification, noninvasive monitoring of treatment response, and early disease detection. In addition, therapeutic targeting of chemotherapy resistance induced by prohibitin dysregulation may offer improved therapies for patients with Wilms' and other relapsed or refractory tumors.
Collapse
Affiliation(s)
- Michael V Ortiz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Saima Ahmed
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Melissa Burns
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Anton G Henssen
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Travis J Hollmann
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ian MacArthur
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Shehana Gunasekera
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Lyvia Gaewsky
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Gary Bradwin
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Jeremy Ryan
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Anthony Letai
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Ying He
- Children's Oncology Group Statistics and Data Center, Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Arlene Naranjo
- Children's Oncology Group Statistics and Data Center, Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Yueh-Yun Chi
- Children's Oncology Group Statistics and Data Center, Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Michael LaQuaglia
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Todd Heaton
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Paolo Cifani
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jeffrey S Dome
- Center for Cancer and Blood Disorders, Children's National Health System, Washington, DC, USA
| | - Samantha Gadd
- Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Elizabeth Perlman
- Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | | | - Hanno Steen
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Alex Kentsis
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Departments of Pediatrics, Pharmacology, and Physiology & Biophysics, Weill Cornell Medical College, Cornell University, New York, New York, USA
| |
Collapse
|
28
|
Samsuzzaman M, Uddin MS, Shah MA, Mathew B. Natural inhibitors on airway mucin: Molecular insight into the therapeutic potential targeting MUC5AC expression and production. Life Sci 2019; 231:116485. [PMID: 31116959 DOI: 10.1016/j.lfs.2019.05.041] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/08/2019] [Accepted: 05/15/2019] [Indexed: 11/19/2022]
Abstract
Airway mucin overproduction is the hallmark risk factor of asthma, which is associated with the reduction of lung function. An aberrant mucin expression is responsible for airway obstruction due to its high viscous characteristics. Among the mucins discovered, MUC5AC is the prime mucin of airway epithelia. Nowadays, mucins induced asthma and chronic obstructive pulmonary disease (COPD) are a great concern all over the world. This review focuses on the effects of natural compounds that can be beneficial to explore new drugs to halt MUC5AC secretion and production in airway epithelial, and also their underlying molecular mechanisms based on recent studies. Several researchers are seeking natural sources to identify a new potent MUC5AC inhibitory agent for clinical applications, because of countable limitations of existing synthetic drugs. Currently, flavonoids, glycoside and steroids like natural compounds have acquired great attention due to their anti-inflammatory and mucoregulatory effects. Most importantly, many natural compounds have shown their potential effects as the modulator of mucin expression, secretion, and production. Therefore, targeting airway MUC5AC expression and production represents an auspicious area of research for the development of drugs against various respiratory diseases.
Collapse
Affiliation(s)
- Md Samsuzzaman
- Department of Molecular Medicine, School of Medicine, Keimyung University, Daegu 42601, South Korea; Department of Food and Life Science, Pukyong National University, Busan 48513, South Korea
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh.
| | - Muhammad Ajmal Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| |
Collapse
|
29
|
Kijima T, Prince T, Neckers L, Koga F, Fujii Y. Heat shock factor 1 (HSF1)-targeted anticancer therapeutics: overview of current preclinical progress. Expert Opin Ther Targets 2019; 23:369-377. [PMID: 30931649 DOI: 10.1080/14728222.2019.1602119] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The heat shock factor 1 (HSF1) plays a pivotal role in guarding proteome stability or proteostasis by induction of heat shock proteins (HSPs). While HSF1 remains mostly latent in unstressed normal cells, it is constitutively active in malignant cells, rendering them addicted to HSF1 for their growth and survival. HSF1 affects tumorigenesis, cancer progression, and treatment resistance by preserving cancer proteostasis, thus suggesting disruption of HSF1 activity as a potential anticancer strategy. Areas covered: In this review, we focus on the HSF1 activation cycle and its interaction with HSPs, the role of HSF1 in oncogenesis, and development of HSF1-targeted drugs as a potential anticancer therapy for disrupting cancer proteostasis. Expert opinion: HSF1 systematically maintains proteostasis in malignant cancer cells. Although genomic instability is widely accepted as a hallmark of cancer, little is known about the role of proteostasis in cancer. Unveiling the complicated mechanism of HSF1 regulation, particularly in cancer cells, will enable further development of proteostasis-targeted anticancer therapy. ABBREVIATIONS AMPK: AMP-activated protein kinase; DBD: DNA-binding domain; HR-A/B; HR-C: heptad repeats; HSE: heat shock elements; HSF1: heat shock factor; HSPs: heat shock proteins; HSR: heat shock response; MEK: mitogen-activated protein kinase kinase; mTOR: mammalian target of rapamycin; NF1: neurofibromatosis type 1; P-TEFb: positive transcription elongation factor b; RD: regulatory domain; RNAi: RNA interference; TAD: transactivation domain; TRiC: TCP-1 ring complex.
Collapse
Affiliation(s)
- Toshiki Kijima
- a Department of Urology , Tokyo Medical and Dental University , Tokyo , Japan
| | - Thomas Prince
- b Departments of Urology and Molecular Functional Genomics , Geisinger Clinic , Danville , PA , USA
| | - Len Neckers
- c Urologic Oncology Branch , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Fumitaka Koga
- d Department of Urology , Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital , Tokyo , Japan
| | - Yasuhisa Fujii
- a Department of Urology , Tokyo Medical and Dental University , Tokyo , Japan
| |
Collapse
|
30
|
Nalli AD, Brown LE, Thomas CL, Sayers TJ, Porco JA, Henrich CJ. Sensitization of renal carcinoma cells to TRAIL-induced apoptosis by rocaglamide and analogs. Sci Rep 2018; 8:17519. [PMID: 30504817 PMCID: PMC6269514 DOI: 10.1038/s41598-018-35908-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/07/2018] [Indexed: 01/07/2023] Open
Abstract
Rocaglamide has been reported to sensitize several cell types to TRAIL-induced apoptosis. In recent years, advances in synthetic techniques have led to generation of novel rocaglamide analogs. However, these have not been extensively analyzed as TRAIL sensitizers, particularly in TRAIL-resistant renal cell carcinoma cells. Evaluation of rocaglamide and analogs identified 29 compounds that are able to sensitize TRAIL-resistant ACHN cells to TRAIL-induced, caspase-dependent apoptosis with sub-µM potency which correlated with their potency as protein synthesis inhibitors and with loss of cFLIP protein in the same cells. Rocaglamide alone induced cell cycle arrest, but not apoptosis. Rocaglates averaged 4–5-fold higher potency as TRAIL sensitizers than as protein synthesis inhibitors suggesting a potential window for maximizing TRAIL sensitization while minimizing effects of general protein synthesis inhibition. A wide range of other rocaglate effects (e.g. on JNK or RAF-MEK-ERK signaling, death receptor levels, ROS, ER stress, eIF4E phosphorylation) were assessed, but did not contribute to TRAIL sensitization. Other than a rapid loss of MCL-1, rocaglates had minimal effects on mitochondrial apoptotic pathway proteins. The identification of structurally diverse/mechanistically similar TRAIL sensitizing rocaglates provides insights into both rocaglate structure and function and potential further development for use in RCC-directed combination therapy.
Collapse
Affiliation(s)
- Ancy D Nalli
- National Cancer Institute, Molecular Targets Program, Frederick, MD, 21702, USA
| | - Lauren E Brown
- Boston University, Center for Molecular Discovery (BU-CMD), Department of Chemistry, Boston, MA, 02215, USA.
| | - Cheryl L Thomas
- National Cancer Institute, Molecular Targets Program, Frederick, MD, 21702, USA
| | - Thomas J Sayers
- National Cancer Institute, Cancer Inflammation Program, Frederick, MD, 21702, USA.,Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - John A Porco
- Boston University, Center for Molecular Discovery (BU-CMD), Department of Chemistry, Boston, MA, 02215, USA.
| | - Curtis J Henrich
- National Cancer Institute, Molecular Targets Program, Frederick, MD, 21702, USA. .,Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA.
| |
Collapse
|
31
|
Sibuyi NRS, Meyer M, Onani MO, Skepu A, Madiehe AM. Vascular targeted nanotherapeutic approach for obesity treatment. Int J Nanomedicine 2018; 13:7915-7929. [PMID: 30538468 PMCID: PMC6260142 DOI: 10.2147/ijn.s173424] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Obesity is a global epidemic that poses a serious health concern due to it being a risk factor for life-threatening chronic diseases, such as type 2 diabetes, cancer, and cardiovascular diseases. Pharmacotherapy remains the mainstay for the management of obesity; however, its usefulness is limited due to poor drug efficacy, non-specificity and toxic side effects. Therefore, novel approaches that could provide insights into obesity and obesity-associated diseases as well as development of novel anti-obesity treatment modalities or improvement on the existing drugs are necessary. While the ideal treatment of obesity should involve early intervention in susceptible individuals, targeted nanotherapy potentially provides a fresh perspective that might be better than the current conventional therapies. Independent studies have shown improved drug efficacy by using prohibitin (PHB)-targeted therapy in obese rodents and non-human primates, thus providing a proof of concept that targeted nanotherapy can be a feasible treatment for obesity. This review presents a brief global survey of obesity, its impact on human health, its current treatment and their limitations, and the role of angiogenesis and PHB in the development of obesity. Finally, the role and potential use of nanotechnology coupled with targeted drug delivery in the treatment of obesity are discussed.
Collapse
Affiliation(s)
- Nicole Remaliah Samantha Sibuyi
- Department of Science and Technology/Mintek Nanotechnology Innovation Centre, Biolabels Unit, Department of Biotechnology, University of the Western Cape, Bellville, South Africa,
| | - Mervin Meyer
- Department of Science and Technology/Mintek Nanotechnology Innovation Centre, Biolabels Unit, Department of Biotechnology, University of the Western Cape, Bellville, South Africa,
| | - Martin Opiyo Onani
- Organometallics and Nanomaterials, Department of Chemistry, University of the Western Cape, Bellville, South Africa
| | - Amanda Skepu
- Department of Science and Technology/Mintek Nanotechnology Innovation Centre, Biolabels Unit, Advanced Materials Division, Mintek, Johannesburg, South Africa
| | - Abram Madimabe Madiehe
- Department of Science and Technology/Mintek Nanotechnology Innovation Centre, Biolabels Unit, Department of Biotechnology, University of the Western Cape, Bellville, South Africa,
| |
Collapse
|
32
|
Chouha N, Hammoud H, Brogi S, Campiani G, Welsch C, Robert C, Vagner S, Cresteil T, Bentouhami E, Désaubry L. Discovery of Iminobenzimidazole Derivatives as Novel Cytotoxic Agents. THE OPEN MEDICINAL CHEMISTRY JOURNAL 2018. [DOI: 10.2174/1874104501812010074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In our quest to identify inhibitors of the eukaryotic translation initiation factor 4F (eIF4F), we serendipitously discovered a novel cytotoxic agent. Even though this compound did not inhibit translation, we explored the structural requirements for its cytotoxicity due to its structural originality. A series of 1,3-disubstituted iminobenzimidazoles was synthesized and evaluated for their in vitro cytotoxicity. The structure-activity relationship studies demonstrate that hydrophobic substituent is essential for activity. The most active compounds displayed a cytotoxicity in KB, HL60 and HCT116 human cancer cells with an IC50 of about 1μM. These first-in-class series of low molecular weight synthetic molecules may provide the basis for the development of new anticancer drugs.
Collapse
|
33
|
Accumulation of prohibitin is a common cellular response to different stressing stimuli and protects melanoma cells from ER stress and chemotherapy-induced cell death. Oncotarget 2018; 8:43114-43129. [PMID: 28562344 PMCID: PMC5522132 DOI: 10.18632/oncotarget.17810] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/19/2017] [Indexed: 01/01/2023] Open
Abstract
Melanoma is responsible for most deaths among skin cancers and conventional and palliative care chemotherapy are limited due to the development of chemoresistance. We used proteomic analysis to identify cellular responses that lead to chemoresistance of human melanoma cell lines to cisplatin. A systems approach to the proteomic data indicated the participation of specific cellular processes such as oxidative phosphorylation, mitochondrial organization and homeostasis, as well as the unfolded protein response (UPR) to be required for the survival of cells treated with cisplatin. Prohibitin (PHB) was among the proteins consistently accumulated, interacting with the functional clusters associated with resistance to cisplatin. We showed PHB accumulated at different levels in melanoma cell lines under stressing stimuli, such as (i) treatment with temozolomide (TMZ), dacarbazine (DTIC) and cisplatin; (ii) serum deprivation; (iii) tunicamycin, an UPR inducer. Prohibitin accumulated in the mitochondria of melanoma cells after cisplatin and tunicamycin treatment and its de novo accumulation led to chemoresistance melanoma cell lines. In contrast, PHB knock-down sensitized melanoma cells to cisplatin and tunicamycin treatment. We conclude that PHB participates in the survival of cells exposed to different stress stimuli, and can therefore serve as a target for the sensitization of melanoma cells to chemotherapy.
Collapse
|
34
|
Taheri Kal Koshvandi A, Heravi MM, Momeni T. Current Applications of Suzuki–Miyaura Coupling Reaction in The Total Synthesis of Natural Products: An update. Appl Organomet Chem 2018. [DOI: 10.10.1002/aoc.4210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | | | - Tayebeh Momeni
- Department of ChemistryAlzahra University Vanak Tehran Iran
| |
Collapse
|
35
|
Yuan G, Chen X, Liu Z, Wei W, Shu Q, Abou-Hamdan H, Jiang L, Li X, Chen R, Désaubry L, Zhou F, Xie D. Flavagline analog FL3 induces cell cycle arrest in urothelial carcinoma cell of the bladder by inhibiting the Akt/PHB interaction to activate the GADD45α pathway. J Exp Clin Cancer Res 2018; 37:21. [PMID: 29415747 PMCID: PMC5804081 DOI: 10.1186/s13046-018-0695-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/31/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Prohibitin 1 (PHB) is a potential target for the treatment of urothelial carcinoma of the bladder (UCB). FL3 is a newly synthesized agent that inhibits cancer cell proliferation by targeting the PHB protein; however, the effect of FL3 in UCB cells remains unexplored. METHODS FL3 was identified to be a potent inhibitor of UCB cell viability using CCK-8 (cell counting kit-8) assay. Then a series of in vitro and in vivo experiments were conducted to further demonstrate the inhibitory effect of FL3 on UCB cell proliferation and to determine the underlying mechanisms. RESULTS FL3 inhibited UCB cell proliferation and growth both in vitro and in vivo. By targeting the PHB protein, FL3 inhibited the interaction of Akt and PHB as well as Akt-mediated PHB phosphorylation, which consequently decreases the localization of PHB in the mitochondria. In addition, FL3 treatment resulted in cell cycle arrest in the G2/M phase, and this inhibitory effect of FL3 could be mimicked by knockdown of PHB. Through the microarray analysis of mRNA expression after FL3 treatment and knockdown of PHB, we found that the mRNA expression of the growth arrest and DNA damage-inducible alpha (GADD45α) gene were significantly upregulated. When knocked down the expression of GADD45α, the inhibitory effect of FL3 on cell cycle was rescued, suggesting that FL3-induced cell cycle inhibition is GADD45α dependent. CONCLUSION Our data provide that FL3 inhibits the interaction of Akt and PHB, which in turn activates the GADD45α-dependent cell cycle inhibition in the G2/M phase.
Collapse
Affiliation(s)
- Gangjun Yuan
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin Chen
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhuowei Liu
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wensu Wei
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qinghai Shu
- School of Material Science and Engineering, Beijing Institute of Technology, Beijing, China
| | - Hussein Abou-Hamdan
- Therapeutic Innovation Laboratory, UMR7200, CNRS/University of Strasbourg, Strasbourg, France
| | - Lijuan Jiang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xiangdong Li
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Rixin Chen
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Laurent Désaubry
- Therapeutic Innovation Laboratory, UMR7200, CNRS/University of Strasbourg, Strasbourg, France.
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
| | - Fangjian Zhou
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Dan Xie
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
36
|
Yu J, Wang C, Kong Q, Wu X, Lu JJ, Chen X. Recent progress in doxorubicin-induced cardiotoxicity and protective potential of natural products. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 40:125-139. [PMID: 29496165 DOI: 10.1016/j.phymed.2018.01.009] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 12/26/2017] [Accepted: 01/14/2018] [Indexed: 05/24/2023]
Abstract
BACKGROUND As an anthracycline antibiotic, doxorubicin (DOX) is one of the most potent and widely used chemotherapeutic agents for various types of solid tumors. Unfortunately, clinical application of this drug results in severe side effects of cardiotoxicity. PURPOSE We aim to review the research focused on elimination or reduction of DOX cardiotoxicity without affecting its anticancer efficacy by natural products. METHODS This study is based on pertinent papers that were retrieved by a selective search using relevant keywords in PubMed and ScienceDirect. The literature mainly focusing on natural products and herb extracts with therapeutic efficacies against experimental models both in vitro and in vivo was identified. RESULTS Current evidence revealed that multiple molecules and signaling pathways, such as oxidative stress, iron metabolism, and inflammation, are associated with DOX-induced cardiotoxicity. Based on these knowledge, various strategies were proposed, and thousands of compounds were screened. A number of natural products and herb extracts demonstrated potency in limiting DOX cardiotoxicity toward cultured cells and experimental animal models. CONCLUSIONS Though a panel of natural products and herb extracts demonstrate protective effects on DOX-induced cardiotoxicity in cells and animal models, their therapeutic potentials for clinical needs further investigation.
Collapse
Affiliation(s)
- Jie Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, PR China
| | - Changxi Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, PR China
| | - Qi Kong
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, PR China
| | - Xiaxia Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, PR China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, PR China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, PR China.
| |
Collapse
|
37
|
Taheri Kal Koshvandi A, Heravi MM, Momeni T. Current Applications of Suzuki–Miyaura Coupling Reaction in The Total Synthesis of Natural Products: An update. Appl Organomet Chem 2018. [DOI: 10.1002/aoc.4210] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
| | | | - Tayebeh Momeni
- Department of ChemistryAlzahra University Vanak Tehran Iran
| |
Collapse
|
38
|
Paz BM, Li Y, Thøgersen MK, Jørgensen KA. Enantioselective synthesis of cyclopenta[ b]benzofurans via an organocatalytic intramolecular double cyclization. Chem Sci 2017; 8:8086-8093. [PMID: 29568457 PMCID: PMC5855134 DOI: 10.1039/c7sc03006a] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 09/30/2017] [Indexed: 12/16/2022] Open
Abstract
An enantioselective organocatalytic strategy, combining Brønsted base and N-heterocyclic carbene catalysis in a unique manner, is demonstrated for a concise construction of the privileged cyclopenta[b]benzofuran scaffold, present in many bioactive compounds having both academic and commercial interests. The reaction concept relies on an intramolecular one-pot double cyclization involving a cycle-specific enantioselective Michael addition followed by a benzoin condensation of ortho-substituted cinnamaldehydes. Cyclopenta[b]benzofurans were achieved in moderate to good yields, with excellent stereoselectivities. A proof of principle for a diastereodivergent variation is demonstrated through the synthesis of cyclopenta[b]benzofurans containing two contiguous aromatic substituents in a substitution pattern present in commercial and natural compounds. Furthermore, several transformations have been performed, demonstrating the synthetic utility of the products. Finally, insights into the activation mode and stereoindution models are presented for this new synthetic strategy.
Collapse
Affiliation(s)
- Bruno Matos Paz
- Department of Chemistry , Aarhus University , DK-8000 Aarhus C , Denmark .
| | - Yang Li
- Department of Chemistry , Aarhus University , DK-8000 Aarhus C , Denmark .
| | | | | |
Collapse
|
39
|
Wang W, Clay A, Krishnan R, Lajkiewicz NJ, Brown LE, Sivaguru J, Porco JA. Total Syntheses of the Isomeric Aglain Natural Products Foveoglin A and Perviridisin B: Selective Excited-State Intramolecular Proton-Transfer Photocycloaddition. Angew Chem Int Ed Engl 2017; 56:14479-14482. [PMID: 28950418 PMCID: PMC5876029 DOI: 10.1002/anie.201707539] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Indexed: 11/10/2022]
Abstract
Selective excited-state intramolecular proton-transfer (ESIPT) photocycloaddition of 3-hydroxyflavones with trans, trans-1,4-diphenyl-1,3-butadiene is described. Using this methodology, total syntheses of the natural products (±)-foveoglin A and (±)-perviridisin B were accomplished. Enantioselective ESIPT photocycloaddition using TADDOLs as chiral hydrogen-bonding additives provided access to (+)-foveoglin A. Mechanistic studies have revealed the possibility for a photoinduced electron-transfer (PET) pathway.
Collapse
Affiliation(s)
- Wenyu Wang
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, 590 Commonwealth Avenue, Boston, Massachusetts, 02215, USA
| | - Anthony Clay
- Present address: Department of Chemistry and Center for Photochemical Sciences, Bowling Geen State University, Bowling Green, OH, 43403, USA
| | - Retheesh Krishnan
- Department of Chemistry, Government College for Women, Thiruvananthapuram, 695014, India
| | - Neil J Lajkiewicz
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, 590 Commonwealth Avenue, Boston, Massachusetts, 02215, USA
| | - Lauren E Brown
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, 590 Commonwealth Avenue, Boston, Massachusetts, 02215, USA
| | - Jayaraman Sivaguru
- Present address: Department of Chemistry and Center for Photochemical Sciences, Bowling Geen State University, Bowling Green, OH, 43403, USA
| | - John A Porco
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, 590 Commonwealth Avenue, Boston, Massachusetts, 02215, USA
| |
Collapse
|
40
|
Wang W, Clay A, Krishnan R, Lajkiewicz NJ, Brown LE, Sivaguru J, Porco JA. Total Syntheses of the Isomeric Aglain Natural Products Foveoglin A and Perviridisin B: Selective Excited‐State Intramolecular Proton‐Transfer Photocycloaddition. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201707539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Wenyu Wang
- Department of Chemistry, Center for Molecular Discovery (BU-CMD) Boston University 590 Commonwealth Avenue Boston Massachusetts 02215 USA
| | - Anthony Clay
- Present address: Department of Chemistry and Center for Photochemical Sciences Bowling Geen State University Bowling Green OH 43403 USA
| | - Retheesh Krishnan
- Department of Chemistry Government College for Women Thiruvananthapuram 695014 India
| | - Neil J. Lajkiewicz
- Department of Chemistry, Center for Molecular Discovery (BU-CMD) Boston University 590 Commonwealth Avenue Boston Massachusetts 02215 USA
| | - Lauren E. Brown
- Department of Chemistry, Center for Molecular Discovery (BU-CMD) Boston University 590 Commonwealth Avenue Boston Massachusetts 02215 USA
| | - Jayaraman Sivaguru
- Present address: Department of Chemistry and Center for Photochemical Sciences Bowling Geen State University Bowling Green OH 43403 USA
| | - John A. Porco
- Department of Chemistry, Center for Molecular Discovery (BU-CMD) Boston University 590 Commonwealth Avenue Boston Massachusetts 02215 USA
| |
Collapse
|
41
|
Ross JA, Robles-Escajeda E, Oaxaca DM, Padilla DL, Kirken RA. The prohibitin protein complex promotes mitochondrial stabilization and cell survival in hematologic malignancies. Oncotarget 2017; 8:65445-65456. [PMID: 29029444 PMCID: PMC5630344 DOI: 10.18632/oncotarget.18920] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 06/16/2017] [Indexed: 12/18/2022] Open
Abstract
Prohibitins (PHB1 and PHB2) have been proposed to play important roles in cancer development and progression, however their oncogenic mechanism of action has not been fully elucidated. Previously, we showed that the PHB1 and PHB2 protein complex is required for mitochondrial homeostasis and survival of normal human lymphocytes. In this study, novel evidence is provided that indicates mitochondrial prohibitins are overexpressed in hematologic tumor cells and promote cell survival under conditions of oxidative stress. Immunofluorescent confocal microscopy revealed both proteins to be primarily confined to mitochondria in primary patient lymphoid and myeloid tumor cells and tumor cell lines, including Kit225 cells. Subsequently, siRNA-mediated knockdown of PHB1 and PHB2 in Kit225 cells significantly enhanced sensitivity to H2O2-induced cell death, suggesting a protective or anti-apoptotic function in hematologic malignancies. Indeed, PHB1 and PHB2 protein levels were significantly higher in tumor cells isolated from leukemia and lymphoma patients compared to PBMCs from healthy donors. These findings suggest that PHB1 and PHB2 are upregulated during tumorigenesis to maintain mitochondrial integrity and therefore may serve as novel biomarkers and molecular targets for therapeutic intervention in certain types of hematologic malignancies.
Collapse
Affiliation(s)
- Jeremy A Ross
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Elisa Robles-Escajeda
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Derrick M Oaxaca
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Diana L Padilla
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Robert A Kirken
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| |
Collapse
|
42
|
Wang S, Darini C, Désaubry L, Koromilas AE. STAT1 Promotes KRAS Colon Tumor Growth and Susceptibility to Pharmacological Inhibition of Translation Initiation Factor eIF4A. Mol Cancer Ther 2017; 15:3055-3063. [PMID: 27913706 DOI: 10.1158/1535-7163.mct-16-0416] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/29/2016] [Accepted: 09/12/2016] [Indexed: 11/16/2022]
Abstract
The transcription factor STAT1 displays antitumor functions for certain forms of cancer via immunoregulatory and cell-autonomous pathways. Paradoxically, STAT1 can promote the survival of different tumor types treated with chemotherapeutic drugs through mechanisms that are not clearly defined. Herein, we demonstrate that STAT1 displays prosurvival effects in human KRAS colon tumor cells by regulating pathways that converge on the initiation of mRNA translation. Specifically, STAT1 increases PI3K class IB signaling and promotes the downregulation of the programmed cell death protein 4 (PDCD4), a protein with tumor-suppressive properties. PDCD4 downregulation by STAT1 increases the activity of the translation initiation factor eIF4A, which facilitates the cap-independent translation of mRNAs encoding for the antiapoptotic XIAP and BCL-XL in colon tumors with mutated but not normal KRAS Genetic inactivation of STAT1 impairs the tumorigenic potency of human KRAS colon tumor cells and renders them resistant to the antitumor effects of the pharmacologic inhibition of eIF4A in culture and immunodeficient mice. Our data demonstrate an important connection between mRNA translation and KRAS tumorigenesis under the control of STAT1, which can determine the susceptibility of KRAS tumors to pharmacologic inhibition of mRNA translation initiation. Mol Cancer Ther; 15(12); 3055-63. ©2016 AACR.
Collapse
Affiliation(s)
- Shuo Wang
- Lady Davis Institute for Medical Research, McGill University, Sir Mortimer B. Davis-Jewish General Hospital, Montreal, Quebec, Canada
| | - Cedric Darini
- Lady Davis Institute for Medical Research, McGill University, Sir Mortimer B. Davis-Jewish General Hospital, Montreal, Quebec, Canada
| | - Laurent Désaubry
- Laboratoire d'Innovation Thérapeutique (UMR 7200), Faculté de Pharmacie de l'Université de Strasbourg, Strasbourg, France
| | - Antonis E Koromilas
- Lady Davis Institute for Medical Research, McGill University, Sir Mortimer B. Davis-Jewish General Hospital, Montreal, Quebec, Canada.
- Department of Oncology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
43
|
Pro-differentiating effects of a synthetic flavagline on human teratocarcinomal cancer stem-like cells. Cell Biol Toxicol 2016; 33:295-306. [DOI: 10.1007/s10565-016-9375-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/04/2016] [Indexed: 01/28/2023]
|
44
|
Malka-Mahieu H, Newman M, Désaubry L, Robert C, Vagner S. Molecular Pathways: The eIF4F Translation Initiation Complex-New Opportunities for Cancer Treatment. Clin Cancer Res 2016; 23:21-25. [PMID: 27789529 DOI: 10.1158/1078-0432.ccr-14-2362] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/07/2016] [Accepted: 09/13/2016] [Indexed: 11/16/2022]
Abstract
The eIF4F complex regulates the cap-dependent mRNA translation process. It is becoming increasingly evident that aberrant activity of this complex is observed in many cancers, leading to the selective synthesis of proteins involved in tumor growth and metastasis. The selective translation of cellular mRNAs controlled by this complex also contributes to resistance to cancer treatments, and downregulation of the eIF4F complex components can restore sensitivity to various cancer therapies. Here, we review the contribution of the eIF4F complex to tumorigenesis, with a focus on its role in chemoresistance as well as the promising use of new small-molecule inhibitors of the complex, including flavaglines/rocaglates, hippuristanol, and pateamine A. Clin Cancer Res; 23(1); 21-25. ©2016 AACR.
Collapse
Affiliation(s)
- Hélène Malka-Mahieu
- Institut Curie, PSL Research University, CNRS UMR 3348, Orsay, France.,Université Paris Sud, Université Paris-Saclay, CNRS UMR 3348, Orsay, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Michelle Newman
- Institut Curie, PSL Research University, CNRS UMR 3348, Orsay, France.,Université Paris Sud, Université Paris-Saclay, CNRS UMR 3348, Orsay, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Laurent Désaubry
- Laboratory of Therapeutic Innovation (UMR 7200), Faculty of Pharmacy, University of Strasbourg-CNRS, Illkirch, France.,Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Caroline Robert
- INSERM U981, Villejuif, France. .,Institut de Cancérologie Gustave Roussy, Villejuif, France.,Université Paris-Sud, Kremlin-Bicêtre, France
| | - Stéphan Vagner
- Institut Curie, PSL Research University, CNRS UMR 3348, Orsay, France. .,Université Paris Sud, Université Paris-Saclay, CNRS UMR 3348, Orsay, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,INSERM U981, Villejuif, France.,Institut de Cancérologie Gustave Roussy, Villejuif, France
| |
Collapse
|
45
|
Zhao Q, Abou-Hamdan H, Désaubry L. Recent Advances in the Synthesis of Flavaglines, a Family of Potent Bioactive Natural Compounds Originating from Traditional Chinese Medicine. European J Org Chem 2016. [DOI: 10.1002/ejoc.201600437] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Qian Zhao
- Laboratory of Therapeutic Innovation (UMR 7200); Faculty of Pharmacy; University of Strasbourg-CNRS; Illkirch France
| | - Hussein Abou-Hamdan
- Laboratory of Therapeutic Innovation (UMR 7200); Faculty of Pharmacy; University of Strasbourg-CNRS; Illkirch France
| | - Laurent Désaubry
- Laboratory of Therapeutic Innovation (UMR 7200); Faculty of Pharmacy; University of Strasbourg-CNRS; Illkirch France
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry; College of Biotechnology; Tianjin University of Science and Technology; 300457 Tianjin P. R. China
| |
Collapse
|
46
|
Wang W, Cencic R, Whitesell L, Pelletier J, Porco JA. Synthesis of Aza-Rocaglates via ESIPT-Mediated (3+2) Photocycloaddition. Chemistry 2016; 22:12006-10. [PMID: 27338157 DOI: 10.1002/chem.201602953] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Indexed: 12/16/2022]
Abstract
Synthesis of aza-rocaglates, nitrogen-containing analogues of the rocaglate natural products, is reported. The route features ESIPT-mediated (3+2) photocycloaddition of 1-alkyl-2-aryl-3-hydroxyquinolinones with the dipolarophile methyl cinnamate. A continuous photoflow reactor was utilized for photocycloadditions. An array of compounds bearing the hexahydrocyclopenta[b]indole core structure was synthesized and evaluated in translation inhibition assays.
Collapse
Affiliation(s)
- Wenyu Wang
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, 590 Commonwealth Avenue, Boston, MA, 02215, USA
| | - Regina Cencic
- Department of Biochemistry and The Rosalind and Morris Goodman Cancer Research Centre Room 810, McGill University, 3655 Drummond St., Montreal, QC, H3G 1Y6, Canada
| | - Luke Whitesell
- Whitehead Institute for Biomedical Research (WIBR), Cambridge, MA, 02142, USA
| | - Jerry Pelletier
- Department of Biochemistry and The Rosalind and Morris Goodman Cancer Research Centre Room 810, McGill University, 3655 Drummond St., Montreal, QC, H3G 1Y6, Canada
| | - John A Porco
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, 590 Commonwealth Avenue, Boston, MA, 02215, USA.
| |
Collapse
|
47
|
Zhao Q, Tijeras-Raballand A, de Gramont A, Raymond E, Désaubry L. Bioisosteric modification of flavaglines. Tetrahedron Lett 2016. [DOI: 10.1016/j.tetlet.2016.05.089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
48
|
Lo Presti A, Cella E, Angeletti S, Ciccozzi M. Molecular epidemiology, evolution and phylogeny of Chikungunya virus: An updating review. INFECTION GENETICS AND EVOLUTION 2016; 41:270-278. [PMID: 27085290 DOI: 10.1016/j.meegid.2016.04.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 04/06/2016] [Accepted: 04/07/2016] [Indexed: 01/08/2023]
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus belonging to the Togaviridae family, causing a febrile illness associated with severe arthralgia and rash. In this review, we summarized a series of articles published from 2013 to 2016 concerning CHIKV epidemiology, phylogeny, vaccine and therapies, to give an update of our most recent article written in 2014 (Lo Presti et al.,2014). CHIKV infection was first reported in 1952 from Makonde plateaus and since this time caused many outbreaks worldwide, involving the Indian Ocean region, African countries, American continent and Italy. CHIKV infection is still underestimated and it is normally associated with clinical symptoms overlapping with dengue virus, recurring epidemics and mutations within the viral genome. These characteristics promote the geographical spread and the inability to control vector-mediated transmission of the virus. For these reasons, the majority of studies were aimed to describe outbreaks and to enhance knowledge on CHIKV biology, pathogenesis, infection treatment, and prevention. In this review, 16 studies on CHIKV phylogenetic and phylodinamics were considered, during the years 2013-2016. Phylogenetic and phylodinamic analysis are useful tools to investigate how the genealogy of a pathogen population is influenced by pathogen's demographic history, host immunological milieu and environmental/ecological factors. Phylogenetic tools were revealed important to reconstruct the geographic spread of CHIKV during the epidemics wave and to have information on the circulating strains of the virus, that are important for the prediction and control of the epidemics, as well as for vaccines and antiviral drugs development. In conclusion, this updating review can give a critical appraisal of the epidemiology, therapeutic and phylogenesis of CHIKV, reinforcing the need to monitor the geographic spread of virus and vectors.
Collapse
Affiliation(s)
- Alessandra Lo Presti
- Department of Infectious Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Eleonora Cella
- Department of Infectious Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, Rome, Italy; Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Silvia Angeletti
- Unit of Clinical Pathology and Microbiology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Massimo Ciccozzi
- Department of Infectious Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, Rome, Italy; Unit of Clinical Pathology and Microbiology, University Campus Bio-Medico of Rome, Rome, Italy.
| |
Collapse
|
49
|
Abstract
Human eukaryotic prohibitin (prohibitin-1 and prohibitin-2) is a membrane protein with different cellular localizations. It is involved in multiple cellular functions, including energy metabolism, proliferation, apoptosis, and senescence. The subcellular localization of prohibitin may determine its functions. Membrane prohibitin regulate the cellular signaling of membrane transport, nuclear prohibitin control transcription activation and the cell cycle, and mitochondrial prohibitin complex stabilize the mitochondrial genome and modulate mitochondrial dynamics, mitochondrial morphology, mitochondrial biogenesis, and the mitochondrial intrinsic apoptotic pathway. Moreover, prohibitin can translocates into the nucleus or the mitochondria under apoptotic signals and the subcellular shuttling of prohibitin is necessary for apoptosis process. Apoptosis is the process of programmed cell death that is important for the maintenance of normal physiological functions. Consequently, any alteration in the content, post-transcriptional modification (i.e. phosphorylation) or the nuclear or mitochondrial translocation of prohibitin may influence cell fate. Understanding the mechanisms of the expression and regulation of prohibitin may be useful for future research. This review provides an overview of the multifaceted and essential roles played by prohibitin in the regulation of cell survival and apoptosis.
Collapse
Affiliation(s)
- Ya-Ting Peng
- Department of Respiratory Medicine, Respiratory Disease Research Institute, Second XiangYa Hospital of Central South University, Changsha, 410011, People's Republic of China
| | | | | | | |
Collapse
|
50
|
Arai MA, Kofuji Y, Tanaka Y, Yanase N, Yamaku K, Fuentes RG, Karmakar UK, Ishibashi M. Synthesis of rocaglamide derivatives and evaluation of their Wnt signal inhibitory activities. Org Biomol Chem 2016; 14:3061-8. [DOI: 10.1039/c5ob02537k] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A new class of rocaglamide derivatives were synthesized by [3 + 2] cycloaddition and their Wnt signal inhibitory activities were evaluated.
Collapse
Affiliation(s)
- Midori A. Arai
- Department of Natural Product Chemistry
- Graduate School of Pharmaceutical Sciences
- Chiba University
- Chiba
- Japan
| | - Yuuki Kofuji
- Department of Natural Product Chemistry
- Graduate School of Pharmaceutical Sciences
- Chiba University
- Chiba
- Japan
| | - Yuuki Tanaka
- Department of Natural Product Chemistry
- Graduate School of Pharmaceutical Sciences
- Chiba University
- Chiba
- Japan
| | - Natsuki Yanase
- Department of Natural Product Chemistry
- Graduate School of Pharmaceutical Sciences
- Chiba University
- Chiba
- Japan
| | - Kazuki Yamaku
- Department of Natural Product Chemistry
- Graduate School of Pharmaceutical Sciences
- Chiba University
- Chiba
- Japan
| | - Rolly G. Fuentes
- Department of Natural Product Chemistry
- Graduate School of Pharmaceutical Sciences
- Chiba University
- Chiba
- Japan
| | - Utpal Kumar Karmakar
- Department of Natural Product Chemistry
- Graduate School of Pharmaceutical Sciences
- Chiba University
- Chiba
- Japan
| | - Masami Ishibashi
- Department of Natural Product Chemistry
- Graduate School of Pharmaceutical Sciences
- Chiba University
- Chiba
- Japan
| |
Collapse
|