1
|
Yin Z, You B, Bai Y, Zhao Y, Liao S, Sun Y, Wu Y. Natural Compounds Derived from Plants on Prevention and Treatment of Renal Cell Carcinoma: A Literature Review. Adv Biol (Weinh) 2024; 8:e2300025. [PMID: 37607316 DOI: 10.1002/adbi.202300025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 08/04/2023] [Indexed: 08/24/2023]
Abstract
Renal cell carcinoma (RCC) accounts for roughly 85% of all malignant kidney cancer. Therapeutic options for RCC have expanded rapidly over the past decade. Targeted therapy and immunotherapy have ushered in a new era of the treatment of RCC, which has facilitated the outcomes of RCC. However, the related adverse effects and drug resistance remain an urgent issue. Natural compounds are optional strategies to reduce mobility. Natural compounds are favored by clinicians and researchers due to their good tolerance and low economic burden. Many studies have explored the anti-RCC activity of natural products and revealed relevant mechanisms. In this article, the chemoprevention and therapeutic potential of natural compounds is reviewed and the mechanisms regarding natural compounds are explored.
Collapse
Affiliation(s)
- Zhenjie Yin
- Department of Urology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian, 365001, P. R. China
| | - Bingyong You
- Department of Urology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian, 365001, P. R. China
| | - Yuanyuan Bai
- Department of Urology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian, 365001, P. R. China
| | - Yu Zhao
- Department of Medical and Radiation Oncology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian, 365001, P. R. China
| | - Shangfan Liao
- Department of Urology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian, 365001, P. R. China
| | - Yingming Sun
- Department of Medical and Radiation Oncology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian, 365001, P. R. China
| | - Yongyang Wu
- Department of Urology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian, 365001, P. R. China
| |
Collapse
|
2
|
Ayed A. The role of natural products versus miRNA in renal cell carcinoma: implications for disease mechanisms and diagnostic markers. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6417-6437. [PMID: 38691151 DOI: 10.1007/s00210-024-03121-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
Natural products are chemical compounds produced by living organisms. They are isolated and purified to determine their function and can potentially be used as therapeutic agents. The ability of some bioactive natural products to modify the course of cancer is fascinating and promising. In the past 50 years, there have been advancements in cancer therapy that have increased survival rates for localized tumors. However, there has been little progress in treating advanced renal cell carcinoma (RCC), which is resistant to radiation and chemotherapy. Oncogenes and tumor suppressors are two roles played by microRNAs (miRNAs). They are involved in important pathogenetic mechanisms like hypoxia and epithelial-mesenchymal transition (EMT); they control apoptosis, cell growth, migration, invasion, angiogenesis, and proliferation through target proteins involved in various signaling pathways. Depending on their expression pattern, miRNAs may identify certain subtypes of RCC or distinguish tumor tissue from healthy renal tissue. As diagnostic biomarkers of RCC, circulating miRNAs show promise. There is a correlation between the expression patterns of several miRNAs and the prognosis and diagnosis of patients with RCC. Potentially high-risk primary tumors may be identified by comparing original tumor tissue with metastases. Variations in miRNA expression between treatment-sensitive and therapy-resistant patients' tissues and serum allow for the estimation of responsiveness to target therapy. Our knowledge of miRNAs' function in RCC etiology has a tremendous uptick. Finding and validating their gene targets could have an immediate effect on creating anticancer treatments based on miRNAs. Several miRNAs have the potential to be used as biomarkers for diagnosis and prognosis. This review provides an in-depth analysis of the current knowledge regarding natural compounds and their modes of action in combating cancer. Also, this study aims to give information about the diagnostic and prognostic value of miRNAs as cancer biomarkers and their involvement in the pathogenesis of RCC.
Collapse
Affiliation(s)
- Abdullah Ayed
- Department of Surgery, College of Medicine, University of Bisha, P.O Box 551, 61922, Bisha, Saudi Arabia.
| |
Collapse
|
3
|
Shi T, Hu W. Asymmetric Carbene Transfer: Enhancing Chemical Diversity for Drug Discovery. Chemistry 2024; 30:e202400971. [PMID: 38735847 DOI: 10.1002/chem.202400971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/10/2024] [Accepted: 05/12/2024] [Indexed: 05/14/2024]
Abstract
The quest to explore chemical space is vital for identifying novel disease targets, impacting both the effectiveness and safety profile of therapeutic agents. The tangible chemical space, currently estimated at a conservative 108 synthesized compounds, pales in comparison to the theoretically conceivable diversity of 1060 molecules. To bridge this vast gap, organic chemists are spearheading innovative methodologies that promise to broaden this limited chemical diversity. A beacon of this progressive wave is Asymmetric Carbene Transfer (ACT), a burgeoning strategy that significantly boosts molecular diversity with efficient bond-formation and precise chiral control. This review focuses on the capabilities of ACT in creating pharmaceutically significant molecules, encompassing an array of natural products and bioactive compounds. Through the lens of ACT, we discern its substantial influence on drug discovery, paving the way for novel therapeutic avenues by expanding the boundaries of molecular diversity. This review will shed light on prospective methodological developments of ACT and articulate their conceivable contributions to the medicinal chemistry arena.
Collapse
Affiliation(s)
- Taoda Shi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China, 510006
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education", Yantai University, Yantai, 264005, China
| | - Wenhao Hu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education", Yantai University, Yantai, 264005, China
| |
Collapse
|
4
|
Hodapp SJ, Gravel N, Kannan N, Newton AC. Cancer-associated mutations in protein kinase C theta are loss-of-function. Biochem J 2024; 481:759-775. [PMID: 38752473 PMCID: PMC11346454 DOI: 10.1042/bcj20240148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 06/11/2024]
Abstract
The Ca2+-independent, but diacylglycerol-regulated, novel protein kinase C (PKC) theta (θ) is highly expressed in hematopoietic cells where it participates in immune signaling and platelet function. Mounting evidence suggests that PKCθ may be involved in cancer, particularly blood cancers, breast cancer, and gastrointestinal stromal tumors, yet how to target this kinase (as an oncogene or as a tumor suppressor) has not been established. Here, we examine the effect of four cancer-associated mutations, R145H/C in the autoinhibitory pseudosubstrate, E161K in the regulatory C1A domain, and R635W in the regulatory C-terminal tail, on the cellular activity and stability of PKCθ. Live-cell imaging studies using the genetically-encoded fluorescence resonance energy transfer-based reporter for PKC activity, C kinase activity reporter 2 (CKAR2), revealed that the pseudosubstrate and C1A domain mutations impaired autoinhibition to increase basal signaling. This impaired autoinhibition resulted in decreased stability of the protein, consistent with the well-characterized behavior of Ca2+-regulated PKC isozymes wherein mutations that impair autoinhibition are paradoxically loss-of-function because the mutant protein is degraded. In marked contrast, the C-terminal tail mutation resulted in enhanced autoinhibition and enhanced stability. Thus, the examined mutations were loss-of-function by different mechanisms: mutations that impaired autoinhibition promoted the degradation of PKC, and those that enhanced autoinhibition stabilized an inactive PKC. Supporting a general loss-of-function of PKCθ in cancer, bioinformatics analysis revealed that protein levels of PKCθ are reduced in diverse cancers, including lung, renal, head and neck, and pancreatic. Our results reveal that PKCθ function is lost in cancer.
Collapse
Affiliation(s)
- Stefanie J. Hodapp
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, U.S.A
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Nathan Gravel
- Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA 30602, U.S.A
| | - Natarajan Kannan
- Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA 30602, U.S.A
| | - Alexandra C. Newton
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, U.S.A
| |
Collapse
|
5
|
Wardas B, Schneider JG, Klugbauer N, Flockerzi V, Beck A. Englerin A Inhibits T-Type Voltage-Gated Calcium Channels at Low Micromolar Concentrations. Mol Pharmacol 2023; 104:144-153. [PMID: 37399325 DOI: 10.1124/molpharm.122.000651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 05/31/2023] [Accepted: 06/08/2023] [Indexed: 07/05/2023] Open
Abstract
Englerin A (EA) is a potent agonist of tetrameric transient receptor potential canonical (TRPC) ion channels containing TRPC4 and TRPC5 subunits. TRPC proteins form cation channels that are activated by plasma membrane receptors. They convert extracellular signals such as angiotensin II into cellular responses, whereupon Na+ and Ca2+ influx and depolarization of the plasma membrane occur. Via depolarization, voltage-gated Ca2+ (CaV) channels can be activated, further increasing Ca2+ influx. We investigated the extent to which EA also affects the functions of CaV channels using the high-voltage-activated L-type Ca2+ channel CaV1.2 and the low-voltage-activated T-type Ca2+ channels CaV3.1, CaV3.2, and CaV3.3. After expression of cDNAs in human embryonic kidney (HEK293) cells, EA inhibited currents through all T-type channels at half-maximal inhibitory concentrations (IC50) of 7.5 to 10.3 μM. In zona glomerulosa cells of the adrenal gland, angiotensin II-induced elevation of cytoplasmic Ca2+ concentration leads to aldosterone release. We identified transcripts of low- and high-voltage-activated CaV channels and of TRPC1 and TRPC5 in the human adrenocortical (HAC15) zona glomerulosa cell line. Although no EA-induced TRPC activity was measurable, Ca2+ channel blockers distinguished T- and L-type Ca2+ currents. EA blocked 60% of the CaV current in HAC15 cells and T- and L-type channels analyzed at -30 mV and 10 mV were inhibited with IC50 values of 2.3 and 2.6 μM, respectively. Although the T-type blocker Z944 reduced basal and angiotensin II-induced 24-hour aldosterone release, EA was not effective. In summary, we show here that EA blocks CaV1.2 and T-type CaV channels at low-micromolar concentrations. SIGNIFICANCE STATEMENT: In this study we showed that englerin A (EA), a potent agonist of tetrameric transient receptor potential canonical (TRPC)4- or TRPC5-containing channels and currently under investigation to treat certain types of cancer, also inhibits the L-type voltage-gated Ca2+ (CaV) channel CaV1.2 and the T-type CaV channels CaV3.1, CaV3.2, and CaV3.3 channels at low micromolar concentrations.
Collapse
Affiliation(s)
- Barbara Wardas
- Experimentelle und Klinische Pharmakologie und Toxikologie/PZMS, Universität des Saarlandes, Homburg, Germany (B.W., V.F., A.B.); Department of Internal Medicine II, Universitätsklinikum des Saarlandes und Medizinische Fakultät der Universität des Saarlandes, Homburg, Germany (J.G.S.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg (J.G.S.); and Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany (N.K.)
| | - Jochen G Schneider
- Experimentelle und Klinische Pharmakologie und Toxikologie/PZMS, Universität des Saarlandes, Homburg, Germany (B.W., V.F., A.B.); Department of Internal Medicine II, Universitätsklinikum des Saarlandes und Medizinische Fakultät der Universität des Saarlandes, Homburg, Germany (J.G.S.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg (J.G.S.); and Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany (N.K.)
| | - Norbert Klugbauer
- Experimentelle und Klinische Pharmakologie und Toxikologie/PZMS, Universität des Saarlandes, Homburg, Germany (B.W., V.F., A.B.); Department of Internal Medicine II, Universitätsklinikum des Saarlandes und Medizinische Fakultät der Universität des Saarlandes, Homburg, Germany (J.G.S.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg (J.G.S.); and Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany (N.K.)
| | - Veit Flockerzi
- Experimentelle und Klinische Pharmakologie und Toxikologie/PZMS, Universität des Saarlandes, Homburg, Germany (B.W., V.F., A.B.); Department of Internal Medicine II, Universitätsklinikum des Saarlandes und Medizinische Fakultät der Universität des Saarlandes, Homburg, Germany (J.G.S.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg (J.G.S.); and Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany (N.K.)
| | - Andreas Beck
- Experimentelle und Klinische Pharmakologie und Toxikologie/PZMS, Universität des Saarlandes, Homburg, Germany (B.W., V.F., A.B.); Department of Internal Medicine II, Universitätsklinikum des Saarlandes und Medizinische Fakultät der Universität des Saarlandes, Homburg, Germany (J.G.S.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg (J.G.S.); and Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany (N.K.)
| |
Collapse
|
6
|
Mieville V, Griffioen AW, Benamran D, Nowak-Sliwinska P. Advanced in vitro models for renal cell carcinoma therapy design. Biochim Biophys Acta Rev Cancer 2023; 1878:188942. [PMID: 37343729 DOI: 10.1016/j.bbcan.2023.188942] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
Renal cell carcinoma (RCC) and its principal subtype, clear cell RCC, are the most diagnosed kidney cancer. Despite substantial improvement over the last decades, current pharmacological intervention still fails to achieve long-term therapeutic success. RCC is characterized by a high intra- and inter-tumoral heterogeneity and is heavily influenced by the crosstalk of the cells composing the tumor microenvironment, such as cancer-associated fibroblasts, endothelial cells and immune cells. Moreover, multiple physicochemical properties such as pH, interstitial pressure or oxygenation may also play an important role. These elements are often poorly recapitulated in in vitro models used for drug development. This inadequate recapitulation of the tumor is partially responsible for the current lack of an effective and curative treatment. Therefore, there are needs for more complex in vitro or ex vivo drug screening models. In this review, we discuss the current state-of-the-art of RCC models and suggest strategies for their further development.
Collapse
Affiliation(s)
- Valentin Mieville
- School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland; Translational Research Center in Oncohaematology, Geneva, Switzerland
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Daniel Benamran
- Division of Urology, Geneva University Hospitals, Geneva, Switzerland
| | - Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland; Translational Research Center in Oncohaematology, Geneva, Switzerland.
| |
Collapse
|
7
|
Chin Y, Gumilar KE, Li XG, Tjokroprawiro BA, Lu CH, Lu J, Zhou M, Sobol RW, Tan M. Targeting HSF1 for cancer treatment: mechanisms and inhibitor development. Theranostics 2023; 13:2281-2300. [PMID: 37153737 PMCID: PMC10157728 DOI: 10.7150/thno.82431] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/06/2023] [Indexed: 05/10/2023] Open
Abstract
Heat Shock Factor 1 (HSF1) is a master regulator of heat shock responsive signaling. In addition to playing critical roles in cellular heat shock response, emerging evidence suggests that HSF1 also regulates a non-heat shock responsive transcriptional network to handle metabolic, chemical, and genetic stress. The function of HSF1 in cellular transformation and cancer development has been extensively studied in recent years. Due to important roles for HSF1 for coping with various stressful cellular states, research on HSF1 has been very active. New functions and molecular mechanisms underlying these functions have been continuously discovered, providing new targets for novel cancer treatment strategies. In this article, we review the essential roles and mechanisms of HSF1 action in cancer cells, focusing more on recently discovered functions and their underlying mechanisms to reflect the new advances in cancer biology. In addition, we emphasize new advances with regard to HSF1 inhibitors for cancer drug development.
Collapse
Affiliation(s)
- Yeh Chin
- Graduate Institute of Biomedical Sciences and Research Center for Cancer Biology, China Medical University, Taichung, Taiwan, R.O.C
| | - Khanisyah E Gumilar
- Graduate Institute of Biomedical Sciences and Research Center for Cancer Biology, China Medical University, Taichung, Taiwan, R.O.C
- The Department of Obstetrics and Gynecology, Medical Faculty, Universitas Airlangga, Surabaya, Indonesia
| | - Xing-Guo Li
- Graduate Institute of Biomedical Sciences and Research Center for Cancer Biology, China Medical University, Taichung, Taiwan, R.O.C
- Institute of Biochemistry & Molecular Biology, China Medical University, Taichung, Taiwan, R.O.C
| | - Brahmana A. Tjokroprawiro
- The Department of Obstetrics and Gynecology, Medical Faculty, Universitas Airlangga, Surabaya, Indonesia
| | - Chien-Hsing Lu
- Department of Gynecology and Obstetrics, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C
| | - Jianrong Lu
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, USA
| | - Ming Zhou
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Robert W. Sobol
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, USA
| | - Ming Tan
- Graduate Institute of Biomedical Sciences and Research Center for Cancer Biology, China Medical University, Taichung, Taiwan, R.O.C
- Institute of Biochemistry & Molecular Biology, China Medical University, Taichung, Taiwan, R.O.C
| |
Collapse
|
8
|
Mushtaq A, Zahoor AF, Bilal M, Hussain SM, Irfan M, Akhtar R, Irfan A, Kotwica-Mojzych K, Mojzych M. Sharpless Asymmetric Dihydroxylation: An Impressive Gadget for the Synthesis of Natural Products: A Review. Molecules 2023; 28:2722. [PMID: 36985698 PMCID: PMC10051988 DOI: 10.3390/molecules28062722] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/21/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Sharpless asymmetric dihydroxylation is an important reaction in the enantioselective synthesis of chiral vicinal diols that involves the treatment of alkene with osmium tetroxide along with optically active quinine ligand. Sharpless introduced this methodology after considering the importance of enantioselectivity in the total synthesis of medicinally important compounds. Vicinal diols, produced as a result of this reaction, act as intermediates in the synthesis of different naturally occurring compounds. Hence, Sharpless asymmetric dihydroxylation plays an important role in synthetic organic chemistry due to its undeniable contribution to the synthesis of biologically active organic compounds. This review emphasizes the significance of Sharpless asymmetric dihydroxylation in the total synthesis of various natural products, published since 2020.
Collapse
Affiliation(s)
- Aqsa Mushtaq
- Medicinal Chemistry Research Lab, Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Ameer Fawad Zahoor
- Medicinal Chemistry Research Lab, Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Muhammad Bilal
- College of Computer Science and Technology, Zhejiang University, Hangzhou 310027, China
| | - Syed Makhdoom Hussain
- Department of Zoology, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Muhammad Irfan
- Department of Pharmaceutics, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Rabia Akhtar
- Medicinal Chemistry Research Lab, Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
- Department of Chemistry, Superior University, Faisalabad 38000, Pakistan
| | - Ali Irfan
- Medicinal Chemistry Research Lab, Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Katarzyna Kotwica-Mojzych
- Laboratory of Experimental Cytology, Medical University of Lublin, Radziwiłłowska 11, 20-080 Lublin, Poland
| | - Mariusz Mojzych
- Department of Chemistry, Siedlce University of Natural Sciences and Humanities, 3-Go Maja 54, 08-110 Siedlce, Poland
| |
Collapse
|
9
|
Zhou Q, Fu X, Xu J, Dong S, Liu C, Cheng D, Gao C, Huang M, Liu Z, Ni X, Hua R, Tu H, Sun H, Shen Q, Chen B, Zhang J, Zhang L, Yang H, Hu J, Yang W, Pei W, Yao Q, Sheng X, Zhang J, Yang WZ, Shen WL. Hypothalamic warm-sensitive neurons require TRPC4 channel for detecting internal warmth and regulating body temperature in mice. Neuron 2023; 111:387-404.e8. [PMID: 36476978 DOI: 10.1016/j.neuron.2022.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 06/28/2022] [Accepted: 11/07/2022] [Indexed: 12/12/2022]
Abstract
Precise monitoring of internal temperature is vital for thermal homeostasis in mammals. For decades, warm-sensitive neurons (WSNs) within the preoptic area (POA) were thought to sense internal warmth, using this information as feedback to regulate body temperature (Tcore). However, the cellular and molecular mechanisms by which WSNs measure temperature remain largely undefined. Via a pilot genetic screen, we found that silencing the TRPC4 channel in mice substantially attenuated hypothermia induced by light-mediated heating of the POA. Loss-of-function studies of TRPC4 confirmed its role in warm sensing in GABAergic WSNs, causing additional defects in basal temperature setting, warm defense, and fever responses. Furthermore, TRPC4 antagonists and agonists bidirectionally regulated Tcore. Thus, our data indicate that TRPC4 is essential for sensing internal warmth and that TRPC4-expressing GABAergic WSNs function as a novel cellular sensor for preventing Tcore from exceeding set-point temperatures. TRPC4 may represent a potential therapeutic target for managing Tcore.
Collapse
Affiliation(s)
- Qian Zhou
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Fu
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianhui Xu
- Thermoregulation and Inflammation Laboratory, Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Shiming Dong
- University of Chinese Academy of Sciences, Beijing 100049, China; Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Changhao Liu
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Dali Cheng
- Department of Electronic Engineering, Tsinghua University, Beijing 100084, China
| | - Cuicui Gao
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Minhua Huang
- Department of Biophysics, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhiduo Liu
- University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Integrated Optoelectronics, Institute of Semiconductors, Chinese Academy of Sciences, Beijing 100083, China
| | - Xinyan Ni
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Rong Hua
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200433, China
| | - Hongqing Tu
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Hongbin Sun
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Qiwei Shen
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200433, China
| | - Baoting Chen
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin Zhang
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| | - Liye Zhang
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Haitao Yang
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Ji Hu
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Wei Yang
- Department of Biophysics, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Weihua Pei
- State Key Laboratory of Integrated Optoelectronics, Institute of Semiconductors, Chinese Academy of Sciences, Beijing 100083, China
| | - Qiyuan Yao
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200433, China
| | - Xing Sheng
- Department of Electronic Engineering, Tsinghua University, Beijing 100084, China
| | - Jie Zhang
- Thermoregulation and Inflammation Laboratory, Chengdu Medical College, Chengdu, Sichuan 610500, China.
| | - Wen Z Yang
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China.
| | - Wei L Shen
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
10
|
Seenadera SD, Long SA, Akee R, Bermudez G, Parsonage G, Strope J, Peer C, Figg WD, Parker KA, Beech DJ, Beutler JA. Biological Effects of Modifications of the Englerin A Glycolate. ACS Med Chem Lett 2022; 13:1472-1476. [PMID: 36105325 PMCID: PMC9465829 DOI: 10.1021/acsmedchemlett.2c00258] [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: 05/27/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
Modifications at the glycolate moiety of englerin A were made to explore variations at the most sensitive site on the molecule for activity in the NCI 60 screen, wherein englerin A is highly potent and selective for renal cancer cells. Replacement of the glycolate by other functionalities as well as esterification of the glycolate hydroxyl yielded compounds which displayed excellent selectivity and potency compared with the natural product. TRPC4/5 ion channel experiments with five compounds showed delayed or reduced agonism with TRPC5, at much higher concentrations than englerin A. With TRPC4, these compounds all had no effect at 10 μM. The same compounds were not detectable in mouse serum after a single oral dose of 12.5 mg/kg. At 100 mg/kg p.o., no toxicity was observed, and blood levels were barely detectable. Intravenous administration led to toxicity but at substantially lower doses than for englerin A.
Collapse
Affiliation(s)
- Sarath
P. D. Seenadera
- Molecular
Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702 United States
| | - Sarah A. Long
- Molecular
Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702 United States
| | - Rhone Akee
- Leidos
Biomedical, FNLCR, Frederick, Maryland 21702 United States
| | - Gabriela Bermudez
- Department
of Chemistry, Stony Brook University, Stony Brook, New York 11790 United States
| | | | - Jonathan Strope
- Genitourinary
Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892 United States
| | - Cody Peer
- Genitourinary
Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892 United States
| | - W. Douglas Figg
- Genitourinary
Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892 United States
| | - Kathlyn A. Parker
- Department
of Chemistry, Stony Brook University, Stony Brook, New York 11790 United States
| | - David J. Beech
- School
of Medicine, University of Leeds, Leeds, LS2 9JT U.K.
| | - John A. Beutler
- Molecular
Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702 United States
| |
Collapse
|
11
|
Huang H, Cai Y, Hong X, Gao W, Tang J, Zhang S, Xu Z. T cell proliferation-related genes: Predicting prognosis, identifying the cold and hot tumors, and guiding treatment in clear cell renal cell carcinoma. Front Genet 2022; 13:948734. [PMID: 36118894 PMCID: PMC9478955 DOI: 10.3389/fgene.2022.948734] [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: 05/20/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Immunotherapy has become a new direction of current research because the effect of traditional radiotherapy and chemotherapy on clear cell renal cell carcinoma (ccRCC) is not satisfactory. T cell proliferation-related genes (TRGs) play a pivotal role in tumor progression by regulating the proliferation, activity, and function of immune cells. The purpose of our study is to construct and verify a prognostic model based on TRGs and to identify tumor subtypes that may guide treatment through comprehensive bioinformatics analyses. Methods: RNA sequencing data, clinical information, and somatic mutation data of ccRCC are obtained from The Cancer Genome Atlas (TCGA) database. We identified the prognosis-related TRGs which were differentially expressed between normal and tumor tissues. After dividing the patients into a train set and a test set according to proportion 1:1 randomly, the least absolute shrinkage and selection operator (LASSO) and multivariate Cox regression analysis were performed to construct a risk-stratified model. Its prediction performance was verified. Then, Gene Set Enrichment Analysis (GSEA), principal component analysis (PCA), tumor microenvironment (TME) analysis, and the half-maximal inhibitory concentration (IC50) prediction were performed between the different groups of patients. To further discuss the immunotherapy between hot and cold tumors, we divided all patients into two clusters based on TRGs through unsupervised learning. Analyzing the gene mutation and calculating the tumor mutation burden (TMB), we further explored the relationship between somatic mutations and grouping or clustering. Results: Risk-stratified model and nomogram predict the prognosis of ccRCC patients accurately. Functional enrichment analyses suggested that TRGs mainly focused on the biological pathways related to tumor progression and immune response. Different tumor microenvironment, drug resistance, and TMB can be distinguished clearly according to both risk stratification and tumor subtype clustering. Conclusion: In this study, a new stratification model of ccRCC based on TRGs was established, which can accurately predict the prognosis of patients. IC50 prediction may guide the application of anti-tumor drugs. The distinction between hot and cold tumors provides a reference for clinical immunotherapy.
Collapse
|
12
|
Neo SP, Alli-Shaik A, Wee S, Lim Z, Gunaratne J. Englerin A Rewires Phosphosignaling via Hsp27 Hyperphosphorylation to Induce Cytotoxicity in Renal Cancer Cells. J Proteome Res 2022; 21:1948-1960. [PMID: 35838755 DOI: 10.1021/acs.jproteome.2c00248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Englerin A (EA) is a small-molecule natural product with selective cytotoxicity against renal cancer cells. EA has been shown to induce apoptosis and cell death through cell-cycle arrest and/or insulin signaling pathways. However, its biological mode of action or targets in renal cancer remains enigmatic. In this study, we employed advanced mass spectrometry-based phosphoproteomics approaches to identify EA's functional roles in renal cancer. We identified 10,940 phosphorylation sites, of which 706 sites exhibited EA-dependent phosphorylation changes. Integrated analysis of motifs and interaction networks suggested activation of stress-activated kinases including p38 upon EA treatment. Of note, a downstream target of p38, Hsp27, was found to be hyperphosphorylated on multiple sites upon EA treatment. Among these, a novel site Ser65 on Hsp27, which was further validated by targeted proteomics, was shown to be crucial for EA-induced cytotoxicity in renal cancer cells. Taken together, these data reveal the complex signaling cascade that is induced upon EA treatment and importantly provide insights into its effects on downstream molecular signaling.
Collapse
Affiliation(s)
- Suat Peng Neo
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Asfa Alli-Shaik
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Sheena Wee
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Zijie Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Jayantha Gunaratne
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
| |
Collapse
|
13
|
Schremmer C, Steinritz D, Gudermann T, Beech DJ, Dietrich A. An ex vivo perfused ventilated murine lung model suggests lack of acute pulmonary toxicity of the potential novel anticancer agent (-)-englerin A. Arch Toxicol 2022; 96:1055-1063. [PMID: 35165752 PMCID: PMC8921049 DOI: 10.1007/s00204-022-03235-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/20/2022] [Indexed: 12/19/2022]
Abstract
(-)-Englerin A (EA), a potential novel anti-cancer drug, is a potent selective activator of classical transient receptor potential 4 and 5 (TRPC4, TRPC5) channels. As TRPC4 channels are expressed and functional in the lung endothelium, possible side effects such as lung edema formation may arise during its administration. Well-established in vivo rodent models for toxicological testing, however, rapidly degrade this compound to its inactive derivative, englerin B. Therefore, we chose an ex vivo isolated perfused and ventilated murine lung (IPVML) model to detect edema formation due to toxicants, which also reduces the number of incriminating animal experiments required. To evaluate the sensitivity of the IPVML model, short-time (10 min) drops of the pH from 7.4 down to 4.0 were applied, which resulted in linear changes of tidal volumes, wet-to-dry weight ratios and incorporation of FITC-coupled dextran particles from the perfusate. As expected, biological activity of EA was preserved after perfusion in the IPVML model. Concentrations of 50-100 nM EA continuously perfused through the IPVML model did not change tidal volumes and lung weights significantly. Wet-to-dry weight ratios were increased after perfusion of 100 nM EA but permeation of FITC-coupled dextran particles from the perfusate to the lung tissues was not significantly different. Therefore, EA shows little or no significant acute pulmonary toxicity after application of doses expected to activate target ion channels and the IPVML is a sensitive powerful ex vivo model for evaluating acute lung toxicity in accordance with the 3R rules for animal experimentation.
Collapse
Affiliation(s)
- Christian Schremmer
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Medical Faculty, LMU-Munich, Nussbaum Str. 26, 80336, Munich, Germany
| | - Dirk Steinritz
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstraße 11, 80937, Munich, Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Medical Faculty, LMU-Munich, Nussbaum Str. 26, 80336, Munich, Germany
| | - David J Beech
- School of Medicine, University of Leeds, LIGHT Building, Clarendon Way, Leeds, LS2 9JT, England, UK
| | - Alexander Dietrich
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Medical Faculty, LMU-Munich, Nussbaum Str. 26, 80336, Munich, Germany.
| |
Collapse
|
14
|
Shi C, Yang EJ, Tao S, Ren G, Mou PK, Shim JS. Natural products targeting cancer cell dependency. J Antibiot (Tokyo) 2021; 74:677-686. [PMID: 34163025 DOI: 10.1038/s41429-021-00438-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023]
Abstract
Precision cancer medicine is a tailored treatment approach for individual cancer patients with different genomic characteristics. Mutated or hyperactive oncogenes have served as main drug targets in current precision cancer medicine, while defective or inactivated tumor suppressors in general have not been considered as druggable targets. Synthetic lethality is one of very few approaches that enable to target defective tumor suppressors with pharmacological agents. Synthetic lethality exploits cancer cell dependency on a protein or pathway, which arises when the function of a tumor suppressor is defective. This approach has been proven to be effective in clinical settings since the successful clinical introduction of BRCA-PARP synthetic lethality for the treatment of breast and ovarian cancer with defective BRCA. Subsequently, large-scale screenings with RNAi, CRISPR/Cas9-sgRNAs, and chemical libraries have been applied to identify synthetic lethal partners of tumor suppressors. Natural products are an important source for the discovery of pharmacologically active small molecules. However, little effort has been made in the discovery of synthetic lethal small molecules from natural products. This review introduces recent advances in the discovery of natural products targeting cancer cell dependency and discusses potentials of natural products in the precision cancer medicine.
Collapse
Affiliation(s)
- Changxiang Shi
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Eun Ju Yang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Shishi Tao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Guowen Ren
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Pui Kei Mou
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Joong Sup Shim
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China. .,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China.
| |
Collapse
|
15
|
Minh Le T, Szakonyi Z. Enantiomeric Isopulegol as the Chiral Pool in the Total Synthesis of Bioactive Agents. CHEM REC 2021; 22:e202100194. [PMID: 34553822 DOI: 10.1002/tcr.202100194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/05/2021] [Indexed: 11/12/2022]
Abstract
Isopulegol, a pool of abundant chiral terpene, has long served as the starting material for the total synthesis of isopulegol-based drugs. As an inexpensive and versatile starting material, this compound continues to serve modern synthetic chemistry. This review highlights the total syntheses of terpenoids in the period from 1980 to 2020 in which with isopulegol applied as a building block.
Collapse
Affiliation(s)
- Tam Minh Le
- Institute of Pharmaceutical Chemistry, University of Szeged, Interdisciplinary Excellent Center, Eötvös utca 6, H-6720, Szeged, Hungary.,Stereochemistry Research Group of the Hungarian Academy Science, Eötvös utca 6, H-6720, Szeged, Hungary
| | - Zsolt Szakonyi
- Institute of Pharmaceutical Chemistry, University of Szeged, Interdisciplinary Excellent Center, Eötvös utca 6, H-6720, Szeged, Hungary.,Interdisciplinary Centre of Natural Products, University of Szeged, Eötvös utca 6, H-6720, Szeged, Hungary
| |
Collapse
|
16
|
Wang Y, Gao N, Feng Y, Cai M, Li Y, Xu X, Zhang H, Yao D. Protein kinase C theta (Prkcq) affects nerve degeneration and regeneration through the c-fos and c-jun pathways in injured rat sciatic nerves. Exp Neurol 2021; 346:113843. [PMID: 34418453 DOI: 10.1016/j.expneurol.2021.113843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/23/2021] [Accepted: 08/15/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Previous finding using DNA microarray and bioinformatics analysis, we have reported some key factors which regulated gene expression and signaling pathways in injured sciatic nerve during Wallerian Degeneration (WD). This research is focused on protein kinase C theta (Prkcq) participates in the regulation of the WD process. METHODS In this study, we explored the molecular mechanism by which Prkcq in Schwann cells (SCs) affects nerve degeneration and regeneration in vivo and in vitro after rat sciatic nerve injury. RESULTS Study of the cross-sectional model showed that Prkcq expression decreased significantly during sciatic nerve repair. Functional analysis showed that upregulation and downregulation of Prkcq could affect the proliferation, migration and apoptosis of Schwann cells and lead to the expression of related factors through the activation of the β-catenin, c-fos, and p-c-jun/c-jun pathways. CONCLUSION The study provides insights into the role of Prkcq in early WD during peripheral nerve degeneration and/or regeneration.
Collapse
Affiliation(s)
- Yi Wang
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, PR China
| | - Nannan Gao
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, PR China
| | - Yumei Feng
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, PR China
| | - Min Cai
- Medical School of Nantong University, Nantong, Jiangsu 226001, PR China.
| | - Yuting Li
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, PR China
| | - Xi Xu
- Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - Huanhuan Zhang
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, PR China
| | - Dengbing Yao
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, PR China.
| |
Collapse
|
17
|
de Sousa Valente J, Alawi KM, Bharde S, Zarban AA, Kodji X, Thapa D, Argunhan F, Barrett B, Nagy I, Brain SD. (-)-Englerin-A Has Analgesic and Anti-Inflammatory Effects Independent of TRPC4 and 5. Int J Mol Sci 2021; 22:6380. [PMID: 34203675 PMCID: PMC8232259 DOI: 10.3390/ijms22126380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/13/2022] Open
Abstract
Recently, we found that the deletion of TRPC5 leads to increased inflammation and pain-related behaviour in two animal models of arthritis. (-)-Englerin A (EA), an extract from the East African plant Phyllanthus engleri has been identified as a TRPC4/5 agonist. Here, we studied whether or not EA has any anti-inflammatory and analgesic properties via TRPC4/5 in the carrageenan model of inflammation. We found that EA treatment in CD1 mice inhibited thermal hyperalgesia and mechanical allodynia in a dose-dependent manner. Furthermore, EA significantly reduced the volume of carrageenan-induced paw oedema and the mass of the treated paws. Additionally, in dorsal root ganglion (DRG) neurons cultured from WT 129S1/SvIm mice, EA induced a dose-dependent cobalt uptake that was surprisingly preserved in cultured DRG neurons from 129S1/SvIm TRPC5 KO mice. Likewise, EA-induced anti-inflammatory and analgesic effects were preserved in the carrageenan model in animals lacking TRPC5 expression or in mice treated with TRPC4/5 antagonist ML204.This study demonstrates that while EA activates a sub-population of DRG neurons, it induces a novel TRPC4/5-independent analgesic and anti-inflammatory effect in vivo. Future studies are needed to elucidate the molecular and cellular mechanisms underlying EA's anti-inflammatory and analgesic effects.
Collapse
Affiliation(s)
- João de Sousa Valente
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| | - Khadija M Alawi
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| | - Sabah Bharde
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| | - Ali A. Zarban
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
- Department of Pharmacological Sciences, Faculty of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Xenia Kodji
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| | - Dibesh Thapa
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| | - Fulye Argunhan
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| | - Brentton Barrett
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| | - Istvan Nagy
- Nociception Group, Section of Anaesthetic, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK;
| | - Susan D. Brain
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| |
Collapse
|
18
|
Fermaintt CS, Peramuna T, Cai S, Takahashi-Ruiz L, Essif JN, Grant CV, O’Keefe BR, Mooberry SL, Cichewicz RH, Risinger AL. Yuanhuacine Is a Potent and Selective Inhibitor of the Basal-Like 2 Subtype of Triple Negative Breast Cancer with Immunogenic Potential. Cancers (Basel) 2021; 13:cancers13112834. [PMID: 34200174 PMCID: PMC8201195 DOI: 10.3390/cancers13112834] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 12/28/2022] Open
Abstract
The heterogeneity of triple negative breast cancer (TNBC) has led to efforts to further subtype this disease with the hope of identifying new molecular liabilities and drug targets. Furthermore, the finding that TNBC is the most inherently immunogenic type of breast cancer provides the potential for effective treatment with immune checkpoint inhibitors and immune adjuvants. Thus, we devised a dual screen to identify compounds from natural product extracts with TNBC subtype selectivity that also promote the expression of cytokines associated with antitumor immunity. These efforts led to the identification of yuanhuacine (1) as a potent and highly selective inhibitor of the basal-like 2 (BL2) subtype of TNBC that also promoted an antitumor associated cytokine signature in immune cells. The mechanism of action of yuanhuacine for both phenotypes depends on activation of protein kinase C (PKC), defining a novel target for the treatment of this clinical TNBC subtype. Yuanhuacine showed potent antitumor efficacy in animals bearing BL2 tumors further demonstrating that PKC could function as a potential pharmacological target for the treatment of the BL2 subtype of TNBC.
Collapse
Affiliation(s)
- Charles S. Fermaintt
- Department of Pharmacology, Mays Cancer Center, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; (C.S.F.); (L.T.-R.); (J.N.E.); (C.V.G.); (S.L.M.)
| | - Thilini Peramuna
- Department of Chemistry and Biochemistry and Natural Products Discovery Group, University of Oklahoma, Norman, OK 73019, USA; (T.P.); (S.C.); (R.H.C.)
| | - Shengxin Cai
- Department of Chemistry and Biochemistry and Natural Products Discovery Group, University of Oklahoma, Norman, OK 73019, USA; (T.P.); (S.C.); (R.H.C.)
| | - Leila Takahashi-Ruiz
- Department of Pharmacology, Mays Cancer Center, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; (C.S.F.); (L.T.-R.); (J.N.E.); (C.V.G.); (S.L.M.)
| | - Jacob Nathaniel Essif
- Department of Pharmacology, Mays Cancer Center, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; (C.S.F.); (L.T.-R.); (J.N.E.); (C.V.G.); (S.L.M.)
| | - Corena V. Grant
- Department of Pharmacology, Mays Cancer Center, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; (C.S.F.); (L.T.-R.); (J.N.E.); (C.V.G.); (S.L.M.)
| | - Barry R. O’Keefe
- Natural Products Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis and Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA;
| | - Susan L. Mooberry
- Department of Pharmacology, Mays Cancer Center, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; (C.S.F.); (L.T.-R.); (J.N.E.); (C.V.G.); (S.L.M.)
| | - Robert H. Cichewicz
- Department of Chemistry and Biochemistry and Natural Products Discovery Group, University of Oklahoma, Norman, OK 73019, USA; (T.P.); (S.C.); (R.H.C.)
| | - April L. Risinger
- Department of Pharmacology, Mays Cancer Center, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; (C.S.F.); (L.T.-R.); (J.N.E.); (C.V.G.); (S.L.M.)
- Correspondence: ; Tel.: +1-210-567-6267
| |
Collapse
|
19
|
Nicolle A, Zhang Y, Belguise K. The Emerging Function of PKCtheta in Cancer. Biomolecules 2021; 11:biom11020221. [PMID: 33562506 PMCID: PMC7915540 DOI: 10.3390/biom11020221] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/22/2021] [Accepted: 02/02/2021] [Indexed: 12/30/2022] Open
Abstract
Protein Kinase C theta (PKCθ) is a serine/threonine kinase that belongs to the novel PKC subfamily. In normal tissue, its expression is restricted to skeletal muscle cells, platelets and T lymphocytes in which PKCθ controls several essential cellular processes such as survival, proliferation and differentiation. Particularly, PKCθ has been extensively studied for its role in the immune system where its translocation to the immunological synapse plays a critical role in T cell activation. Beyond its physiological role in immune responses, increasing evidence implicates PKCθ in the pathology of various diseases, especially autoimmune disorders and cancers. In this review, we discuss the implication of PKCθ in various types of cancers and the PKCθ-mediated signaling events controlling cancer initiation and progression. In these types of cancers, the high PKCθ expression leads to aberrant cell proliferation, migration and invasion resulting in malignant phenotype. The recent development and application of PKCθ inhibitors in the context of autoimmune diseases could benefit the emergence of treatment for cancers in which PKCθ has been implicated.
Collapse
|
20
|
Synthesis of Polycyclic Ether-Benzopyrans and In Vitro Inhibitory Activity against Leishmania tarentolae. Molecules 2020; 25:molecules25225461. [PMID: 33233418 PMCID: PMC7700287 DOI: 10.3390/molecules25225461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/14/2020] [Accepted: 11/18/2020] [Indexed: 11/21/2022] Open
Abstract
Construction of a focused library of polycyclic ether-benzopyrans was undertaken in order to discover new therapeutic compounds that affect Leishmania growth and infectivity. This is especially of interest since there are few drug therapies for leishmaniasis that do not have serious drawbacks such high cost, side effects, and emerging drug resistance. The construction of these polycyclic ether-benzopyrans utilized an acetoxypyranone-alkene [5+2] cycloaddition and the Suzuki-Miyaura cross-coupling. The multi-gram quantity of the requisite aryl bromide was obtained followed by effective Pd-catalyzed coupling with boronic acid derivatives. Compounds were tested in vitro using the parasitic protozoan, Leishmania tarentolae. Effects of concentration, time, and exposure to light were evaluated. In addition, the effects on secreted acid phosphatase activity and nitric oxide production were investigated, since both have been implicated in parasite infectivity. The data presented herein are indicative of disruption of the Leishmania tarentolae and thus provide impetus for the development and testing of a more extensive library.
Collapse
|
21
|
Wang J, Jin W, Zhou X, Li J, Xu C, Ma Z, Wang J, Qin L, Zhou B, Ding W, Gao T, Yao H, Chen Z. Identification, Structure-Activity Relationships of Marine-Derived Indolocarbazoles, and a Dual PKCθ/δ Inhibitor with Potent Antipancreatic Cancer Efficacy. J Med Chem 2020; 63:12978-12991. [PMID: 33100009 DOI: 10.1021/acs.jmedchem.0c01271] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Protein kinases C (PKCs) are a family of serine/threonine kinases involved in various cellular processes, including proliferation, differentiation, cell survival, and apoptosis. Here, we report the identification, structure-activity relationship (SAR), and 3D-QSAR studies of 69 natural indolocarbazoles, including 15 new compounds, from marine streptomyces strains. Interestingly, we found that the chair conformational isomer of 7-oxo-staurosporine (compound 15) inhibited PKCθ more potently than the corresponding boat isomer. An evaluation of kinase selectivity and antitumor efficacy revealed that 15 was a potent dual PKCθ/δ inhibitor and that it could efficiently inhibit tumor growth in pancreatic cancer (PC) by inducing cellular apoptosis and suppressing the NF-κB/p-P65 pathway. In addition, we demonstrated that overexpression of p-PKCδ and p-P65 was associated with poor survival rates in patients with PC, and p-PKCθ expression also showed significant positive correlations with p-PKCδ and p-P65 levels. Finally, the PC patient-derived xenograft model further confirmed the potential anti-PC efficacy of 15.
Collapse
Affiliation(s)
- Jinhui Wang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, No. 1 Zheda Road, Zhoushan 316021, China
| | - Weiyang Jin
- College of Life and Environmental Sciences, Hangzhou Normal University, No. 2318, Yuhangtang Road, Hangzhou 311121, China
| | - Xiaoxin Zhou
- College of Life and Environmental Sciences, Hangzhou Normal University, No. 2318, Yuhangtang Road, Hangzhou 311121, China
| | - Jiaqi Li
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, No. 1 Zheda Road, Zhoushan 316021, China
| | - Chengdong Xu
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, No. 1 Zheda Road, Zhoushan 316021, China
| | - Zhongjun Ma
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, No. 1 Zheda Road, Zhoushan 316021, China
| | - Jianan Wang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, No. 1 Zheda Road, Zhoushan 316021, China
| | - Lele Qin
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, No. 1 Zheda Road, Zhoushan 316021, China
| | - Biao Zhou
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, No. 1 Zheda Road, Zhoushan 316021, China
| | - Wanjing Ding
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, No. 1 Zheda Road, Zhoushan 316021, China
| | - Tingting Gao
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, No. 1 Zheda Road, Zhoushan 316021, China
| | - Hangping Yao
- The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou 310003, China
| | - Zhe Chen
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medicine, Zhejiang Chinese Medical University, No. 548, Binwen Road, Hangzhou 310053, China
| |
Collapse
|
22
|
Development of synthetic lethality in cancer: molecular and cellular classification. Signal Transduct Target Ther 2020; 5:241. [PMID: 33077733 PMCID: PMC7573576 DOI: 10.1038/s41392-020-00358-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/27/2022] Open
Abstract
Recently, genetically targeted cancer therapies have been a topic of great interest. Synthetic lethality provides a new approach for the treatment of mutated genes that were previously considered unable to be targeted in traditional genotype-targeted treatments. The increasing researches and applications in the clinical setting made synthetic lethality a promising anticancer treatment option. However, the current understandings on different conditions of synthetic lethality have not been systematically assessed and the application of synthetic lethality in clinical practice still faces many challenges. Here, we propose a novel and systematic classification of synthetic lethality divided into gene level, pathway level, organelle level, and conditional synthetic lethality, according to the degree of specificity into its biological mechanism. Multiple preclinical findings of synthetic lethality in recent years will be reviewed and classified under these different categories. Moreover, synthetic lethality targeted drugs in clinical practice will be briefly discussed. Finally, we will explore the essential implications of this classification as well as its prospects in eliminating existing challenges and the future directions of synthetic lethality.
Collapse
|
23
|
Wu Z, Suppo JS, Tumova S, Strope J, Bravo F, Moy M, Weinstein ES, Peer CJ, Figg WD, Chain WJ, Echavarren AM, Beech DJ, Beutler JA. Bridgehead Modifications of Englerin A Reduce TRPC4 Activity and Intravenous Toxicity but not Cell Growth Inhibition. ACS Med Chem Lett 2020; 11:1711-1716. [PMID: 32944138 DOI: 10.1021/acsmedchemlett.0c00186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/03/2020] [Indexed: 11/29/2022] Open
Abstract
Modifications at the bridgehead position of englerin A were made to explore the effects of variation at this site on the molecule for biological activity, as judged by the NCI 60 screen, in which englerin A is highly potent and selective for renal cancer cells. Replacement of the isopropyl group by other, larger substituents yielded compounds which displayed excellent selectivity and potency comparable to the natural product. Selected compounds were also evaluated for their effect on the ion channel TRPC4 as well as for intravenous toxicity in mice, and these had lower potency in both assays compared to englerin A.
Collapse
Affiliation(s)
- Zhenhua Wu
- Department of Chemistry & Biochemistry, University of Delaware, 163 The Green, Newark, Delaware 19716, United States
| | - Jean-Simon Suppo
- Institute of Chemical Research of Catalonia (ICIQ), 43007 Tarragona, Spain
| | - Sarka Tumova
- School of Medicine, University of Leeds, Leeds LS2 9JT, U.K
| | - Jonathan Strope
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - Fernando Bravo
- Institute of Chemical Research of Catalonia (ICIQ), 43007 Tarragona, Spain
| | - Melody Moy
- Department of Chemistry & Biochemistry, University of Delaware, 163 The Green, Newark, Delaware 19716, United States
| | - Ethan S. Weinstein
- Department of Chemistry & Biochemistry, University of Delaware, 163 The Green, Newark, Delaware 19716, United States
| | - Cody J. Peer
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - William D. Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - William J. Chain
- Department of Chemistry & Biochemistry, University of Delaware, 163 The Green, Newark, Delaware 19716, United States
| | | | - David J. Beech
- School of Medicine, University of Leeds, Leeds LS2 9JT, U.K
| | - John A. Beutler
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
24
|
ZNF322A-mediated protein phosphorylation induces autophagosome formation through modulation of IRS1-AKT glucose uptake and HSP-elicited UPR in lung cancer. J Biomed Sci 2020; 27:75. [PMID: 32576196 PMCID: PMC7310457 DOI: 10.1186/s12929-020-00668-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 06/09/2020] [Indexed: 12/19/2022] Open
Abstract
Background ZNF322A is an oncogenic transcription factor that belongs to the Cys2His2-type zinc-finger protein family. Accumulating evidence suggests that ZNF322A may contribute to the tumorigenesis of lung cancer, however, the ZNF322A-mediated downstream signaling pathways remain unknown. Methods To uncover ZNF322A-mediated functional network, we applied phosphopeptide enrichment and isobaric labeling strategies with mass spectrometry-based proteomics using A549 lung cancer cells, and analyzed the differentially expressed proteins of phosphoproteomic and proteomic profiles to determine ZNF322A-modulated pathways. Results ZNF322A highlighted a previously unidentified insulin signaling, heat stress, and signal attenuation at the post-translational level. Consistently, protein-phosphoprotein-kinase interaction network analysis revealed phosphorylation of IRS1 and HSP27 were altered upon ZNF322A-silenced lung cancer cells. Thus, we further investigated the molecular regulation of ZNF322A, and found the inhibitory transcriptional regulation of ZNF322A on PIM3, which was able to phosphorylate IRS1 at serine1101 in order to manipulate glucose uptake via the PI3K/AKT/mTOR signaling pathway. Moreover, ZNF322A also affects the unfolded protein response by phosphorylation of HSP27S82 and eIF2aS51, and triggers autophagosome formation in lung cancer cells. Conclusions These findings not only give new information about the molecular regulation of the cellular proteins through ZNF322A at the post-translational level, but also provides a resource for the study of lung cancer therapy.
Collapse
|
25
|
Mou SB, Xiao W, Wang HQ, Wang SJ, Xiang Z. Syntheses of Epoxyguaiane Sesquiterpenes (−)-Englerin A, (−)-Oxyphyllol, (+)-Orientalol E, and (+)-Orientalol F: A Synthetic Biology Approach. Org Lett 2020; 22:1976-1979. [DOI: 10.1021/acs.orglett.0c00325] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Shu-Bin Mou
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Wen Xiao
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Hua-Qi Wang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Su-Jing Wang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Zheng Xiang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| |
Collapse
|
26
|
Alasady MJ, Mendillo ML. The Multifaceted Role of HSF1 in Tumorigenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1243:69-85. [PMID: 32297212 DOI: 10.1007/978-3-030-40204-4_5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Heat Shock Factor 1 (HSF1), the master transcriptional regulator of the heat shock response (HSR), was first cloned more than 30 years ago. Most early research interrogating the role that HSF1 plays in biology focused on its cytoprotective functions, as a factor that promotes the survival of organisms by protecting against the proteotoxicity associated with neurodegeneration and other pathological conditions. However, recent studies have revealed a deleterious role of HSF1, as a factor that is co-opted by cancer cells to promote their own survival to the detriment of the organism. In cancer, HSF1 operates in a multifaceted manner to promote oncogenic transformation, proliferation, metastatic dissemination, and anti-cancer drug resistance. Here we review our current understanding of HSF1 activation and function in malignant progression and discuss the potential for HSF1 inhibition as a novel anticancer strategy. Collectively, this ever-growing body of work points to a prominent role of HSF1 in nearly every aspect of carcinogenesis.
Collapse
Affiliation(s)
- Milad J Alasady
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Marc L Mendillo
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
27
|
Hall G, Wang L, Spurney RF. TRPC Channels in Proteinuric Kidney Diseases. Cells 2019; 9:cells9010044. [PMID: 31877991 PMCID: PMC7016871 DOI: 10.3390/cells9010044] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 12/20/2022] Open
Abstract
Over a decade ago, mutations in the gene encoding TRPC6 (transient receptor potential cation channel, subfamily C, member 6) were linked to development of familial forms of nephrosis. Since this discovery, TRPC6 has been implicated in the pathophysiology of non-genetic forms of kidney disease including focal segmental glomerulosclerosis (FSGS), diabetic nephropathy, immune-mediated kidney diseases, and renal fibrosis. On the basis of these findings, TRPC6 has become an important target for the development of therapeutic agents to treat diverse kidney diseases. Although TRPC6 has been a major focus for drug discovery, more recent studies suggest that other TRPC family members play a role in the pathogenesis of glomerular disease processes and chronic kidney disease (CKD). This review highlights the data implicating TRPC6 and other TRPC family members in both genetic and non-genetic forms of kidney disease, focusing on TRPC3, TRPC5, and TRPC6 in a cell type (glomerular podocytes) that plays a key role in proteinuric kidney diseases.
Collapse
|
28
|
Liu HT, Huang DA, Li MM, Liu HD, Guo K. HSF1: a mediator in metabolic alteration of hepatocellular carcinoma cells in cross-talking with tumor-associated macrophages. Am J Transl Res 2019; 11:5054-5064. [PMID: 31497221 PMCID: PMC6731433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 07/16/2019] [Indexed: 06/10/2023]
Abstract
Recently, heat shock transcription factor 1 (HSF1) is observed to be involved in the process of cellular metabolism in cancer. However, the roles of HSF1 in the metabolic alteration of hepatocellular carcinoma (HCC) in tumor microenvironment remain elusive. Here, HCC cells were co-cultured with tumor-associated macrophages (TAM). The levels of glucose uptake, the lactate release, reactive oxygen species (ROS) and mtDNA content were measured by the associated Kits; all detected protocols were correspondingly according to the manufacturers' instructions. Recombinant lentiviruses with shRNA against HSF1 and MCT4 were transfected into HCC cells or TAMs. Western blot analysis was conducted to detect the relative levels of HSF1, MCT1 and MCT4 proteins. CCK-8 assay was utilized to assess cell proliferation. Based on the co-culture system with HCC cells and TAMs, metabolic alteration of HCC cells after co-culture with TAMs was observed. Furthermore, glucose consumption rate, lactate production rate and intercellular ROS level were decreased, while the copy number of mtDNA was increased in HSF1-knockdown HCC cells. Besides, metabolic crosstalk between HCC cells and TAMs was induced by HSF1 not only in HCC cells but also in TAMs through regulating individually MCT1 and MCT4 expressions. To the best of our knowledge, this is an important study to demonstrate the roles of HSF1 in regulating metabolic alteration of HCC cells induced by TAMs, which implies the potential use of HSF1 as a target modulating malignant behaviors of HCC cells.
Collapse
Affiliation(s)
- Hua Tian Liu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of EducationShanghai, China
- Cancer Research Center, Institutes of Biomedical Sciences, Fudan UniversityShanghai, China
| | - Dan Ai Huang
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical UniversityNanning, Guangxi, China
| | - Miao Miao Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of EducationShanghai, China
| | - He Deng Liu
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical UniversityNanning, Guangxi, China
| | - Kun Guo
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of EducationShanghai, China
| |
Collapse
|
29
|
Guo L, Plietker B. β‐Ketoesters as Mono‐ or Bisnucleophiles: A Concise Enantioselective Total Synthesis of (−)‐Englerin A and B. Angew Chem Int Ed Engl 2019; 58:8346-8350. [DOI: 10.1002/anie.201900401] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/21/2019] [Indexed: 11/12/2022]
Affiliation(s)
- Lei Guo
- Institut für Organische ChemieUniversität Stuttgart Pfaffenwaldring 55 70569 Stuttgart Germany
| | - Bernd Plietker
- Institut für Organische ChemieUniversität Stuttgart Pfaffenwaldring 55 70569 Stuttgart Germany
| |
Collapse
|
30
|
Takahashi T, Ichikawa H, Morimoto Y, Tsuneyama K, Hijikata T. Inhibition of EP2/EP4 prostanoid receptor-mediated signaling suppresses IGF-1-induced proliferation of pancreatic cancer BxPC-3 cells via upregulating γ-glutamyl cyclotransferase expression. Biochem Biophys Res Commun 2019; 516:388-396. [PMID: 31217077 DOI: 10.1016/j.bbrc.2019.06.054] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 06/10/2019] [Indexed: 12/12/2022]
Abstract
Inhibition of prostaglandin E2 signaling via EP2/EP4 prostanoid receptors suppresses Insulin-like growth factor (IGF)-1-induced proliferation of pancreatic cancer BxPC-3 cells. To better understand the mechanism of EP2/EP4 signaling for controlling cell proliferation, we performed metabolome analyses in BxPC-3 cells treated with IGF-1 alone or IGF-1 plus EP2/EP4 inhibitors. These analyses revealed increased g-aminobutyric acid and 5-oxoproline production following the addition of EP2/EP4 inhibitors to IGF-1-treated cells. The expression of a 5-oxoproline-catalyzing enzyme, γ-glutamylcyclotransferase (GGCT), was also upregulated by IGF-1 treatment and further enhanced by the addition of EP2/EP4 inhibitors. Knockdown of GGCT expression resulted in the loss of suppressive effects of EP2/EP4 inhibitors on IGF-1-induced BxPC-3 cell proliferation, whereas GGCT overexpression repressed the basal proliferation of BxPC-3 cells but did not affect the suppressive effects of EP2/EP4 inhibitors. To summarize, we propose a role for EP2/EP4 signaling in regulating IGF-1-induced cell proliferation, in which EP2/EP4 signaling represses IGF-1-induced GGCT expression, which mediates and whose amount controls a branch of IGF-1 signaling to promote cell proliferation via extracellular signal-regulated kinase phosphorylation.
Collapse
Affiliation(s)
- Tetsuyuki Takahashi
- Department of Anatomy and Cell Biology, Faculty of Pharmacy, Research Institute of Pharmaceutical Sciences, Musashino University, Nishi-Tokyo, Tokyo, Japan
| | - Hirona Ichikawa
- Department of Anatomy and Cell Biology, Faculty of Pharmacy, Research Institute of Pharmaceutical Sciences, Musashino University, Nishi-Tokyo, Tokyo, Japan
| | - Yuuki Morimoto
- Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Takao Hijikata
- Department of Anatomy and Cell Biology, Faculty of Pharmacy, Research Institute of Pharmaceutical Sciences, Musashino University, Nishi-Tokyo, Tokyo, Japan.
| |
Collapse
|
31
|
Guo L, Plietker B. β‐Ketoesters as Mono‐ or Bisnucleophiles: A Concise Enantioselective Total Synthesis of (−)‐Englerin A and B. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201900401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Lei Guo
- Institut für Organische ChemieUniversität Stuttgart Pfaffenwaldring 55 70569 Stuttgart Germany
| | - Bernd Plietker
- Institut für Organische ChemieUniversität Stuttgart Pfaffenwaldring 55 70569 Stuttgart Germany
| |
Collapse
|
32
|
Rubaiy HN. Treasure troves of pharmacological tools to study transient receptor potential canonical 1/4/5 channels. Br J Pharmacol 2019; 176:832-846. [PMID: 30656647 PMCID: PMC6433652 DOI: 10.1111/bph.14578] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/25/2018] [Accepted: 12/18/2018] [Indexed: 12/13/2022] Open
Abstract
Canonical or classical transient receptor potential 4 and 5 proteins (TRPC4 and TRPC5) assemble as homomers or heteromerize with TRPC1 protein to form functional nonselective cationic channels with high calcium permeability. These channel complexes, TRPC1/4/5, are widely expressed in nervous and cardiovascular systems, also in other human tissues and cell types. It is debatable that TRPC1 protein is able to form a functional ion channel on its own. A recent explosion of molecular information about TRPC1/4/5 has emerged including knowledge of their distribution, function, and regulation suggesting these three members of the TRPC subfamily of TRP channels play crucial roles in human physiology and pathology. Therefore, these ion channels represent potential drug targets for cancer, epilepsy, anxiety, pain, and cardiac remodelling. In recent years, a number of highly selective small-molecule modulators of TRPC1/4/5 channels have been identified as being potent with improved pharmacological properties. This review will focus on recent remarkable small-molecule agonists: (-)-englerin A and tonantzitlolone and antagonists: Pico145 and HC7090, of TPRC1/4/5 channels. In addition, this work highlights other recently identified modulators of these channels such as the benzothiadiazine derivative, riluzole, ML204, clemizole, and AC1903. Together, these treasure troves of agonists and antagonists of TRPC1/4/5 channels provide valuable hints to comprehend the functional importance of these ion channels in native cells and in vivo animal models. Importantly, human diseases and disorders mediated by these proteins can be studied using these compounds to perhaps initiate drug discovery efforts to develop novel therapeutic agents.
Collapse
Affiliation(s)
- Hussein N. Rubaiy
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical SchoolUniversity of HullHullUK
| |
Collapse
|
33
|
Cheung SY, Henrot M, Al-Saad M, Baumann M, Muller H, Unger A, Rubaiy HN, Mathar I, Dinkel K, Nussbaumer P, Klebl B, Freichel M, Rode B, Trainor S, Clapcote SJ, Christmann M, Waldmann H, Abbas SK, Beech DJ, Vasudev NS. TRPC4/TRPC5 channels mediate adverse reaction to the cancer cell cytotoxic agent (-)-Englerin A. Oncotarget 2018; 9:29634-29643. [PMID: 30038709 PMCID: PMC6049859 DOI: 10.18632/oncotarget.25659] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 05/01/2018] [Indexed: 01/09/2023] Open
Abstract
(-)-Englerin A (EA) is a natural product which has potent cytotoxic effects on renal cell carcinoma cells and other types of cancer cell but not non-cancer cells. Although selectively cytotoxic to cancer cells, adverse reaction in mice and rats has been suggested. EA is a remarkably potent activator of ion channels formed by Transient Receptor Potential Canonical 4 and 5 proteins (TRPC4 and TRPC5) and TRPC4 is essential for EA-mediated cancer cell cytotoxicity. Here we specifically investigated the relevance of TRPC4 and TRPC5 to the adverse reaction. Injection of EA (2 mg.kg-1 i.p.) adversely affected mice for about 1 hour, manifesting as a marked reduction in locomotor activity, after which they fully recovered. TRPC4 and TRPC5 single knockout mice were partially protected and double knockout mice fully protected. TRPC4/TRPC5 double knockout mice were also protected against intravenous injection of EA. Importance of TRPC4/TRPC5 channels was further suggested by pre-administration of Compound 31 (Pico145), a potent and selective small-molecule inhibitor of TRPC4/TRPC5 channels which did not cause adverse reaction itself but prevented adverse reaction to EA. EA was detected in the plasma but not the brain and so peripheral mechanisms were implicated but not identified. The data confirm the existence of adverse reaction to EA in mice and suggest that it depends on a combination of TRPC4 and TRPC5 which therefore overlaps partially with TRPC4-dependent cancer cell cytotoxicity. The underlying nature of the observed adverse reaction to EA, as a consequence of TRPC4/TRPC5 channel activation, remains unclear and warrants further investigation.
Collapse
Affiliation(s)
- Sin Ying Cheung
- School of Medicine, University of Leeds, Leeds, LS2 9JT, England, UK
| | - Matthias Henrot
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Mohammad Al-Saad
- School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | | | - Heiko Muller
- Lead Discovery Center GmbH, D-44227 Dortmund, Germany
| | - Anke Unger
- Lead Discovery Center GmbH, D-44227 Dortmund, Germany
| | - Hussein N Rubaiy
- School of Medicine, University of Leeds, Leeds, LS2 9JT, England, UK
| | - Ilka Mathar
- Institute of Pharmacology, Universität Heidelberg, D-69120 Heidelberg, Germany
| | - Klaus Dinkel
- Lead Discovery Center GmbH, D-44227 Dortmund, Germany
| | | | - Bert Klebl
- Lead Discovery Center GmbH, D-44227 Dortmund, Germany
| | - Marc Freichel
- Institute of Pharmacology, Universität Heidelberg, D-69120 Heidelberg, Germany
| | - Baptiste Rode
- School of Medicine, University of Leeds, Leeds, LS2 9JT, England, UK
| | - Sebastian Trainor
- School of Medicine, University of Leeds, Leeds, LS2 9JT, England, UK
| | - Steven J Clapcote
- School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Mathias Christmann
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Herbert Waldmann
- Max-Planck-Institut für Molekulare Physiologie, D-44227 Dortmund, Germany.,Technische Universität Dortmund, Fakultät für Chemie und Chemische Biologie, D-44227 Dortmund, Germany
| | - Syed Khawar Abbas
- School of Medicine, University of Leeds, Leeds, LS2 9JT, England, UK
| | - David J Beech
- School of Medicine, University of Leeds, Leeds, LS2 9JT, England, UK
| | - Naveen S Vasudev
- School of Medicine, University of Leeds, Leeds, LS2 9JT, England, UK
| |
Collapse
|
34
|
Rubaiy HN, Ludlow MJ, Siems K, Norman K, Foster R, Wolf D, Beutler JA, Beech DJ. Tonantzitlolone is a nanomolar potency activator of transient receptor potential canonical 1/4/5 channels. Br J Pharmacol 2018; 175:3361-3368. [PMID: 29859013 DOI: 10.1111/bph.14379] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 05/24/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND AND PURPOSE The diterpene ester tonantzitlolone (TZL) is a natural product, which displays cytotoxicity towards certain types of cancer cell such as renal cell carcinoma cells. The effect is similar to that of (-)-englerin A, and so, although it is chemically distinct, we investigated whether TZL also targets transient receptor potential canonical (TRPC) channels of the 1, 4 and 5 type (TRPC1/4/5 channels). EXPERIMENTAL APPROACH The effects of TZL on renal cell carcinoma A498 cells natively expressing TRPC1 and TRPC4, modified HEK293 cells overexpressing TRPC4, TRPC5, TRPC4-TRPC1 or TRPC5-TRPC1 concatemer, TRPC3 or TRPM2, or CHO cells overexpressing TRPV4 were studied by determining changes in intracellular Ca2+ , or whole-cell or excised membrane patch-clamp electrophysiology. KEY RESULTS TZL induced an elevation of intracellular Ca2+ in A498 cells, similar to that evoked by englerin A. TZL activated overexpressed channels with EC50 values of 123 nM (TRPC4), 83 nM (TRPC5), 140 nM (TRPC4-TRPC1) and 61 nM (TRPC5-TRPC1). These effects of TZL were reversible on wash-out and potently inhibited by the TRPC1/4/5 inhibitor Pico145. TZL activated TRPC5 channels when bath-applied to excised outside-out but not inside-out patches. TZL failed to activate endogenous store-operated Ca2+ entry or overexpressed TRPC3, TRPV4 or TRPM2 channels in HEK 293 cells. CONCLUSIONS AND IMPLICATIONS TZL is a novel potent agonist for TRPC1/4/5 channels, which should be useful for testing the functionality of this type of ion channel and understanding how TRPC1/4/5 agonists achieve selective cytotoxicity against certain types of cancer cell.
Collapse
Affiliation(s)
| | | | | | | | | | | | - John A Beutler
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | | |
Collapse
|
35
|
Minard A, Bauer CC, Wright DJ, Rubaiy HN, Muraki K, Beech DJ, Bon RS. Remarkable Progress with Small-Molecule Modulation of TRPC1/4/5 Channels: Implications for Understanding the Channels in Health and Disease. Cells 2018; 7:E52. [PMID: 29865154 PMCID: PMC6025525 DOI: 10.3390/cells7060052] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/21/2018] [Accepted: 05/23/2018] [Indexed: 12/12/2022] Open
Abstract
Proteins of the TRPC family can form many homo- and heterotetrameric cation channels permeable to Na⁺, K⁺ and Ca2+. In this review, we focus on channels formed by the isoforms TRPC1, TRPC4 and TRPC5. We review evidence for the formation of different TRPC1/4/5 tetramers, give an overview of recently developed small-molecule TRPC1/4/5 activators and inhibitors, highlight examples of biological roles of TRPC1/4/5 channels in different tissues and pathologies, and discuss how high-quality chemical probes of TRPC1/4/5 modulators can be used to understand the involvement of TRPC1/4/5 channels in physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Aisling Minard
- School of Chemistry, University of Leeds, Leeds LS2 9JT, UK.
| | - Claudia C Bauer
- Department of Discovery and Translational Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK.
| | - David J Wright
- Department of Discovery and Translational Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK.
| | - Hussein N Rubaiy
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, Hull HU6 7RX, UK.
| | - Katsuhiko Muraki
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, 1-100 Kusumoto, Chikusa, Nagoya 464-8650, Japan.
| | - David J Beech
- Department of Discovery and Translational Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK.
| | - Robin S Bon
- Department of Discovery and Translational Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
36
|
Gold(I)-Catalysis for the Synthesis of Terpenoids: From Intramolecular Cascades to Intermolecular Cycloadditions. Isr J Chem 2018. [DOI: 10.1002/ijch.201800006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
37
|
Rubaiy HN, Seitz T, Hahn S, Choidas A, Habenberger P, Klebl B, Dinkel K, Nussbaumer P, Waldmann H, Christmann M, Beech DJ. Identification of an (-)-englerin A analogue, which antagonizes (-)-englerin A at TRPC1/4/5 channels. Br J Pharmacol 2018; 175:830-839. [PMID: 29247460 PMCID: PMC5811624 DOI: 10.1111/bph.14128] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/30/2017] [Accepted: 12/01/2017] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND PURPOSE (-)-Englerin A (EA) is a potent cytotoxic agent against renal carcinoma cells. It achieves its effects by activation of transient receptor potential canonical (TRPC)4/TRPC1 heteromeric channels. It is also an agonist at channels formed by the related protein, TRPC5. Here, we sought an EA analogue, which might enable a better understanding of these effects of EA. EXPERIMENTAL APPROACH An EA analogue, A54, was synthesized by chemical elaboration of EA. The effects of EA and A54 on the activity of human TRPC4 or TRPC5 channels overexpressed on A498 and HEK 293 cells were investigated, firstly, by measuring intracellular Ca2+ and, secondly, current using whole-cell patch clamp recordings. KEY RESULTS A54 had weak or no agonist activity at endogenous TRPC4/TRPC1 channels in A498 cells or TRPC4 or TRPC5 homomeric channels overexpressed in HEK 293 cells. A54 strongly inhibited EA-mediated activation of TRPC4/TRPC1 or TRPC5 and weakly inhibited activation of TRPC4. Studies of TRPC5 showed that A54 shifted the EA concentration-response curve to the right without changing its slope, consistent with competitive antagonism. In contrast, Gd3+ -activated TRPC5 or sphingosine-1-phosphate-activated TRPC4 channels were not inhibited but potentiated by A54. A54 did not activate TRPC3 channels or affect the activation of these channels by the agonist 1-oleoyl-2-acetyl-sn-glycerol. CONCLUSIONS AND IMPLICATIONS This study has revealed a new tool compound for EA and TRPC1/4/5 channel research, which could be useful for characterizing endogenous TRPC1/4/5 channels and understanding EA-binding sites and their physiological relevance.
Collapse
Affiliation(s)
| | - Tobias Seitz
- Institute of Chemistry and BiochemistryFreie Universität BerlinBerlinGermany
| | - Sven Hahn
- Institute of Chemistry and BiochemistryFreie Universität BerlinBerlinGermany
| | | | | | - Bert Klebl
- Lead Discovery Center GmbHDortmundGermany
| | | | | | | | - Mathias Christmann
- Institute of Chemistry and BiochemistryFreie Universität BerlinBerlinGermany
| | | |
Collapse
|
38
|
The Role of Compounds Derived from Natural Supplement as Anticancer Agents in Renal Cell Carcinoma: A Review. Int J Mol Sci 2017; 19:ijms19010107. [PMID: 29301217 PMCID: PMC5796057 DOI: 10.3390/ijms19010107] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/24/2017] [Accepted: 12/28/2017] [Indexed: 12/24/2022] Open
Abstract
Renal Cell Carcinoma (RCC) is the most prominent kidney cancer derived from renal tubules and accounts for roughly 85% of all malignant kidney cancer. Every year, over 60,000 new cases are registered, and about 14,000 people die from RCC. The incidence of this has been increasing significantly in the U.S. and other countries. An increased understanding of molecular biology and the genomics of RCC has uncovered several signaling pathways involved in the progression of this cancer. Significant advances in the treatment of RCC have been reported from agents approved by the Food and Drug Administration (FDA) that target these pathways. These agents have become drugs of choice because they demonstrate clinical benefit and increased survival in patients with metastatic disease. However, the patients eventually relapse and develop resistance to these drugs. To improve outcomes and seek approaches for producing long-term durable remission, the search for more effective therapies and preventative strategies are warranted. Treatment of RCC using natural products is one of these strategies to reduce the incidence. However, recent studies have focused on these chemoprevention agents as anti-cancer therapies given they can inhibit tumor cell grow and lack the severe side effects common to synthetic compounds. This review elaborates on the current understanding of natural products and their mechanisms of action as anti-cancer agents. The present review will provide information for possible use of these products alone or in combination with chemotherapy for the prevention and treatment of RCC.
Collapse
|
39
|
Na + entry through heteromeric TRPC4/C1 channels mediates (-)Englerin A-induced cytotoxicity in synovial sarcoma cells. Sci Rep 2017; 7:16988. [PMID: 29209034 PMCID: PMC5717101 DOI: 10.1038/s41598-017-17303-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/22/2017] [Indexed: 12/22/2022] Open
Abstract
The sesquiterpene (-)Englerin A (EA) is an organic compound from the plant Phyllanthus engleri which acts via heteromeric TRPC4/C1 channels to cause cytotoxicity in some types of cancer cell but not normal cells. Here we identified selective cytotoxicity of EA in human synovial sarcoma cells (SW982 cells) and investigated the mechanism. EA induced cation channel current (Icat) in SW982 cells with biophysical characteristics of heteromeric TRPC4/C1 channels. Inhibitors of homomeric TRPC4 channels were weak inhibitors of the Icat and EA-induced cytotoxicity whereas a potent inhibitor of TRPC4/C1 channels (Pico145) strongly inhibited Icat and cytotoxicity. Depletion of TRPC1 converted Icat into a current with biophysical and pharmacological properties of homomeric TRPC4 channels and depletion of TRPC1 or TRPC4 suppressed the cytotoxicity of EA. A Na+/K+-ATPase inhibitor (ouabain) potentiated EA-induced cytotoxicity and direct Na+ loading by gramicidin-A caused Pico145-resistant cytotoxicity in the absence of EA. We conclude that EA has a potent cytotoxic effect on human synovial sarcoma cells which is mediated by heteromeric TRPC4/C1 channels and Na+ loading.
Collapse
|
40
|
Wang X, Dande RR, Yu H, Samelko B, Miller RE, Altintas MM, Reiser J. TRPC5 Does Not Cause or Aggravate Glomerular Disease. J Am Soc Nephrol 2017; 29:409-415. [PMID: 29061651 DOI: 10.1681/asn.2017060682] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022] Open
Abstract
Transient receptor potential channel 5 (TRPC5) is highly expressed in brain and kidney and mediates calcium influx and promotes cell migration. In the kidney, loss of TRPC5 function has been reported to benefit kidney filter dynamics by balancing podocyte cytoskeletal remodeling. However, in vivo gain-in-function studies of TRPC5 with respect to kidney function have not been reported. To address this gap, we developed two transgenic mouse models on the C57BL/6 background by overexpressing either wild-type TRPC5 or a TRPC5 ion-pore mutant. Compared with nontransgenic controls, neither transgenic model exhibited an increase in proteinuria at 8 months of age or a difference in LPS-induced albuminuria. Moreover, activation of TRPC5 by Englerin A did not stimulate proteinuria, and inhibition of TRPC5 by ML204 did not significantly lower the level of LPS-induced proteinuria in any group. Collectively, these data suggest that the overexpression or activation of the TRPC5 ion channel does not cause kidney barrier injury or aggravate such injury under pathologic conditions.
Collapse
Affiliation(s)
- Xuexiang Wang
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Ranadheer R Dande
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Hao Yu
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Beata Samelko
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Rachel E Miller
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Mehmet M Altintas
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
41
|
Gomez-Pastor R, Burchfiel ET, Thiele DJ. Regulation of heat shock transcription factors and their roles in physiology and disease. Nat Rev Mol Cell Biol 2017; 19:4-19. [PMID: 28852220 DOI: 10.1038/nrm.2017.73] [Citation(s) in RCA: 459] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The heat shock transcription factors (HSFs) were discovered over 30 years ago as direct transcriptional activators of genes regulated by thermal stress, encoding heat shock proteins. The accepted paradigm posited that HSFs exclusively activate the expression of protein chaperones in response to conditions that cause protein misfolding by recognizing a simple promoter binding site referred to as a heat shock element. However, we now realize that the mammalian family of HSFs comprises proteins that independently or in concert drive combinatorial gene regulation events that activate or repress transcription in different contexts. Advances in our understanding of HSF structure, post-translational modifications and the breadth of HSF-regulated target genes have revealed exciting new mechanisms that modulate HSFs and shed new light on their roles in physiology and pathology. For example, the ability of HSF1 to protect cells from proteotoxicity and cell death is impaired in neurodegenerative diseases but can be exploited by cancer cells to support their growth, survival and metastasis. These new insights into HSF structure, function and regulation should facilitate the development tof new disease therapeutics to manipulate this transcription factor family.
Collapse
Affiliation(s)
- Rocio Gomez-Pastor
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine
| | | | - Dennis J Thiele
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine.,Department of Biochemistry, Duke University School of Medicine.,Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| |
Collapse
|
42
|
Elliott DC, Beutler JA, Parker KA. Importance of a 4-Alkyl Substituent for Activity in the Englerin Series. ACS Med Chem Lett 2017; 8:746-750. [PMID: 28740610 DOI: 10.1021/acsmedchemlett.7b00161] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 06/05/2017] [Indexed: 11/30/2022] Open
Abstract
The ring closing metathesis/transannular etherification approach to the englerin nucleus was adapted to provide two key intermediates for analogue synthesis: the 4-desmethyl Δ5,6 tricycle and the 4-oxo Δ5,6 tricycle. The former was elaborated to 4-desmethyl englerin A and the latter served as a common precursor for englerin A, 4-ethyl englerin A, and 4-isopropyl englerin A. 4-Desmethyl englerin A was less active than the natural product by an order of magnitude, but the 4-ethyl and 4-isopropyl analogues were comparable in activity to englerin A. These results are consistent with the premise that the 4-alkyl group enforces the binding conformation of the cinnamoyl ester substituent. Furthermore, they suggest that 4-alkyl englerin structures may prove to be useful tool compounds.
Collapse
Affiliation(s)
- Daniel C. Elliott
- Department
of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - John A. Beutler
- Molecular
Targets Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Kathlyn A. Parker
- Department
of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
| |
Collapse
|
43
|
Wu Z, Zhao S, Fash DM, Li Z, Chain WJ, Beutler JA. Englerins: A Comprehensive Review. JOURNAL OF NATURAL PRODUCTS 2017; 80:771-781. [PMID: 28170253 PMCID: PMC6198806 DOI: 10.1021/acs.jnatprod.6b01167] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
In the decade since the discovery of englerin A (1) and its potent activity in cancer models, this natural product and its analogues have been the subject of numerous chemical, biological, and preclinical studies by many research groups. This review summarizes published findings and proposes further research directions required for entry of an englerin analogue into clinical trials for kidney cancer and other conditions.
Collapse
Affiliation(s)
- Zhenhua Wu
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - Senzhi Zhao
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - David M. Fash
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - Zhenwu Li
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - William J. Chain
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - John A. Beutler
- Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
44
|
Batova A, Altomare D, Creek KE, Naviaux RK, Wang L, Li K, Green E, Williams R, Naviaux JC, Diccianni M, Yu AL. Englerin A induces an acute inflammatory response and reveals lipid metabolism and ER stress as targetable vulnerabilities in renal cell carcinoma. PLoS One 2017; 12:e0172632. [PMID: 28296891 PMCID: PMC5351975 DOI: 10.1371/journal.pone.0172632] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 02/06/2017] [Indexed: 12/14/2022] Open
Abstract
Renal cell carcinoma (RCC) is among the top ten most common forms of cancer and is the most common malignancy of the kidney. Clear cell renal carcinoma (cc-RCC), the most common type of RCC, is one of the most refractory cancers with an incidence that is on the rise. Screening of plant extracts in search of new anti-cancer agents resulted in the discovery of englerin A, a guaiane sesquiterpene with potent cytotoxicity against renal cancer cells and a small subset of other cancer cells. Though a few cellular targets have been identified for englerin A, it is still not clear what mechanisms account for the cytotoxicity of englerin A in RCC, which occurs at concentrations well below those used to engage the targets previously identified. Unlike any prior study, the current study used a systems biology approach to explore the mechanism(s) of action of englerin A. Metabolomics analyses indicated that englerin A profoundly altered lipid metabolism by 24 h in cc-RCC cell lines and generated significant levels of ceramides that were highly toxic to these cells. Microarray analyses determined that englerin A induced ER stress signaling and an acute inflammatory response, which was confirmed by quantitative PCR and Western Blot analyses. Additionally, fluorescence confocal microscopy revealed that englerin A at 25 nM disrupted the morphology of the ER confirming the deleterious effect of englerin A on the ER. Collectively, our findings suggest that cc-RCC is highly sensitive to disruptions in lipid metabolism and ER stress and that these vulnerabilities can be targeted for the treatment of cc-RCC and possibly other lipid storing cancers. Furthermore, our results suggest that ceramides may be a mediator of some of the actions of englerin A. Lastly, the acute inflammatory response induced by englerin A may mediate anti-tumor immunity.
Collapse
Affiliation(s)
- Ayse Batova
- Department of Pediatrics, University of California, San Diego, California, United States of America
- * E-mail:
| | - Diego Altomare
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina, United States of America
| | - Kim E. Creek
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina, United States of America
| | - Robert K. Naviaux
- Department of Pediatrics, University of California, San Diego, California, United States of America
- The Mitochondrial and Metabolic Disease Center, Department of Pathology, University of California, San Diego, San Diego, California, United States of America
- Department of Medicine, University of California, San Diego, California, United States of America
| | - Lin Wang
- The Mitochondrial and Metabolic Disease Center, Department of Pathology, University of California, San Diego, San Diego, California, United States of America
| | - Kefeng Li
- The Mitochondrial and Metabolic Disease Center, Department of Pathology, University of California, San Diego, San Diego, California, United States of America
| | - Erica Green
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina, United States of America
| | - Richard Williams
- Department of Pediatrics, University of California, San Diego, California, United States of America
| | - Jane C. Naviaux
- The Mitochondrial and Metabolic Disease Center, Department of Pathology, University of California, San Diego, San Diego, California, United States of America
| | - Mitchell Diccianni
- Department of Pediatrics, University of California, San Diego, California, United States of America
| | - Alice L. Yu
- Department of Pediatrics, University of California, San Diego, California, United States of America
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| |
Collapse
|
45
|
Ludlow MJ, Gaunt HJ, Rubaiy HN, Musialowski KE, Blythe NM, Vasudev NS, Muraki K, Beech DJ. (-)-Englerin A-evoked Cytotoxicity Is Mediated by Na+ Influx and Counteracted by Na+/K+-ATPase. J Biol Chem 2016; 292:723-731. [PMID: 27875305 PMCID: PMC5241745 DOI: 10.1074/jbc.m116.755678] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/09/2016] [Indexed: 11/21/2022] Open
Abstract
(−)-Englerin A ((−)-EA) has a rapid and potent cytotoxic effect on several types of cancer cell that is mediated by plasma membrane ion channels containing transient receptor potential canonical 4 (TRPC4) protein. Because these channels are Ca2+-permeable, it was initially thought that the cytotoxicity arose as a consequence of Ca2+ overload. Here we show that this is not the case and that the effect of (−)-EA is mediated by a heteromer of TRPC4 and TRPC1 proteins. Both TRPC4 and TRPC1 were required for (−)-EA cytotoxicity; however, although TRPC4 was necessary for the (−)-EA-evoked Ca2+ elevation, TRPC1 was not. TRPC1 either had no role or was a negative regulator of Ca2+ entry. By contrast, both TRPC4 and TRPC1 were necessary for monovalent cation entry evoked by (−)-EA, and (−)-EA-evoked cell death was dependent upon entry of the monovalent cation Na+. We therefore hypothesized that Na+/K+-ATPase might act protectively by counteracting the Na+ load resulting from sustained Na+ entry. Indeed, inhibition of Na+/K+-ATPase by ouabain potently and strongly increased (−)-EA-evoked cytotoxicity. The data suggest that (−)-EA achieves cancer cell cytotoxicity by inducing sustained Na+ entry through heteromeric TRPC1/TRPC4 channels and that the cytotoxic effect of (−)-EA can be potentiated by Na+/K+-ATPase inhibition.
Collapse
Affiliation(s)
- Melanie J Ludlow
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom and
| | - Hannah J Gaunt
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom and
| | - Hussein N Rubaiy
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom and
| | - Katie E Musialowski
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom and
| | - Nicola M Blythe
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom and
| | - Naveen S Vasudev
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom and
| | - Katsuhiko Muraki
- the School of Pharmacy, Aichi-Gakuin University, 1-100 Kusumoto, Chikusa, Nagoya 464-8650, Japan
| | - David J Beech
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom and
| |
Collapse
|
46
|
Nelson R, Gulías M, Mascareñas JL, López F. Concise, Enantioselective, and Versatile Synthesis of (-)-Englerin A Based on a Platinum-Catalyzed [4C+3C] Cycloaddition of Allenedienes. Angew Chem Int Ed Engl 2016; 55:14359-14363. [PMID: 27735111 DOI: 10.1002/anie.201607348] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Indexed: 01/01/2023]
Abstract
A practical synthesis of (-)-englerin A was accomplished in 17 steps and 11 % global yield from commercially available achiral precursors. The key step consists of a platinum-catalyzed [4C+3C] allenediene cycloaddition that directly delivers the trans-fused guaiane skeleton with complete diastereoselectivity. The high enantioselectivity (99 % ee) stems from an asymmetric ruthenium-catalyzed transfer hydrogenation of a readily assembled diene-ynone. The synthesis also features a highly stereoselective oxygenation, and a late-stage cuprate alkylation that enables the preparation of previously inaccessible structural analogues.
Collapse
Affiliation(s)
- Ronald Nelson
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Moisés Gulías
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - José L Mascareñas
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Fernando López
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain. .,Instituto de Química Orgánica General (CSIC), Juan de la Cierva, 3, 28006, Madrid, Spain.
| |
Collapse
|
47
|
Nelson R, Gulías M, Mascareñas JL, López F. Concise, Enantioselective, and Versatile Synthesis of (−)-Englerin A Based on a Platinum-Catalyzed [4C+3C] Cycloaddition of Allenedienes. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201607348] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ronald Nelson
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica; Universidade de Santiago de Compostela; 15782 Santiago de Compostela Spain
| | - Moisés Gulías
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica; Universidade de Santiago de Compostela; 15782 Santiago de Compostela Spain
| | - José L. Mascareñas
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica; Universidade de Santiago de Compostela; 15782 Santiago de Compostela Spain
| | - Fernando López
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica; Universidade de Santiago de Compostela; 15782 Santiago de Compostela Spain
- Instituto de Química Orgánica General (CSIC); Juan de la Cierva, 3 28006 Madrid Spain
| |
Collapse
|
48
|
Sourbier C, Scroggins BT, Mannes PZ, Liao PJ, Siems K, Wolf D, Beutler JA, Linehan WM, Neckers L. Tonantzitlolone cytotoxicity toward renal cancer cells is PKCθ- and HSF1-dependent. Oncotarget 2016; 6:29963-74. [PMID: 26298773 PMCID: PMC4745775 DOI: 10.18632/oncotarget.4676] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 07/10/2015] [Indexed: 11/25/2022] Open
Abstract
Elucidating the targets and mechanism of action of natural products is strategically important prior to drug development and assessment of potential clinical applications. In this report, we elucidated the main targets and mechanism of action of the natural product tonantzitlolone (TZL) in clear cell renal cell carcinoma (CCRCC). We identified TZL as a dual PKCα and PKCθ activator in vitro, although in CCRCC cells its activity was mostly PKCθ-dependent. Through activation of PKCθ, TZL induced an insulin resistant phenotype by inhibiting IRS1 and the PI3K/Akt pathway. Simultaneously, TZL activated the heat shock factor 1 (HSF1) transcription factor driving glucose dependency. Thus, similar to the selective PKCθ activator englerin A, TZL induces a metabolic catastrophe in CCRCC, starving cells of glucose while simultaneously increasing their glycolytic dependency.
Collapse
Affiliation(s)
- Carole Sourbier
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Bradley T Scroggins
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Philip Z Mannes
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Pei-Jyun Liao
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | | | - Dietmar Wolf
- AnalytiCon Discovery GmbH, D-14473 Potsdam, Germany
| | - John A Beutler
- Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Leonard Neckers
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
49
|
Gaunt HJ, Vasudev NS, Beech DJ. Transient receptor potential canonical 4 and 5 proteins as targets in cancer therapeutics. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2016; 45:611-620. [PMID: 27289383 PMCID: PMC5045487 DOI: 10.1007/s00249-016-1142-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 05/12/2016] [Accepted: 05/16/2016] [Indexed: 12/05/2022]
Abstract
Novel approaches towards cancer therapy are urgently needed. One approach might be to target ion channels mediating Ca2+ entry because of the critical roles played by Ca2+ in many cell types, including cancer cells. There are several types of these ion channels, but here we address those formed by assembly of transient receptor potential canonical (TRPC) proteins, particularly those which involve two closely related members of the family: TRPC4 and TRPC5. We focus on these proteins because recent studies point to roles in important aspects of cancer: drug resistance, transmission of drug resistance through extracellular vesicles, tumour vascularisation, and evoked cancer cell death by the TRPC4/5 channel activator (−)-englerin A. We conclude that further research is both justified and necessary before these proteins can be considered as strong targets for anti-cancer cell drug discovery programmes. It is nevertheless already apparent that inhibitors of the channels would be unlikely to cause significant adverse effects, but, rather, have other effects which may be beneficial in the context of cancer and chemotherapy, potentially including suppression of innate fear, visceral pain and pathological cardiac remodelling.
Collapse
Affiliation(s)
- Hannah J Gaunt
- School of Medicine, University of Leeds, LIGHT Building, Clarendon Way, Leeds, LS2 9JT, UK.
| | - Naveen S Vasudev
- School of Medicine, University of Leeds, LIGHT Building, Clarendon Way, Leeds, LS2 9JT, UK
| | - David J Beech
- School of Medicine, University of Leeds, LIGHT Building, Clarendon Way, Leeds, LS2 9JT, UK.
| |
Collapse
|
50
|
Fash DM, Peer CJ, Li Z, Talisman IJ, Hayavi S, Sulzmaier FJ, Ramos JW, Sourbier C, Neckers L, Figg WD, Beutler JA, Chain WJ. Synthesis of a stable and orally bioavailable englerin analogue. Bioorg Med Chem Lett 2016; 26:2641-4. [PMID: 27107948 PMCID: PMC4862412 DOI: 10.1016/j.bmcl.2016.04.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 04/06/2016] [Accepted: 04/07/2016] [Indexed: 12/26/2022]
Abstract
Synthesis of analogues of englerin A with a reduced propensity for hydrolysis of the glycolate moiety led to a compound which possessed the renal cancer cell selectivity of the parent and was orally bioavailable in mice.
Collapse
Affiliation(s)
- David M Fash
- Department of Chemistry, University of Hawaii at Manoa, 2545 McCarthy Mall, Honolulu, HI 96822, United States
| | - Cody J Peer
- Genitourinary Malignancies Branch, National Cancer Institute, Frederick, MD 21702, United States
| | - Zhenwu Li
- Department of Chemistry, University of Hawaii at Manoa, 2545 McCarthy Mall, Honolulu, HI 96822, United States
| | - Ian J Talisman
- Department of Chemistry, University of Hawaii at Manoa, 2545 McCarthy Mall, Honolulu, HI 96822, United States
| | - Sima Hayavi
- Developmental Therapeutics Program, National Cancer Institute, Frederick, MD 21702, United States
| | - Florian J Sulzmaier
- The University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI 96813, United States
| | - Joe W Ramos
- The University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI 96813, United States
| | - Carole Sourbier
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States
| | - Leonard Neckers
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States
| | - W Douglas Figg
- Genitourinary Malignancies Branch, National Cancer Institute, Frederick, MD 21702, United States
| | - John A Beutler
- Molecular Targets Laboratory, National Cancer Institute, Frederick, MD 21702, United States.
| | - William J Chain
- Department of Chemistry, University of Hawaii at Manoa, 2545 McCarthy Mall, Honolulu, HI 96822, United States; The University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI 96813, United States.
| |
Collapse
|